Publication Cover
Redox Report
Communications in Free Radical Research
Volume 29, 2024 - Issue 1
2,037
Views
0
CrossRef citations to date
0
Altmetric
Review Article

Integrated approach to reducing polypharmacy in older people: exploring the role of oxidative stress and antioxidant potential therapy

ORCID Icon, , ORCID Icon, , & ORCID Icon

ABSTRACT

Increased life expectancy, attributed to improved access to healthcare and drug development, has led to an increase in multimorbidity, a key contributor to polypharmacy. Polypharmacy is characterised by its association with a variety of adverse events in the older persons. The mechanisms involved in the development of age-related chronic diseases are largely unknown; however, altered redox homeostasis due to ageing is one of the main theories. In this context, the present review explores the development and interaction between different age-related diseases, mainly linked by oxidative stress. In addition, drug interactions in the treatment of various diseases are described, emphasising that the holistic management of older people and their pathologies should prevail over the individual treatment of each condition.

1. Introduction

The world is experiencing a demographic shift in which the population aged 65 and over is increasing rapidly compared to other age groups. This group of people worldwide is expected to grow from 761 million in 2021 to 1.6 billion in 2050, according to United Nations estimates [Citation1]. This demographic transition is the result of declining fertility rates coupled with an increase in life expectancy, as older adults are now living longer due to technological advances. However, living longer does not necessarily involve a better life quality. In fact, as the number of older people increases, so does the prevalence of chronic non-communicable diseases and disability. In this context, multimorbidity, understood as the development of 2 or more chronic pathologies in an individual, is currently considered a public health problem that affects, according to some estimates, more than half of the population over 60 years of age [Citation2]. The main diseases being experienced by older adults are cardiovascular diseases (CVD) first, followed by type 2 diabetes mellitus (DM2), musculoskeletal disorders, respiratory diseases, cancer and mental health disorders [Citation3]. This increase in age-related chronic diseases has led to more prescription of medicines for the management of these diseases, with the main consequence being an increase in the prevalence of polypharmacy.

1.1. Polypharmacy

Polypharmacy corresponds to a geriatric syndrome whose definition is often very heterogeneous; however, according to a systematic review, the most widely used definition is usually the one that considers the consumption of 5 or more drugs. Whereas hyperpolypharmacy or also called excessive polypharmacy is the concurrent use of 10 or more drugs [Citation4]. This heterogeneity in definition between countries makes it difficult to calculate prevalence; however, some studies worldwide estimated overall 37% of the population over 65 years of age consume 5 or more drugs [Citation5] This geriatric syndrome is associated with an increase in various adverse health outcomes in older people, such as falls, cognitive impairment, disability, hospitalizations, adverse drug reactions, increased hospital stay, among others. These adverse outcomes are increased in frail older people, where frailty is understood as a clinically recognizable condition of increased vulnerability as a result of age-related decline in the reserve and function of various physiological systems, leading to reduced resistance to endogenous and exogenous stressors [Citation6,Citation7].

Thus, polypharmacy was born as a form of management implemented by health systems to address the multimorbidity of the elderly, where factors such as limited consultation time, inadequate coordination of services, lack of evidence, limited availability of geriatricians, among other factors, end up promoting the treatment of different conditions in an individualized manner without taking into consideration the integrity of the geriatric patient, where interactions between diseases and medications are common [Citation8]. This favors phenomena such as cascade prescribing, which is one of the main forms of inappropriate prescribing [Citation9]. In this context, the development of geroscience has enabled research into possible interventions that address the physiology of ageing in an attempt to prevent and reduce the severity of age-related chronic diseases [Citation10]. It is in this sense that this manuscript aims to address therapies aimed at modulating oxidative stress, given that by targeting one of the main substrates of ageing, in the long term they would be expected to eliminate multimorbidity, the indication of therapeutic drugs and thus polypharmacy.

2. Geriatric pharmacology

Ageing has been defined as a normal physiological process characterized by the degeneration over time of cellular and molecular structures that lead to a deterioration of function and organic reserve, limiting the capacity to respond to stressors. All these changes eventually lead to the development of diseases and the death of organisms [Citation4]. The changes attributed to ageing include alterations in response to medication due to pharmacokinetic and pharmacodynamic changes. Pharmacokinetics can be understood in simple terms as how the organism affects the drug, while pharmacodynamics corresponds to how the drug affects the organism [Citation11]. The main changes in geriatric pharmacology are largely due to reduced target organ function, changes in receptor sensitivity, altered homeostasis, concurrent drug use, complexity of concomitant disease states, among others. This section briefly describes the main pharmacokinetic changes produced by age.

2.1. Absorption

Ageing generates substantial changes in the gastrointestinal tract that have a negative impact on the uptake and absorption of orally administered medicines. The main changes observed are at the level of the stomach and distal gastrointestinal tract. At the level of the stomach, a decrease in acid secretion is described as a result of the increased prevalence of atrophic gastritis, which is enhanced by the chronic use of proton pump inhibitors in the older adult population [Citation12,Citation13]. All this generates an increase in pH, altering the solubilization of drugs that behave as weak acids or bases. Thus, an increase in pH would be expected to decrease in vivo dissolution and absorption of weakly basic drugs, but increase in vivo dissolution and absorption of weakly acidic drugs [Citation14,Citation15]. Gastrointestinal motility is also affected by the decrease in cholinergic neurons in the myenteric plexus due to age and concomitant diseases, which is reflected in a decrease in peristalsis and gastric emptying, leading to delayed absorption of certain drugs and increased exposure to the gastrointestinal tract depending on where they are absorbed [Citation16–18].

Not only gastrointestinal absorption is affected by age but also other routes of absorption such as transdermal and inhalation. For example, it has been observed that older adults are more sensitive to transdermal fentanyl patches than younger subjects [Citation19], which could be due to a decrease in the thickness of the epidermis and a weakening of the dermis [Citation20]. In addition, the inhalation pathway also suffers from alterations due to age-related changes in respiratory physiology. This is why elderly patients, as a result of the degradation of elastic fibers, experience a decrease in the total alveolar surface area due to an increase in air spaces, which can be enhanced by smoking, in addition to a loss in the elasticity of the rib cage due to ossification of the rib cartilages, weakening of the respiratory muscles and loss of height of the intervertebral discs [Citation21,Citation22]. These changes lead to an impairment in the respiratory mechanics of the older adult that may influence the absorption of inhaled drugs [Citation23].

2.2. Distribution

Another pharmacokinetic parameter that is altered during ageing is the distribution of drugs in the body. Body water decreases by 10-15% in older adults compared to younger people. This decrease is presumed to be due to a loss of intracellular water, as the extracellular volume remains relatively constant. Parallel to the decrease in body water, there is a decrease in muscle mass and an increase in body fat, particularly visceral fat tissue [Citation24–28]. These changes lead to alterations in the volume of distribution of water- and fat-soluble drugs. In this way, a decrease in the volume of distribution of water-soluble drugs (e.g. digoxin, ethanol, aminoglycosides, theophylline, etc.) is generated, reaching increased plasma concentrations of the latter in a shorter time compared to younger subjects [Citation29,Citation30]. In turn, fat-soluble drugs (e.g. benzodiazepines) will suffer an increased volume of distribution and an increased half-life due to accumulation in adipose tissue [Citation31].

In addition to changes in body composition, there are changes linked to plasma protein binding. In this sense, the decrease in serum albumin concentration and the modification of drug binding sites due to ageing are described. However, age alone is not a determinant of serum albumin alterations, but rather due to age-related pathologies such as malnutrition, sarcopenia and inflammatory diseases [Citation32–34]. These changes in albumin can lead to increased free plasma concentrations of acidic drugs that have a high affinity for albumin, such as diazepam, warfarin, salicylic acid, phenytoin, among others. In parallel to changes in albumin, changes in the concentrations of acid glycoprotein 1 alpha have been described as a result of the inflammatory state of the pathologies that accompany ageing [Citation35,Citation36]. These changes in protein concentrations will not have a major effect on most drugs, since changes in the free fraction will be compensated in parallel by changes in drug excretion, but they will be relevant to the pharmacodynamic action of drugs toward a narrow therapeutic index [Citation37].

2.3. Metabolism and excretion

Most tissues have some capacity to metabolize drugs, however, the main organ responsible for drug metabolism is the liver. It is precisely this process that is affected by ageing due to different factors, such as the decrease in hepatic blood flow, describing a decrease of approximately 20-50% of blood flow in older adults [Citation38], which could alter the transport of drugs to the liver and the supply of oxygen to hepatocytes, which is necessary for phase I oxidation reactions. This is compounded by a decrease in liver volume and blood flow, affecting the cytochrome P450 activity and changing the drug metabolism, increasing susceptibility to drug-induced liver injury [Citation39]. Lipophilic drugs may exhibit an expanded distribution volume, resulting in an extended half-life, while drugs that readily dissolve in water often show a more limited distribution volume. Among the elderly population, certain medications can experience a reduction of up to 30% in hepatic drug clearance, with a higher likelihood of impaired CYP-mediated phase I reactions compared to phase II metabolism. Related to this, CYP3A activity, the most important cytochrome P450 isoforms responsible for drug metabolism by humans, is reduced in ageing [Citation40,Citation41]. Another phenomenon that alters hepatic metabolism in older adults is polypharmacy due to the action of different drugs on liver enzyme levels and activity, making it difficult to study their pharmacokinetics and the role of enzymes in decreasing hepatic clearance.

Renal function is also altered in older adults; in fact, the kidneys are among the organs that undergo the most marked changes during the normal ageing process [Citation42]. These changes are characterized by a progressive reduction in parenchymal thickness and total renal volume. Some studies have described a decrease in volume of around 16 and 22 cm3 per decade from the age of 50 [Citation43–45]. These microstructural alterations are mainly associated with the reduction of functioning nephrons due to nephrosclerosis [Citation46,Citation47], which is reflected in the glomerular filtration rate (GFR). Thus, some studies postulate that the average annual decline in GFR ranges from 0.4 to 2.6 ml/min/1.73 m2 and that after the age of 35 years GFR falls by approximately 5-10% per decade [Citation48–50]. It should be mentioned that the progression of these changes will be enhanced in the presence of hypertension. These morphological and functional renal impairments are accompanied by a decrease in renal blood flow and alterations in the renal flow autoregulatory mechanism, characterized by increased vasoconstrictor responses and decreased vasodilator responses [Citation51]. These age-related changes in renal function have a number of clinical implications, including increased half-life of renally excreted drugs, and increased susceptibility to acute kidney injury (AKI) due to the nephrotoxicity of some drugs [Citation52].

3. Changes in redox homeostasis in the older life stages

The mechanisms involved in ageing are not fully understood. However, attempts have been made to characterize the classical ageing phenotype, given nine features that accompany the normal ageing process. These include: genomic instability, telomere attrition, epigenetic alterations, prosthesis loss, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell depletion, altered intercellular communication, disabled macroautophagy, chronic inflammation and dysbiosis [Citation53]. In this context, different theories have emerged as an attempt to link these characteristics so that they do not remain isolated events that occur during ageing. One of these theories is the one that proposes that the accumulation of oxidative stress is associated with the development of chronic inflammation, which would generate a deterioration of all the cells of an individual, having a greater impact on the homeostatic systems, i.e. nervous, endocrine and immune systems, which would explain the increased morbidity and mortality of population during ageing [Citation54].

Oxidative and nitrosative stress represent mechanisms of damage in various pathologies. One of the latest definitions given by the scientific community characterizes it as an alteration of redox signaling and control, leading to disturbances in redox homeostasis and damage to biomolecules [Citation55]. The main free radicals generated are reactive oxygen species (ROS), which, in the presence of nitric oxide (NO), can be converted into reactive oxygen and nitrogen species, being much more reactive than when on their own. These molecules, at certain concentrations, support fundamental life processes, serving as a defense mechanism against infectious agents and in the function of various cell signaling systems. However, when they increase above the physiological range and overwhelm adaptive systems, this leads to altered redox signaling and cellular damage [Citation56,Citation57].

The generation of free radicals in the cell occurs both by enzymatic action and in free form. The main sources of free radicals are oxidative phosphorylation in the inner membrane of mitochondria and NADPH oxidase (NOX) in activated leukocytes during the respiratory burst process [Citation58]. In addition, reactions catalyzed by the enzymes myeloperoxidase and nitric oxide synthase (NOS) are added when it is uncoupled due to the loss of the cofactor tetrahydrobiopterin (BH4) and acquires the ability to synthesize superoxide anion instead of nitric oxide [Citation59]. However, the enzymatic pathway is not the only route by which free radicals are generated; they can also be generated spontaneously, as in the Fenton and Haber–Weiss reactions in the presence of free ionic iron [Citation60].

Antioxidant defenses can be divided into enzymatic and non-enzymatic antioxidants. Antioxidant enzymes include superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPX), among others () [Citation61]. The antioxidant response is mainly regulated by the Nrf2/Keap1 system. Nuclear factor erythroid 2 (Nrf2) is a transcription factor that, upon dimerization, binds to promoter regions called antioxidant response elements (ARE), regulating the expression of many cytoprotective enzyme genes, most notably antioxidant enzymes. Under normal conditions, the adaptor protein Keap1 negatively regulates Nrf2; however, upon exposure to oxidative stress, Keap1 loses its ability to ubiquitinate Nrf2, allowing the activation of its target genes () [Citation62].

Figure 1. Molecular mechanism of oxidative stress and potential targets for antioxidant treatment in aging-related diseases. ARE, antioxidant response elements; CAT, catalase, GLP-1, glucagon-like peptide 1;GPX, glutathione peroxidase; GST, glutathione transferase; HO-1, heme oxygenase 1; Keap1, Kelch-like ECH-associated protein 1; NF-κB, nuclear factor kappa-light-chain-enhancer of the activated B cells; Nrf2, nuclear factor-erythroid 2-related factor 2; NSAIDs, non-steroidal anti-inflammatory drugs; ROS, reactive oxygen species; SOD, superoxide dismutase.

Figure 1. Molecular mechanism of oxidative stress and potential targets for antioxidant treatment in aging-related diseases. ARE, antioxidant response elements; CAT, catalase, GLP-1, glucagon-like peptide 1;GPX, glutathione peroxidase; GST, glutathione transferase; HO-1, heme oxygenase 1; Keap1, Kelch-like ECH-associated protein 1; NF-κB, nuclear factor kappa-light-chain-enhancer of the activated B cells; Nrf2, nuclear factor-erythroid 2-related factor 2; NSAIDs, non-steroidal anti-inflammatory drugs; ROS, reactive oxygen species; SOD, superoxide dismutase.

Non-enzymatic antioxidants include those produced naturally by cells, such as glutathione, uric acid, bilirubin, and iron-sequestering ferritin, and those that are not produced naturally and must therefore be administered through diet or supplements, such as certain vitamins (A, C, and E), and polyphenols such as curcumin and resveratrol, among many others [Citation63].

It is precisely the homeostasis of this system that is altered during ageing, since on the one hand there is an increase in ROS production, and a parallel decrease in the activity of enzymatic antioxidant systems, leaving cells susceptible to oxidative damage. The increase in free radicals is largely due to mitochondrial dysfunction, in which the efficiency of the electron transport chain is reduced, leading to increased electron leakage parallel to reduced ATP production. This has been studied in different models and tissues, showing that for example in cardiomyocytes complexes III and IV are those that lose their activity with age [Citation64–66], whereas in brain, skeletal muscle and liver complex I is the most sensitive to ageing [Citation67]. On the other hand, the deterioration of antioxidant systems has also been studied, as exemplified by a study in which the activity of the enzyme GPX was measured in a cohort of disabled older women, observing that after the age of 65 years, GPX activity decreased with age [Citation68]. In addition, studies in different models have shown that the expression of SOD decreases with age in cartilage, thereby increasing the risk of developing degenerative diseases such as osteoarthritis [Citation69–71].

Mitochondrial dysfunction represents a common mechanism of damage in various diseases related to ageing, in this sense, in addition to the aforementioned defect in the efficiency of the electron chain, multiple changes have been described in the mitochondria that alter its function, such as; mitochondrial DNA (mtDNA) mutations [Citation72], alterations in mitochondrial proteolysis [Citation73,Citation74], alterations in the dynamic processes of mitochondrial fusion, fission and mitophagy [Citation75–77], among others. It is noteworthy that mtDNA, unlike the cell's nuclear DNA, is more exposed to damage from ROS generated in the mitochondrial inner membrane and has limited repair mechanisms to counteract mutations without histone protection, and it is precisely the accumulation of mtDNA mutations that is one of the central mechanisms that explain mitochondrial dysfunction during aging [Citation78,Citation79]. In this context, during the last decade, various therapies have been described based on lifestyle changes that promote the consumption of natural antioxidants and physical activity, since both processes are presumed to be related to regulating mitochondrial function, which would help treat diseases and prolong longevity [Citation80–84].

In this way, ageing is naturally related to an increase in oxidative stress, which leads to damage and the development of various diseases, although there are various factors that can contribute to modulating this process. In this way, we can describe those factors that accelerate this process, such as the consumption of drugs, alcohol, smoking, sedentary lifestyle, overeating, radiation, atmospheric pollutants, among others [Citation85–88]. It is also possible to describe those factors that reduce oxidative stress, such as the consumption of foods rich in antioxidants or physical activity. Thus, oxidative stress becomes a pharmacological target for the treatment and prevention of various diseases associated with ageing.

4. Effect of aging diseases and their treatment on the redox balance

As mentioned above, the increase in life expectancy due to improved access to healthcare and drug development has been accompanied by an increase in the prevalence of multimorbidity, which is the main driver of polypharmacy.

On the other hand, longevity assurance processes are intricately governed by a group of genes known as vitagenes which play a pivotal role in maintaining cellular homeostasis, particularly in stress conditions, while also exerting control over the aging process. These genes encode various proteins, including heat shock proteins (Hsp). This is a crucial factor in understanding and potentially mitigating the impact of these health challenges due the role of Hsp and their response in establishing a cytoprotective state across a wide spectrum of human diseases such as inflammatory disorders, cancer, aging-related ailments, and neurodegenerative disorders [Citation89,Citation90].

Recent scientific investigations have shed light on the protective properties of dietary antioxidants that have been demonstrated to confer protection through the activation of hormetic pathways, which notably include vitagenes. Within these pathways are Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), Nrf-2/ARE pathway and other kinases and transcription factors [Citation90–92]. The hormetic response and polypharmacy are interconnected through their impact on medication management and healthcare decisions leading to personalized medication approach.

The following section aims to address the development and interaction between different age-related diseases, the main link of which is oxidative stress. In addition, some drug interactions between the different diseases will be described, with the aim of making it clear that the comprehensive treatment of the elderly and their pathologies should be superimposed on the individualized management of each condition.

4.1. Hypertension

According to the WHO, the current definition of hypertension is systolic blood pressure (SBP) equal to or greater than 140 mmHg and/or diastolic greater than 90 mmHg with at least two measurements in consultation and on at least two separate occasions to diagnose it [Citation93] is more prevalent in older people and affects 1.28 billion people worldwide. In recent years, there has been an increase in its prevalence, with 76% of adults aged 65–74 years and 82% of adults aged 75 years and older suffering from hypertension [Citation94–96]. Hypertension is one of the major risk factors predisposing to fatal complications of CVD [Citation97], being a key contributor to the development of stroke, myocardial infarction, heart failure and renal failure [Citation93].

The pathophysiology of essential hypertension is directly related to the development of other age-related diseases, where chronic inflammation and increased oxidative stress are mainly responsible for endothelial dysfunction, which together with impaired compensatory blood pressure mechanisms, such as renal dysfunction leading to increased sodium absorption, abnormal renin-angiotensin-aldosterone system (RAAS) response and overactivation of the sympathetic nervous system, lead to increased total peripheral resistance and afterload, contributing to the development of hypertension [Citation93].

Treatment is the most frequent reason for medical visits and use of chronic prescription drugs and adherence to treatment is poor, with 46% of adults with hypertension unaware that they have the disease. Hypertension can be managed by monotherapy and combination therapies, the latter being the most common, aggravating polypharmacy. There are angiotensin-converting enzyme (ACE) inhibitors, such as enalapril and lisinopril; angiotensin-2 receptor blockers (ARBs), such as losartan and telmisartan. Both ACE inhibitors and ARBs have a protective effect on the kidney, as well as relaxing blood vessels. Dihydropyridine calcium antagonists, such as amlodipine and felodipine, also relax blood vessels. Other drugs used include beta-blockers, such as metoprolol and carvedilol, and thiazide diuretics, such as hydrochlorothiazide and chlorthalidone, which remove excess sodium and water from the body, reducing blood pressure. Among these drugs, the most important are those that pharmacologically target the RAAS, since it has been shown that the decrease in angiotensin II and aldosterone activity results in a decrease in oxidative stress and the release of inflammatory mediators at the cellular level, thereby improving endothelial dysfunction [Citation98]. Thus, research with losartan has shown a reduction in oxidative stress after 12 months, with a decrease in serum and urinary levels of 8-hydroxy-deoxyguanosine (8-OHdG) and nitrotyrosine, as well as an increase in SOD activity. However, this effect is not observed with all antihypertensives, because in this same study Amlodipine did not obtain significant decreases after 12 months in the aforementioned markers [Citation99].

Therapeutic goals in older persons require multiple considerations in relation to their comorbidities, clinical, psychosocial and functional contexts, overall prognosis and goals of care, as all older persons are different, for example, in older patients with high functional reserve aggressive hypertensive treatment would be of great benefit, whereas in frail older patients it would increase adverse events such as orthostasis, falls, syncope and cognitive impairment [Citation100]. Another consideration is the interaction of pharmacotherapy for hypertension with pharmacotherapy for other age-related diseases, for example, the use of nonsteroidal anti-inflammatory drugs (NSAIDs) leading to hypertension leads to an increase in the dose of antihypertensive drugs, leading to an increased risk of AKI [Citation101]. Another common example is the prescription of diuretics to counteract edema secondary to the use of dihydropyridine calcium channel blockers, which significantly decreases the quality of life of older people [Citation102].

It is in this context that redox balance modulation takes on great importance, since the most relevant drugs in the treatment of hypertension are those aimed at reducing endothelial dysfunction by reducing oxidative stress and inflammation, which not only has implications for the management of blood pressure, but also for the treatment of other age-related diseases such as Alzheimer's disease and DM2, as will be discussed below.

4.2. Alzheimer's disease

Among the dementias, Alzheimer's disease (AD) is the most common. AD is a neurodegenerative disease characterized by marked latent cognitive impairment, loss of memory and spatio-temporal orientation. Patients with advanced AD cannot recognize their relatives or where they are. This disease can have a prolonged silent course, however, it can present cognitive impairment to varying degrees which is important to suspect in order to provide a comprehensive approach to the patient and their family early on. Currently, AD is becoming increasingly lethal and is a major cost to the healthcare system, for example, in the United States in 2020 and 2021 it was ranked as the seventh leading cause of death nationwide and the economic costs associated with medical care, long-term care and palliative care for people aged 65 years and older with dementia amounted to $321 billion [Citation103,Citation104]

The brain has characteristics that make it particularly vulnerable to oxidative stress, including its high oxygen consumption (approximately 20% of the oxygen supplied by the respiratory system), its high energy and metabolic demand, as well as its composition of neurons, which are cells with a low antioxidant defense, low regenerative capacity and whose membranes are rich in long-chain polyunsaturated fatty acids (PUFAs) that are preferential targets of ROS in the lipid peroxidation reactions that occur in living cells [Citation105]. As for the pathophysiology of AD, the key point is the deposition of amyloid-β (Aβ) (whose oligomeric forms are the most neurotoxic by causing oxidative stress), which is considered the main inducer of the disease, a phenomenon to which is added Tau hyperphosphorylation, tangle formation and mitochondrial dysfunction, which ultimately lead to neurodegeneration and cognitive impairment.

Oxidative stress would be one of the main promoting factors in Alzheimer's disease as it is the direct result of the damage generated by the accumulation of Aβ, as well as participating directly in the pathogenesis of the disease by promoting the production and aggregation of Aβ, as well as facilitating the phosphorylation of Tau, which leads to a vicious circle marked by neurotoxicity and neuroinflammation, which ultimately leads to neuronal death (Zhao, Yan, 2013). For example, one of the main mechanisms of oxidative stress damage is lipid peroxidation of PUFAs, which has been associated with plasma membrane damage, as well as an increase in toxic second mediators such as 4-hydroxy-2-nonenal aldehyde HNE, malondialdehyde (MDA) and ceramides. In this regard, natural fat-soluble antioxidants such as vitamin E have been shown in in vitro studies to decrease lipid peroxidation and with it toxic mediators [Citation106–109]. As for damage to nucleic acids and proteins, this has been verified by studying biomarkers of oxidative stress in AD, with the accumulation of products such as 8-OHdG, 8-hydroxyguanosine, protein carbonyls and 3-nitrotyrosine residues [Citation110–112]. This whole process of macromolecule damage is aggravated by alterations in the concentrations of transition metals such as iron, zinc and copper, and by alterations in the function of NO, which physiologically at the brain level has functions linked to synaptic plasticity, sleep regulation, appetite and neurosecretion [Citation113–118].

As for the relationship of AD with other age-related comorbidities, a strong association has now been proven, given that those with various comorbidities are at greater risk of suffering and exacerbating cognitive decline [Citation119]. In this regard, the most common comorbidities in people with dementia include DM2, CVD and hypertension. For example, hypertension as a risk factor for the development of AD is directly linked to the role of angiotensin II, which through its G protein-coupled receptor AT1 pathway, induces a variety of deleterious effects at the brain level, including the induction of proinflammatory pathways, ROS synthesis and apoptosis [Citation120,Citation121]. Against this background, treatment with different RAAS modulators has been shown in in vitro and in vivo models to have a beneficial role in the prevention and treatment of cognitive impairment, with ARBs and ACE inhibitors having the strongest evidence. ARBs have shown some superiority in the prevention of cognitive decline compared to ACE inhibitors, for example some clinical trials with telmisartan alone or in combination with other drugs such as rosuvastatin, have been shown to reduce the risk of incidences of dementia or cognitive decline in certain hypertensive patients, however these are still preliminary results that need to be studied with further evidence [Citation122,Citation123].

Currently, there is no curative treatment for AD, as the main focus is on treating symptoms according to the stage of the disease with the aim of providing well-being, dignity and independence for individuals and their families over a longer period of time. For mild to moderate dementia, the pharmacological treatment of choice is cholinesterase inhibitors, such as donepezil, rivastigmine and galantamine, which increase cholinergic transmission by inhibiting cholinesterase in the synaptic gap [Citation124]. A study done in another line of research in relation to metabolic syndrome has shown that galantamine can increase antioxidant enzyme activity, such as SOD and CAT activity, as well as decrease lipid peroxidation and systemic nitrite levels [Citation125]. In this line, a recent study showed an identification of polyphenols as potential inhibitors of cholinesterase and oxidative stress for their antioxidant properties useful in the treatment of AD [Citation126]. Another drug used is Memantine, which has been considered as a neuroprotectant by antagonizing the overstimulation of the N-methyl-D-aspartate (NMDA) receptor [Citation127]. Thus, combined treatment with memantine and cholinesterase inhibitors can be considered, being more useful for AD, but less acceptable to patients [Citation128,Citation129]. However, caution should be exercised with cholinesterase inhibitors in older adults, as they potentiate vagal tone and are contraindicated in cases of initial bradycardia or cardiac conduction pathologies due to the risk of syncope, falls and fractures. In contrast, memantine has no significant side effects [Citation130]. Another precaution to consider in the treatment of people with cognitive impairment is the inappropriate prescription of drugs with anticholinergic effects, which are often prescribed for the treatment of other age-related conditions.

Thus, finally, current evidence supports that the most cost-effective management seems to be the prevention of AD [Citation131], which is achieved with adequate control of the main risk factors, including comorbidities, which as we have seen, are directly related to each other and must be managed in a comprehensive manner.

4.3. Osteoarthritis

Osteoarthritis (OA) is the most common form of arthritis. Over time it degrades cartilage, causes bone remodeling and produces formation of osteophytes visible on radiological examinations. This produces alterations and clinical manifestations such as pain, stiffness, swelling and limitations in joint function, producing high mobility, especially in older adults. Joint's alterations commonly occur in the knee, hip, hand, foot and ankle [Citation132]. OA affects 1 in 3 people over age 65 and is more frequent in women than men. The main risk factors are obesity, physical inactivity, and joint injury [Citation133,Citation134]. Effects of aging on cartilage play an important role in the pathophysiology of OA as well as cellular senescence, inflammation, mitochondrial dysfunction, oxidative stress and energy metabolism dysfunction [Citation135]. OA is associated with chronic low-grade systemic and local inflammation including active synovitis, term known as ‘inflammaging’ provoked by a continuous antigenic load and stress [Citation136]. An important mechanism and a hallmark of aging in OA is the senescence-associated secretory phenotype which increases production of proinflammatory mediators and matrix-degrading enzymes, maintaining the cell in a cycle arrest [Citation137]. Other support for the inflammation is the visceral fat mass that is associated with a decline in muscle mass greater in older adults and with a higher adipocyte and proinflammatory macrophages, increasing cytokines and adipokines [Citation138,Citation139]. Moreover, there exists the term ‘meta-inflammation’ which is related to metabolic syndrome associated with OA and obviously individual factors that are crucial at this point and intermingles with OA such as diabetes, dyslipidemia, and hypertension [Citation140]. These factors can all damage the joint tissue and promote matrix destruction with an alteration of gene expression in chondrocytes [Citation141]. In aged mice, the intrinsic circadian clock is disrupted and in humans OA chondrocytes the alteration in the circadian clock is associated with stress-induced senescence in fibroblasts [Citation142]. Another hallmark of ageing in OA is the mitochondrial dysfunction, leading to the development of the ageing phenotype and oxidative stress [Citation143]. OA chondrocytes have mitochondrial biogenesis impairment, thereby producing chondrocyte pro catabolic responses. Hydrogen peroxide (H2O2) downregulates the expression of mitochondrial antioxidant enzymes and may escape from the mitochondria causing oxidative damage to cellular macromolecules [Citation144–146].

Mice with OA showed elevated intracellular and mitochondrial superoxide anion radical (O2·_) generation and a decrease in mitochondrial SOD2 together with increased degeneration cartilage age-related. However, the use of antioxidants such as a derivative of vitamin C suppresses mitochondrial superoxide generation and ameliorates cartilage degeneration in vivo [Citation147].

In synovial fluid xanthine oxidoreductase level is elevated in OA and its amount correlates with the severity of joint damage [Citation148]. A reduction in extracellular matrix gene expression and protein synthesis is observed in the anormal function of human chondrocyte insulin-like growth factor 1 signaling due to oxidative stress [Citation149].

The thiol groups that are in proteins which contain reactive cysteines suffer oxidative post-translational modifications in OA chondrocytes, a process called S-sulfenylation that promoted the production of matrix metalloproteinase 13 and increases the cartilage destruction [Citation150]. Hyperoxidation leads to age-related inhibition of pro-survival cell signaling and chondrocyte cell death and the expression of CAT prevents this phenomenon [Citation151]. On the other hand, ferroptosis, a type of nonapoptotic iron-dependent cell death associated with lipid peroxidation and modulated by GPX4 is involved, at least in part, in OA pathogenesis. A recent study showed that iron level in synovial fluid increased with the progression of OA as well as Fe2+, Fe3+ and total iron concentrations, thus suggesting an iron accumulation during OA progression. GPX4 function is decreased in OA along with decrease of glutathione (GSH) contents in the cartilage. Downregulation of GPX4 increased the sensitivity of chondrocytes to oxidative stress and mitochondrial morphological alteration together with degradation of extracellular matrix through MAPK/NF-κB pathway [Citation152].

There are non-pharmacological treatments based on exercise, weight loss, braces and foot orthoses and education [Citation153]. On the other hand, the classic pharmacological is NSAIDs that should only be used during periods when symptoms are present. Depending on the patient's clinical setting topical capsaicin, duloxetine, and intraarticular glucocorticoids may be required [Citation154], but may have deleterious effects on the hyaline cartilage and may accelerate OA progression [Citation155]. NSAIDs have several anti-inflammatory, analgesic and antipyretic benefits. Although NSAIDs are the first line of treatment, it produces adverse effects in our organism especially when it is self-medicated which include gastrointestinal, cardiovascular, hepatic, renal, cerebral and pulmonary problems. They are toxic compounds due to the activation of mitochondrial oxidative stress and the key target for some of them is the mitochondrial electron transport chain complex-I, leading to abnormal redox-active chain reactions [Citation156].

Given the polypharmacy of patients who use NSAIDs, these drugs can potentiate gastrointestinal, cardiovascular, and renal complications dose-dependent. Gastrointestinal damage are upper gastrointestinal complications, especially peptic ulcer and also can lead to impairment of intestinal mucosal barrier function producing proinflammatory cytokine cascade and NLRP3 inflammasome activation which both attract the neutrophils causing oxidative burst, chronic inflammation, apoptosis and eventually intestinal ulceration [Citation157,Citation158]. Main cardiovascular complications are major vascular events, heart failure and stroke [Citation159]. Regarding kidney complications, users of NSAIDs have 3-fold greater risk for AKI [Citation160] and the treatment should be used with caution in patients with hypertension or heart failure because both treatments significantly increase the risk of AKI and alters the hemodynamic equilibrium [Citation101].

Due to the complexity of OA development it's impossible to analyze all causal factors. However, OA is certainly an ageing-associated process although it has its own independent processes. The treatment with NSAIDs produces a vicious circle of damage and oxidative stress given the chronic use of these drugs.

4.4. Cancer

Another age-related disease is cancer, which has increased in prevalence worldwide, with significant economic and social repercussions. Older adults will account for about 60% of all newly diagnosed cancer cases projected to 2035, with prevention among young adults a key pillar. The main cancers diagnosed worldwide are lung, colorectal, prostate, stomach and breast cancer [Citation161,Citation162]. Regarding the relationship of this pathology with polypharmacy, it has been reported that polypharmacy exceeds 90% in older adults diagnosed with cancer, being even higher in those receiving radiotherapy because the adverse effects of this therapy must be managed with more drugs, thereby increasing their consumption [Citation163]. The link between cancer and oxidative stress can be approached in two contexts, firstly in the pathophysiology that initiates cancer progression and secondly in cancer therapy, where pro-oxidant and antioxidant substances stand out.

Various studies have shown that cancer cells in general have a redox imbalance in favor of the oxidative burden, for example, it has been observed that in lung cancer there is a decrease in vitamin E and a depletion of serum antioxidant capacity, together with an increase in lipid peroxidation, leading to a significant increase in systemic oxidative stress [Citation164]. The increase of ROS in cancer cells, observed to come mainly from mitochondria and the enzyme NOX, thus dysregulation of NOX activity or mitochondria has been found to be linked to transformation and progression of some common cancers, such as bladder, breast, kidney, colon, stomach, chronic lymphocytic leukemia among others [Citation165–171]. This prooxidant state in cancer induces a variety of biological effects including increased cell proliferation and survival [Citation172–174], DNA damage and genomic instability [Citation175,Citation176] and drug and radiotherapy resistance [Citation177,Citation178]. However, this pro-oxidant state is in equilibrium, as toxic levels of ROS induce cell cycle arrest, apoptosis and senescence, whereby cancer cells develop mechanisms by which they partially decrease ROS levels. For example, mutations in the Nrf2/Keap1 system have been described that generate a higher expression of Nrf2 and its target genes, increasing the transcription of antioxidant enzymes [Citation179,Citation180], and tumor cells also adapt to hypoxia by increasing glycolysis metabolism, in a phenomenon known as the Warburg effect, which also plays a role in redox homeostasis by inducing reducing agents such as NADPH via the pentose phosphate pathway or GSH via glutaminolysis [Citation181,Citation182]. In this way, tumor cells manage to maintain a prooxidant redox state that favors tumorigenesis, without reaching high levels of ROS that interfere with their progression.

On the other hand, treatment can also create an environment of oxidative stress. In this sense, radio- and chemotherapeutic drugs have as their main objective to increase ROS production to toxic levels, depleting antioxidant systems and causing apoptosis of tumor cells. An example of this are the anthracyclines, which are cytotoxic drugs commonly used in the treatment of a broad spectrum of cancers. The main theory currently accepted to explain the generation of ROS by anthracyclines indicates that they are forming an unstable metabolite semiquinone, which when oxidized by the mitochondria is accompanied by the release of large quantities of ROS together with apoptosis and ferroptosis [Citation183]. Another theory indicates that anthracyclines form complexes with iron which induces lipid peroxidation, protein carbonylation, DNA oxidation by ROS, leading to tumor cell death by apoptosis and ferroptosis [Citation184]. While these pro-oxidant therapies are often favorable for cancer treatment, they also induce long-term damage in other organs distant from the origin of the cancer, given the non-targeted nature of oxidative damage. Thus, for example, multiple side effects have been described in the use of chemotherapeutics, including cardiotoxicity, neurotoxicity, hepatotoxicity, among others, most of them mediated by oxidative stress [Citation185–193]. In this context, antioxidants have been tested to reduce the side effects of anticancer therapy, including the use of polyphenols, which have been shown to have an antioxidant and anti-inflammatory effect by modulating the NF-κB signaling pathway (), which is the main pathway involved in lung, liver and colorectal cancer [Citation194]. Thus, the net effect of the polyphenol is to reduce cytokines and ROS, increase cancer cell apoptosis and enhance vasodilator function through activation of endothelial NOS (eNOS) [Citation195]. In addition, vitamin C also activates eNOS through the same pathway as polyphenols (PI3 kinase/Akt signaling) [Citation196]. Therefore, the use of antioxidants may serve as a complementary therapy for cancer treatment by protecting normal tissues from oxidative stress damage, while enhancing the efficacy of chemo- and radiotherapy.

4.5. Obesity and metabolic disorders

Obesity is a complex multifactorial disease. According to WHO, it is defined as ‘excess or abnormal fat accumulation that presents a risk to health’ [Citation197]. This pathology has an worldwide impact due its increase of prevalence and public health management. Nowadays, despite the fact that body mass index (BMI) has low sensitivity and there is a large inter-individual variability in the percent body fat, it is still used in the clinical setting. According to this index, nearly a third of the world's population is classified as overweight or obese in all ages and both sexes, although the prevalence is generally in older persons and women [Citation198]. The importance of obesity is rooted in its relation with sarcopenia [Citation199], a prevalent condition in elderly, which is overlooked and undertreated in the biomedical setting. This interconnection leads to sarcopenia or even osteosarcopenia exacerbation due to fat infiltration into muscle, hampering of physical functions and underscore adverse outcomes [Citation200].

Intrinsically, obesity is related to many conditions such as metabolic syndrome, DM2, CVD, and hypertension [Citation201]. Oxidative stress is involved in their pathophysiology. While the majority of mechanisms mentioned in other diseases contribute to the pathophysiology of oxidative stress in obesity, there exists a distinct group of antioxidant enzymes known as the paraoxonase family. Notably, paraoxonase 1 is associated with high-density lipoprotein (HDL) and plays a role in hydrolyzing oxidized low-density lipoprotein-cholesterol (LDL). This enzyme possesses atheroprotective properties, but its activity is reduced in conditions such as DM2, metabolic syndrome, hypercholesterolemia, and chronic renal failure [Citation202,Citation203]. Previous studies showed a positive correlation between oxidative stress and insulin resistance with increased LDL as well as low HDL in animal models. HDL sub particles have antioxidant activity and in overweight individuals its antioxidant mechanism is dysfunctional which leads to more metabolic disorders and is associated with accelerated atherosclerosis due to increased macrophage infiltration [Citation204,Citation205]. Moreover, ROS can influence the activity of enzymes involved in lipolysis and lipogenesis [Citation206] and play a role in signal transduction and the regulation of adipocyte differentiation and it is influenced by various factors that are susceptible to redox metabolism, including transcription factors, cell-cycle proteins, hormones, and small molecules. In preadipocytes, NADPH has a role in the generation of ROS [Citation207] On the other hand, mature adipocytes (white, brown, and beige) have different functions throughout life that contribute to maintaining adequate adaptive thermogenesis and homeostasis. In this line, ROS may play a crucial role in the browning of adipose tissue, potentially mitigating obesity, as certain natural compounds with antioxidant properties have been shown to facilitate browning [Citation208]. Chronic excessive nutrient availability leads to disruptions in mitochondrial homeostasis and induces abnormalities in biogenesis, ROS production, and apoptosis [Citation209]. This is aggravated by its association with insulin resistance and DM2 due to elevated glucose levels that contribute to increased ROS production, modifying mitochondrial enzymes and contributing to the development of metabolic disorders. Impaired mitochondrial function in adipocytes is associated with low grade inflammation, disturbances in adipogenesis, lipolysis, fatty acid esterification, adiponectin production and apoptosis [Citation210]. After the mitophagy process undergone by the mitochondria, they are replaced in a process that produces more ROS [Citation211], so it's a vicious circle. Another mechanism occurs when exposed to ROS, redox-sensitive transcription factors like NF-kB become activated, leading to the transcription of various proinflammatory cytokines. This process contributes to an escalation in ROS overproduction, especially H2O2 which affects the degradation of IκB, an NF-κB inhibitor [Citation212,Citation213].

Leptin is an adipokine that regulates hunger and appetite and food intake. In overweight individuals there is a elevation of this molecule leading to secretion of pro-inflammatory cytokines, such as TNF-α and IL-6 as well as TGF-β and has a role in cardiac fibrosis and oxidative stress through mTOR pathway [Citation214].

Otherwise, adiponectin is an anti-inflammatory and insulin-sensitizing hormone secreted by adipose tissue and in obese patients is diminished. Adiponectin exhibits anti-inflammatory effects by decreasing the levels of NF-κB [Citation215]. Increased concentrations of adiponectin result in reduced production and activity of TNF-α, IL-6, and other interleukins, promoting lipolysis and stimulating de novo synthesis and secretion of hepatic fatty acids [Citation216].

This same chronic inflammation also increases hepcidin, a hormone which regulates iron metabolism. The elevated levels of hepcidin leads to iron deficiency [Citation217]. Despite this, iron triggers oxidative stress due to its redox activity as a metal. Additionally, it contributes to inflammation, and endocrine dysfunction. Intracellular free iron is cytotoxic as it promotes the Fenton reaction, further intensifying oxidative stress, producing hydroxyl radical [·OH] () [Citation218].

The treatment for obesity and metabolic syndrome is non-pharmacological as an essential pillar. This includes regular physical activity, healthy diet, behavior modification and others actions that include a supportive environment. The diet should be a low-fat low-carbohydrate diet with plenty of antioxidants such as vitamin C and E, and polyphenols [Citation208].

The pharmacological treatment is reserved for those with a BMI >30 kg/m2, or a BMI of 27–29.9 kg/m2 with weight-related comorbidities, who have not met weight loss goals [Citation219]. This is based especially on Glucagon-like peptide-1 (GLP-1) agonists such as Liraglutide or Semaglutide. G⁣⁣LP-1, an incretin hormone, stimulates insulin secretion, suppresses glucagon secretion, and reduces food intake. Besides its hypoglycemic effects, GLP-1 exhibits anti-inflammatory, antioxidative and vascular protective properties in various cells and tissues as well as the suppression of NF-κB [Citation220,Citation221]. In terms of oxidative stress, GLP-1 engages the Nrf2-ARE pathway, which decreases oxidative stress markers and activates genes associated with mitochondrial biogenesis and preservation, including mitochondrial transcription factors [Citation222,Citation223]. Another GLP-1 analog, exendin-4, has demonstrated the ability to reduce H2O2-induced ROS production and enhance the synthesis of antioxidant enzymes such as CAT, GPX, and SOD through Nrf2 [Citation224,Citation225]. In this context, it is necessary to consider whether the patient has DM2, their frailty, and whether they are taking other hypoglycemic medications to take into account the risk of falls and hypoglycemia, as well as adverse effects. To end, obesity, metabolic syndrome, and oxidative stress are interconnected conditions that have a significant impact on overall health and well-being and contribute to the development and progression of various chronic diseases that may be managed with antioxidants drugs.

4.6. Ischemic heart disease

Ischemic heart disease is a condition in which there is reduced blood flow to the heart muscle due to the narrowing or blockage of the coronary arteries. The majority of cardiac deaths in adults from countries of any income are due to ischemic heart disease secondary to coronary atherosclerosis [Citation226], a key development factor related to chronic inflammation, oxidative stress, disrupted homeostasis, imbalance in growth, proliferation and migration cell regulation [Citation227]. It is associated with an imbalance of blood supply to the myocardium due to obstruction of the epicardial coronary arteries, often caused by plaque buildup in the wall of the arteries that may cause vascular obstruction and ischemia [Citation228]. During the ischemic phase, there is a decrease in oxygen supply and cellular changes inside that produce an interruption of cellular metabolism and derangements of cellular membrane pumps, causing the anaerobic to predominate over the aerobic metabolism which decreases the cellular pH thus producing that Na+/H+ exchanger excretes excess hydrogen ions, which produces an influx of Na + . In this phase there is also a endoplasmic reticulum stress, decrease intracellular ATP, reduction of active Ca2+ efflux, limiting its reuptake by other organelles such as endoplasmic reticulum or mitochondria, producing an abnormal rise of this ion in the cell which then mediate excitotoxic cell death. These intracellular changes occur in conjunction with the opening of the mitochondrial permeability transition pore (mPTP), which causes depolarization of the mitochondrial membrane potential, further impairing ATP production. Ca2+ at high concentrations can contribute to activation of proteases, calpains, and endonucleases, which cause damage to the muscular heart tissue, and DNA, and cell death [Citation229–231].

On the other hand, the reperfusion phase, frequently through percutaneous coronary intervention (PCI), produces a burst of O2·_ that causes further damage. Indeed, a reperfusion paradoja occurs since the damage can reach up to 50% of the final size of the infarct, term called myocardial ischemia-reperfusion injury (IRI) [Citation232]. ROS production is an important event in cell signaling transduction maintenance. However, different mechanisms occur in this phase due to disruption of the respiratory chains, commonly associated with complex I, which can lead to an excessive ROS production, aggravating the damage, causing DNA, lipids and protein peroxidation. In fact, it has been known for many years that this phase accelerates the development of necrosis [Citation233] and there is still no effective treatment to prevent myocardial IRI. Other mechanisms are calcium overload, opening of the mPTP, endothelial dysfunction, prothrombogenic phenotype, and inflammatory responses [Citation232]. The main IRI ROS is O2·_ which is produced by mitochondria and xanthine oxidase, NOX, cytochrome P450 oxidases and uncoupled NOS. Under physiological conditions is not toxic due the fast dismutation to H2O2, a conversion accelerated by SOD. However, under low pH conditions, O2·_ converted to its conjugate acid, the more highly potent oxidant, hydroperoxyl radical (HOO•) and although H2O2 is less reactive, it diffuses across membranes and can act as a second messenger, participating in the generation of •OH, through the Fenton reaction () in the presence of transition metals [Citation231,Citation234,Citation235]. Besides that, in myocardial IRI there are proinflammatory processes due to the release of cytokines like TNF-α, IL-1β and IL-6 and chemokines which also produces autophagy, necrosis, apoptosis [Citation236].

Globally, the damage is given by both phases. However, the reperfusion phase is a dynamic process that not only occurs at a single time when performing PCI, but contractile and endothelial dysfunction and cell death can continue up to 3 days after the start of reperfusion [Citation237].

Oxidative stress-induced endothelial dysfunction is an early event in the development of this disease, leading to an imbalance between the availability of NO and ROS. This dysfunction is characterized by abnormal regulation of blood vessel tone and growth, impaired anti-inflammatory and anti-thrombotic properties of the endothelium, and disturbances in vascular remodeling [Citation238]. A healthy endothelium plays a crucial role in responding to vascular stress by inducing vasorelaxation, controlling vascular permeability, and preventing platelet aggregation. However, it is highly reactive to various stimuli, including NO, to maintain proper vascular tone and structural integrity. NO, synthesized by eNOS, plays a significant role in the body's antioxidant defense system and reduced bioavailability of NO primarily occurs due to increased degradation by ROS and decreased expression of eNOS. Additionally, uncoupling of eNOS can further decrease NO availability as it shifts its enzymatic activity to generate O2·− and H2O2 instead of NO [Citation239]. On the other hand, angiotensin II and thrombin can upregulate NADPH in response to ROS [Citation240,Citation241].

As previously mentioned, atherosclerosis is the primary underlying cause of ischemic heart disease. It is characterized by a chronic inflammatory condition involving the interaction between immune cells and endothelial cells, facilitated by adhesion molecules present on the surface of the vascular endothelium [Citation242]. Excessive production of ROS contributes to the formation of atherogenic oxidized low-density lipoprotein, a key factor in the development of atherosclerosis [Citation243]. Additionally, ROS overproduction leads to an increase in NADPH levels and impairs mitochondrial capacity, resulting in mitochondrial dysfunction [Citation244]. During the progression of atherosclerosis, accumulated neutrophils generate additional ROS and activate poly [ADP-ribose] polymerase (PARP), causing damage to mtDNA and further ROS production. This oxidative stress and the activation of various cellular pathways lead to the release of inflammatory cytokines, contributing to vascular inflammation and participating in the oxidation process of LDL. These effects are further exacerbated by factors such as age, obesity, smoking, hypertension, DM2, and dyslipidemia [Citation245]. Clinically, the first step in the resolution of heart ischemia in patients who meet the criteria for ST elevation myocardial infarction are thrombolytics or PCI, which is strongly preferred. At this point, patients usually require nitrates, dual antiplatelet therapy, anticoagulation drugs, beta blockers, statins, ACE inhibitors, morphine and oxygen. The long term management includes use of multiple medications prior to being beneficial in the secondary prevention of atherosclerotic disease and congestive heart failure [Citation246].

On the other hand, current research has not provided evidence that antioxidants like flavonoids and vitamins decrease the risk of acute myocardial infarction (AMI) or mitigate myocardial IRI in humans [Citation247,Citation248]. However, other studies have demonstrated that cocoa flavonoids exhibit anti-inflammatory properties, reduce oxidative stress, and inhibit myocardial apoptosis following AMI and reperfusion. These effects are achieved by counteracting membrane peroxidation and nitro-oxidative stress, as well as by reducing levels of inflammatory markers such as IL-6 and NF-κB [Citation249].

It should be noted that the phenomenon of ischemia-reperfusion not only happens in myocardial infarction, but also in stroke, and peripheral vascular disease [Citation231], pathologies that also increase with age and have different management.

4.7. Diabetes mellitus 2

The American Diabetes Association defines DM2 as a progressive loss of adequate β-cell insulin secretion frequently on the background of insulin resistance that manifests clinically as hyperglycemia. Various genetic and environmental factors are involved [Citation250]. The worldwide prevalence is 10.5 percent among adults aged 20–79 years and it is estimated to affect 537 million adults [Citation251].

Chronic hyperglycemia leads to an imbalance between the antioxidant defense system and ROS production with predominance of the latter. Consequently, the development of oxidative stress contributes to diabetic complications by damaging cells, tissues, and organs. It can impair insulin cell signaling, promote endothelial dysfunction, cause DNA damage, and induce inflammation. These effects contribute to the pathogenesis of complications associated with DM2, such as macrovascular damage and microvascular damage (nephropathy, retinopathy, and neuropathy) [Citation252,Citation253].

The main mechanisms are associated with glucose and lipid metabolism [Citation254], which include glucose autoxidation, mitochondrial dysfunction, activation of protein kinase C (PKC) pathway, advanced glycation end products (AGEs) formation and accumulation, and inflammation.

Glucose oxidation pathway is essential for glucose to be oxidized in body cells and is composed of multiple steps that under normal conditions result in increased mitochondrial production of O2·_. In hyperglycemic conditions, the antioxidant compensatory mechanism will become overwhelmed, thereby resulting in the production of macromolecules damage, such as DNA oxidation, which responses with activation of PARP-1, a repair enzyme. Consequently, PARP-1 increases the levels of glycolytic intermediates, thus increasing H2O2 production, as well as the metabolic flux across pro-oxidative pathways, such as AGE, PKC, hexosamine and polyol ones [Citation255]

Advanced glycation end-products are a complex group of molecules formed through a process called non-enzymatic glycation. This process is accelerated in conditions of hyperglycemia and has an important role in CVD progression in DM2. AGEs can be formed both within the body (endogenously) and in foods during cooking or processing (exogenously). In the context of DM2, endogenous AGEs are of particular interest due to their formation involving a series of chemical reactions that result in the irreversible modification of macromolecules due to cross-linking of proteins, cellular dysfunction, oxidative stress, inflammation and activation of other oxidative stress-related pro-oxidative pathways such as that of PKC when AGEs bind to their receptor [Citation256,Citation257].

The presence of AGEs, particularly in connective tissue, results in age-related damage. It has been demonstrated that the covalent cross-linking of adhesion proteins in the extracellular matrix contributes to the loss of skin and blood vessel elasticity, as well as the degeneration of cartilage, ligaments, and the lens. The accumulation of AGEs, along with the buildup of lipofuscin, is associated with the process of aging and chronic diseases related to age. Another AGEs ageing hallmark is the activation of NF-κB pathway in patients with age-related macular degeneration [Citation258,Citation259]. Uncontrolled DM2 leads to the activation of the polyol pathway, which results in intracellular sorbitol accumulation and its incapability to pass through the cell membrane in insulin-independent tissues, thus contributing to cell damage. The intermediates of this pathway can glycate proteins leading to AGEs formation, aggravating the vascular damage [Citation260]. Moreover, another source of oxidative stress is the formation of the pro-oxidant glyoxal, a precursor of AGEs, which can result from the buildup of glucose in cells [Citation261].

Protein Kinase C is an enzyme playing a crucial role in a wide range of cellular processes and cell signaling pathways. It functions by modifying the activities of other proteins through a cascade of phosphorylation reactions. PKC activation is closely associated with oxidative stress in DM2, creating a vicious cycle. Activation of PKC in DM2 has been linked to several mechanisms that contribute to diabetic complications such as insulin resistance due to impairment of insulin signaling, leading to insulin resistance [Citation262]. In addition, this pathway interferes with the normal translocation of glucose transporter type 4 (GLUT4) to the cell surface, reducing glucose uptake by cells. Moreover, it produces vascular complications, fibrosis and oxidative stress [Citation263]

Heightened activation of the PKC pathway has been shown to stimulate enzymes that generate ROS, such as NADPH-oxidases and lipoxygenases [Citation262]. In addition, PARP activation due to DNA oxidation affects the signal and induces oxidative stress through PKC activation [Citation264]. Inflammation, aging, and oxidative stress are closely intertwined processes that contribute to the pathogenesis of DM2 [Citation265]. The balance between proinflammatory and anti-inflammatory cytokines is essential for the development of muscle insulin resistance and in DM2. Chronic low-grade inflammation is commonly observed, being characterized by increased levels of pro-inflammatory cytokines and activation of immune cells. This inflammatory state is mediated by various factors, including adipose tissue dysfunction, insulin resistance, and metabolic abnormalities [Citation266]. The pro-inflammatory cytokines, such as TNF-α, IL-6, and IL-1β, released during inflammation can further impair insulin signaling and promote insulin resistance. IL-6 has been shown to induce hyperglycemia and compensatory hyperinsulinemia [Citation267,Citation268]. TNF-α, a cytokine with proinflammatory properties, affects lipid metabolism and contributes to insulin resistance by influencing the activity of GLUT4 and inducing phosphorylation of insulin receptor substrate-1 [Citation269,Citation270]. Lipotoxicity, besides causing insulin resistance, also plays a role in pancreatic β cell dysfunction. The activation of ROS and endoplasmic reticulum stress have been identified as the mechanisms that contribute to lipotoxicity in β cells [Citation271].

In the development of this pathology, it is important not to overlook prediabetes, which involves the infiltration of immune cells into adipose tissue and holds a significant role. Macrophages with an M1-like phenotype are known for their proinflammatory characteristics leading to a failure of pancreatic β-cells, while macrophages exhibiting an M2-like phenotype secreting cytokines with anti-inflammatory properties [Citation272]

The treatment of patients diagnosed with DM2 encompasses various aspects. These include providing education to patients, evaluating the presence of micro- and macrovascular complications, aiming for optimal blood glucose control, reducing cardiovascular and other long-term risk factors, and avoiding medications that may worsen insulin or lipid metabolism abnormalities. Managing inflammation and oxidative stress are essential in the management of DM2 that comprise lifestyle interventions, including regular exercise, a healthy diet, and weight management. Antioxidant-rich foods and/or supplementation with antioxidants may also be beneficial. Additionally, medications targeting inflammation and oxidative stress pathways may be prescribed to alleviate these processes and improve metabolic control in DM2.

The first line of pharmacological treatment is metformin, an oral antidiabetic biguanide agent which has greater anti-inflammatory activity. It should be avoided insulin secretagogues because they are dangerous for frail older adults due the increased risk of hypoglycemia and falls [Citation273]. Moreover, serious hypoglycemia and the concomitant use of warfarin with sulfonylureas or metformin should be taken in consideration for the treatment of older adults [Citation274]

Metformin activates AMP-activated protein kinase (AMPK) pathway, that reduces excessive ROS generation and limits cell apoptosis. Also, it increases signaling through the ROS-mediated PI3K/Akt, AKT/Nrf2 and Nrf2/HO-1 pathways, and markedly elevates the levels of its downstream antioxidants [Citation275,Citation276]. Moreover, metformin may be able to protect cells against H2O2-induced ROS by autophagy activation [Citation277]. Some studies indicate that metformin may induce anti-aging transcriptional changes and beneficial effects of metformin on aging and healthspan are primarily indirect via its effects on cellular metabolism and result from its anti-hyperglycemic action, enhancing insulin sensitivity, reduction of oxidative stress and protective effects on the endothelium and vascular function [Citation278,Citation279], although these data remain controversial. Apart from its hypoglycemic effects, it has been observed to offer further advantages in terms of antioxidant function, weight loss, and the management of several diseases. These include cancer, cardiovascular disorders, and metabolic conditions, encompassing thyroid diseases as well [Citation280].

4.8. Depression

Depression is a severe mental disorder that significantly raises the risk of illness and death, particularly in the later stages of life. Additionally, it diminishes the overall quality of life for both the individuals battling depression and their families. The prevalence of geriatric depression represents a significant challenge to public health, particularly when it occurs alongside chronic medical conditions, often escaping proper diagnosis. It frequently coexists with other health issues, such as DM2 and CVD, synergistically affecting the well-being and disease management of older people [Citation281,Citation282].

The treatment will depend upon the severity, type, and chronicity of the disease. Psychotherapy and pharmacotherapy may be used singly or in combination. For moderate to severe forms of depression, it is better to use pharmacotherapy. However, there are too many drug–drug interactions and greater risk for adverse events.

When dealing with depression in older adults, it is crucial to conduct a thorough review of all the medications the patient is currently taking. This is essential as the medications prescribed for physical ailments can trigger or exacerbate depressive symptoms or the condition itself [Citation283]. Studies conducted have shown significant heterogeneity in the treatment outcomes. However, it was demonstrated that they were effective for late-life major depressive disorder but with less effective results than in younger patients [Citation284].

The primary categories of antidepressant medications include selective serotonin reuptake inhibitors (SSRIs), serotonin–norepinephrine reuptake inhibitors, tricyclic antidepressants, and monoamine oxidase inhibitors. Among these, SSRIs are the most safe and commonly prescribed class of antidepressants, making them the first-line treatment for older patients due to their limited effects on autonomic system activity and a lower impact on blood pressure [Citation285]. Nevertheless, SSRIs may be associated with falls and osteoporosis [Citation286].

Within SSRIs, the most well-known ones are sertraline, paroxetine, and escitalopram. It's worth noting that paroxetine is linked to cholinergic muscarinic antagonism and robust inhibition of CYP2D6, while sertraline exhibits moderate concerns regarding drug interactions compared to escitalopram. When it comes to choosing an antidepressant for oncological patients, it's a delicate balance between assessing the side effect profile and pharmacokinetics. Drug interactions between antidepressants and antineoplastic drugs (ANs) can potentially compromise the effectiveness of ANs and elevate their toxicity, which has evident implications for prognosis. For most other ANs that rely on CYP 450 3A4 metabolism, caution is advised when administering them concurrently with inhibitors of this isoenzyme, such as sertraline, paroxetine, and fluvoxamine [Citation287].

In laboratory studies, a significant modulation of intracellular ROS levels and GSH levels, as well as changes in antioxidant enzymatic activities and transcriptional profiles of antioxidant genes, were observed in response to sertraline and fluoxetine. These changes led to a delay in cell growth and the induction of oxidative stress [Citation288]. Similarly, other studies have indicated that both fluoxetine and citalopram reduced SOD and MDA levels while increasing ascorbic acid levels after a 12-week treatment in a group of major depressive disorder patients [Citation289]

4.9. Chronic kidney disease

Chronic kidney disease (CKD) is a pathological condition characterized by a decrease in the glomerular filtration rate, closely related to the pathologies previously described, with diabetes and hypertension being the main causes of this condition. Currently, it is one of the main causes of death and morbidity worldwide, where its more advanced stages have a higher prevalence in the elderly [Citation290]. As a consequence of the advanced stages, patients must undergo renal replacement therapies, where hemodialysis corresponds to the main method [Citation291]. However, these patients have a lower life expectancy than people of the same age and sex who are not undergoing hemodialysis (4.3 and 5.3 years less). Furthermore, the median survival on dialysis of elderly patients has been reported to be 1.6–5.4 years shorter than those of patients younger than 45 years [Citation291,Citation292].

Oxidative stress plays a fundamental role in the development and progression of CKD, as it is related to the etiopathogenesis of its causes, as well as by being directly involved in glomerular damage, interstitial fibrosis, inflammation and microvascular dysfunction present in kidney damage [Citation293]. The main sources of ROS in the kidney correspond to the mitochondria and the NOX enzyme [Citation294]. The chronic increase in ROS in the tubular epithelium exposes the tissue to direct cellular damage, but also induces elevations of proinflammatory cytokines such as TGF-B, which promotes the synthesis of collagen fibronectin, and plasminogen activator inhibitor-1, while attenuating activity of extracellular matrix degradation factors in mesangium and endothelial cells [Citation293].

As a result of the above, various biomarkers of oxidative stress have been studied in subjects with CKD. First, in relation to lipid peroxidation, elevated plasma levels of MDA, 8-isoprostanes [Citation295] and thiobarbituric acid reactive substances (TBARS) have been reported in patients with CKD. Also, high levels of 8-hydroxy-20-deoxyguanosine are found as a biomarker of oxidative damage in the leukocyte DNA of patients with CKD [Citation296]. Likewise, urine 8-hydroxy-20-deoxyguanosine and serum MDA levels were significantly higher in diabetic patients with proteinuria [Citation297]. Also, increased levels of protein oxidation, such as carbonylation, nitrotyrosine, advanced oxidation protein products and nonenzymatic advanced glycation end products [Citation298,Citation299].

The management of CKD is based mainly on strict control of blood pressure and glycemia in all patients, including older adults. For this, drugs such as ACE inhibitors or ARBs are the most recommended for hypertension and proteinuria. While the effect on oxidative stress was described in the hypertension section, the antioxidant/oxidant effect has also been reported in CKD [Citation300].

On the other hand, for the management of diabetes in patients with CKD, the use of metformin and SGLT2 inhibitors (SGLT2i) is recommended as first-line hypoglycemic agents. Canaglifozin has been shown to decrease ROS production and cellular damage in cultures of mouse proximal tubular cells by suppressing the increase in renal proximal tubular angiotensinogen mediated by hyperglycemia [Citation301]. Furthermore, it has been reported that the administration of dapagliflozin in cultures of Human proximal tubular cells decreases cytosolic and mitochondrial ROS and prevents cellular damage associated with oxidative stress [Citation302]. Likewise, it has been reported that canagliflozin prevents the increase in MDA, advanced protein oxidation product and nitric oxide mediated by the enzymes iNOS and NOX isoform 4 and in turn increases the activity of SOD and CAT and the levels of reduced glutathione in rats with increased oxidative stress induced by isoprenaline [Citation303]. Empagliflozin has also reported the decrease of superoxide and hydrogen peroxide induced by hyperglycemia in cultures of human proximal tubular epithelial cells, and in turn prevents the activation of inflammatory signaling pathways [Citation304]. On the other hand, recent studies reported that empagliflozin would have antioxidant and anti-renal senescence effects mediated by SIRT1 in a d-galactose-induced aging model in rats [Citation305] and also managed to restore endothelial dysfunction induced by hyperglycemia through the modulation of oxidative stress in human cell cultures [Citation306].

5. Antioxidants treatments for aging related diseases

One way to reduce polypharmacy-related adverse effects may be to incorporate or replace conventional therapies with therapies based on natural antioxidant products, such as ascorbic acid, quercetin, resveratrol or curcumin. These agents act through mechanisms of action different from conventional drugs, have a safe adverse events profile and beneficial effects in various conditions that have oxidative stress as a common pathophysiological pillar, reporting evidence in hypertension, DM2, dyslipidemia, polyneuropathy, cognitive decline, among other pathologies and aging-related conditions with a high prevalence in the elderly. Therefore, its use could allow achieving therapeutic goals at lower doses of standard therapy by enhancing its individual effects with those of antioxidants, avoiding the addition of new drugs to the base therapy, which increases the risk of adverse events associated with polypharmacy or the use of high doses of each drug.

5.1. Vitamin C

One of the most studied antioxidants in various clinical models corresponds to vitamin C, which is a water-soluble molecule that acts as a direct antioxidant as it is a powerful radical scavenger of ROS. In addition, it has other indirect effects on the redox state by recycling vitamin E, reducing NF-κB levels or preventing BH4 oxidation [Citation307]. Furthermore, clinical studies in older adults have reported that oral consumption of vitamin C achieves an increase in antioxidant defenses and a decrease in markers of lipid peroxidation in patients with DM2 [Citation308,Citation309]

Several clinical studies have been carried out to demonstrate its antihypertensive effect, based on the report of an inverse relationship between plasmatic vitamin C levels and blood pressure levels, which could be explained mechanistically by the activity of up- regulation of eNOS and down-regulation of NOX [Citation310]. Furthermore, this is consistent with the improvement in endothelial dysfunction reported in patients with various pathologies [Citation307,Citation311]. Our team conducted a randomized placebo-controlled clinical trial, where it was reported a significant decrease in systolic, diastolic, and mean blood pressure in patients with untreated essential hypertension after daily consumption of 1 g vitamin C and 400 IU of vitamin E orally for 8 weeks, associated with an improvement in oxidative stress, specifically a decrease in MDA and F2-isoprostane levels and an increase in ferric reducing antioxidant power (FRAP) and the GSH/GSSG ratio [Citation312]. However, the subjects enrolled in this study had no other comorbidities and were between 35 and 60 years of age, with a mean of 46 years. On the other hand, another randomized placebo-controlled study reported that after consuming 1 g of vitamin C orally for 6 weeks, the blood pressure response to low-intensity exercise would improve in patients with DM2, obtaining lower systolic and diastolic pressures immediately before and after exercising, added to a decrease in the levels of MDA and F2-isoprostanes in patients supplemented with vitamin C [Citation311]. Likewise, another study reported that the use of oral vitamin C decreases systolic and diastolic blood pressure in patients with hypertension with a mean age of 69 years [Citation313]. Furthermore, a recent systematic review and meta-analysis concluded that vitamin C causes a significant reduction in blood pressure in patients with essential hypertension, even in those subgroups of patients older than 60 years [Citation314]. Furthermore, the use of vitamin C in patients with refractory hypertension has also shown benefits in the elderly patient group, significantly reducing systolic blood pressure after a 6-month administration of 600 mg/day compared to a group of non-elderly patients with refractory hypertension who were also given the same dose of vitamin C [Citation315] ().

Table 1. Main antioxidant drugs used in age-related diseases and their therapeutic effects in clinical trials that evaluated oxidative stress parameters.

Vitamin C has also presented an improvement in metabolic parameters [Citation309,Citation316–318], where a clinical trial reported that the use of vitamin C for 8 weeks managed to reduce the levels of fast blood glucose and inflammatory parameters, the IL6 and high-sensitivity C-reactive protein (hs-CRP), in a group of patients with hypertension and/or obesity and a mean age of 50.69 years [Citation319]. In addition, Mason et al. reported that supplementation of 1 g/day of vitamin c for 4 months improves postprandial glycaemic control in patients with DM2 and a mean age of 61.8 years, also showing a decrease in F2-isoprostanes and systolic and diastolic pressure by −7 mmHg and −5 mmHg, respectively[Citation318]. Moreover, the use of vitamin C and/or E in men who daily consume metformin has been found to improve insulin resistance and fasting blood glucose levels, with an increase in GSH [Citation317].

However, despite this evidence, the recommendation to use vitamin C as antihypertensive therapy remains inconclusive, so further clinical studies must be performed to elucidate it. In addition, there is a need to carry out clinical studies that evaluate the effects of associating vitamin C with conventional hypertension therapy.

5.2. Polyphenols

Polyphenols are a wide variety of compounds with different subclasses, such as flavonols (as quercetin), flavanones, flavones, isoflavones, anthocyanins, stilbenes (as resveratrol), and curcuminoids (as curcumin), among others [Citation320]. Within the scientific community, it has been suggested that these natural compounds may provide benefits in various areas of health, including effects on the cardiovascular, musculoskeletal, neurological, metabolic systems, as well as in the prevention and treatment of non-communicable diseases and cancer.

A systematic review and meta-analysis reported that quercetin reduces systolic and diastolic BP after analyzing 7 randomized controlled trials (RCTs), suggesting that the effect would be greater in those who received doses higher than 500 mg/day. However, only 1 of the articles included patients with arterial hypertension, another included patients with prehypertension, and all others included subjects without hypertension [Citation321]. Similarly, in another RCT with doses lower than 500 mg/day, overweight or obese patients with prehypertension and stage I hypertension experienced a slight reduction in blood pressure at 24 h with a dose of 162 mg/day compared to the placebo group. However, this dose did not have a significant effect on blood pressure in the overall group [Citation322]. The same effect was observed in an RCT in a population of 93 overweight or obese subjects where a dose of 150 mg/day of quercetin was administered for 6 weeks which led to a decrease in blood pressure in the hypertensive subgroup, in addition to significantly decreasing the plasma concentration of atherogenic oxidized LDL, which positions quercetin as a promising natural antioxidant with cardioprotective properties [Citation323]. Another polyphenol that has shown diverse results in reducing cardiovascular risk is resveratrol. In a review where several RCTs were analyzed, it was found that this antioxidant significantly reduced body weight in obese patients and partially lowered systolic blood pressure, in addition to improving parameters such as fasting glycemia and glycosylated hemoglobin in diabetic patients. However, the range of doses used in the different RCTs varies between 5 and 5000 mg/day and over wide periods (from a couple of days to months) [Citation324]. Importantly, although there is evidence that some natural antioxidants, such as those obtained by consuming berries, do not appear to reduce blood pressure when used as monotherapy [Citation325], evidence suggests that polyphenols may have cardioprotective effects when consumed in a diet rich in them, thanks to the pleiotropic effects generated by the different subclasses of polyphenols [Citation326].

In addition to the cardioprotective effects of antioxidants mentioned above, it is important to highlight the role that these compounds can play in improving the cardiovascular risk profile, being beneficial in the prevention and progression of pathologies such as DM2. In a systematic review focused on the analysis of RCTs it could be seen that curcumin can reduce blood glucose and triglyceride levels, as well as other liver function markers such as the enzyme alanine aminotransferase and aspartate aminotransferase in patients with DM2, concluding that curcumin is beneficial in improving metabolic complications, but that future research is still needed to clearly determine other aspects related to its absorption and bioavailability [Citation327]. In relation to the above, studies conducted with other antioxidants such as resveratrol, have shown beneficial effects on the control of glycosylated hemoglobin (Hb1Ac) and glucose control [Citation328]. Supporting the previous hypothesis, a recent RCT conducted in diabetic patients receiving a daily dose of 200 mg of resveratrol showed that, after 24 weeks of supplementation, a significant reduction in plasma glucose and an increase in the homeostatic insulin resistance model were observed, in addition to a decrease in inflammatory parameters such as hs-CRP and TNF-α. These effects were observed without the presence of side effects, suggesting the possibility that resveratrol supplementation, in combination with oral hypoglycemic agents, may be useful in diabetes therapies [Citation329] ().

Regarding musculoskeletal health, one of the polyphenols with relative evidence is resveratrol. OA is one of the most common disorders in this area worldwide, in which inflammation plays a major role. In an RCT where 40 g of lyophilized blueberry powder were assigned for 4 months to 33 patients with OA, it was observed that there was a decrease in pain, stiffness, and difficulty in performing daily activities at the end of the study, which was observed despite not observing a decrease in inflammatory parameters such as TNF-α, interleukins, or matrix metalloproteinases [Citation330]. Similarly, in an RCT where resveratrol was used as an adjuvant therapy with meloxicam for patients with OA, this antioxidant significantly improved pain, functional capacity, and associated symptoms compared to placebo, while demonstrating a good safety profile and being well tolerated by patients [Citation331]. It is worth noting that the doses used were more than double those commonly used in the RCTs mentioned in this review (500 mg vs 200 mg per day), which, combined with the therapy's good tolerability, suggests that further studies with higher doses of this antioxidant may yield better results.

Another of the health benefits for which polyphenols stand out are their anticarcinogenic effects. In a review where the effect of various flavonoids on different types of cancer was extensively analyzed, the beneficial effects of these antioxidants on lung, breast, gastric, colorectal, liver, cervical and prostate cancer, among others, could be seen by regulating the inflammatory response and decreasing oxidative stress. One of the flavonoids that stands out in different types of cancer is quercetin, which acts through mechanisms such as decreasing the inflammatory response associated with NF-κB signaling, possibly inducing apoptosis through phosphorylation of AMPK and p53 and blocking pathways such as PI3K and WNT signaling pathway. However, this review concludes that there is still a lack of clinical trials of flavonoids on the use of flavonoids to treat cancer [Citation194]. In the same line and supporting what was mentioned by the previous review, in an RCT of 26 patients with breast cancer who consumed 3 capsules/day (296.4 phenolic compounds per capsule, including curcumin and resveratrol) the presence of metabolites derived from curcuminoids, isoflavones and lignans in malignant breast tissues was analyzed for the first time, which could exert anticancer effects after long-term exposure [Citation332] according to the mechanisms mentioned above.

In addition to the benefits mentioned above, it is important to highlight the potential role that polyphenols may have in benefiting cognitive functions or in the prevention and treatment of neurodegenerative diseases. Although there are clinical trials where it was shown that the consumption of 200 mg of resveratrol on the memory of older adult participants aged 60–79 years for 26 weeks did not achieve significant improvements in aspects such as verbal memory after 6 months [Citation333], there is evidence that the use of higher doses of this antioxidant (up to 1 g orally twice a day) for longer periods (52 weeks) may reduce neuroinflammation and induce adaptive immunity in AD [Citation334]. Furthermore, RCTs have demonstrated that the consumption of curcumin can lead to an elevation in plasma Vitamin E levels and an increase in serum levels of Aβ40. This effect potentially indicates curcumin's capability to disintegrate deposits of this biomarker in the brains of individuals with AD [Citation335]. These findings suggest a potential beneficial role of these antioxidants in therapeutic interventions for this condition. However, additional clinical trials with varying concentrations of these compounds and extended durations are necessary to comprehensively assess the long-term neuroprotective effects.

5.3. N-acetylcysteine

N-acetylcysteine (NAC) is an antioxidant that acts as a radical scavenger and precursor of GSH, which is currently widely used to treat patients with acetaminophen poisoning or as a mucolytic. In a recent RCT, it was reported that supplementation with glycine and NAC in older adults between 61 and and 80 years of age for 16 weeks increases GSH levels in muscle and red blood cells, decreased the levels of TBARS and F2-isoprostanes significantly, until presenting no differences with the levels of young adults with an average age of 25.6 years, who presented a baseline difference of 424% of TBARS and 301% of F2-iso between both groups, decreased systolic blood pressure and improved insulin resistance, endothelial dysfunction and other parameters associated with aging [Citation336].

One of the pathologies where research on the benefits of NAC use has been expanded is rheumatoid arthritis, where it has been reported that the oral administration of 600 mg of NAC twice a day for 3 months in patients with rheumatoid arthritis manages to improve certain parameters of oxidative stress, such as MDA and NO [Citation337] when compared with the placebo group, however, other parameters do not present an improvement, such as the total antioxidant capacity measured in plasma () [Citation337,Citation338] and GPX activity [Citation338] do not present significant differences. On the other hand, studies that have evaluated clinical parameters present contradictory results, where Jamali et al. reported a significant reduction of disease activity assessed by DAS28, but Esalatmanesh et al. and Batooei et al. reported that there were no significant differences in this parameter. Morning stiffness decreased significantly with respect to the baseline of the patients, but not when compared with the placebo group [Citation338] (). Finally, pain severity assessed by visual analog scale, physical performance health assessed by Health Assessment Questionnaire and global health as a parameter of the disease activity evaluated by the same patient presented significant differences with respect to the placebo group [Citation339]. Therefore, the use of NAC in rheumatoid arthritis is debatable, and more studies are needed to demonstrate the true usefulness of this drug.

Also, the use of NAC as an adjuvant treatment to improve cognition in patients with AD has been evaluated. Adair et al. conducted a RCT orally administered 50 mg/kg/day for six months to patients with probable AD, reporting that there were no differences between the control group and placebo in the measurements of oxidative stress parameters (SOD and GPX activity and levels of GSH and TBARS) in peripheral blood, as well as their primary measurements (Mini-Mental State Examination and Activities of Daily Living Scale) did not show significant differences either. However, other parameters such as the Wechsler Memory Scale figure reproduction and the letter fluency task showed significant improvement at 3 and 6 months, respectively, in patients receiving NAC [Citation340]. Other studies have included NAC supplementation in patients with AD but within a multivitamin complex together with other antioxidants [Citation341–343], so the results cannot be directly associated with the use of NAC, which is widely described in the systematic review conducted by Skvarc et al. [Citation344].

5.4. Alpha-Lipoic acid

Alpha-Lipoic acid (ALA) is an endogenous antioxidant derived from octanoic acid, which is found in various foods of animal or vegetable origin. It has a high antioxidant capacity, acting in multiple ways to reduce oxidative stress, among which it has been reported that it acts as a radical scavenger and ionic metal chelator, promotes the synthesis of GSH and recycles other antioxidants such as vitamin C, vitamin E, and coenzyme Q10 (CoQ10) [Citation345]. In addition, this antioxidant has an amphipathic nature, which allows it to act both at the extracellular and intracellular levels. For this reason, in the last years, interest in testing its potential benefits in different pathologies has been increasing, leading to clinical trials in patients with diabetic polyneuropathy, metabolic diseases, cancer, among others [Citation346].

Diabetes mellitus has been one of the pathologies in which various clinical studies have been conducted to test the enhancement of antioxidant defenses through ALA, given the significant role of oxidative stress underlying the disease and its complications described in the previous section [Citation347]. A clinical study reported that the administration of 600 mg/day of ALA in combination with 165 mg of L-carnosine, 7.5 mg of zinc, and B-group vitamins in a dietary supplement for 3 months improved oxidative stress parameters by increasing the activity of SOD and GPX while reducing the levels of MDA [Citation348]. However, another study in which 600 mg/day of ALA was administered orally without other compounds for 6 months in patients with DM2 did not show differences in SOD and GPX activity or levels of 8-isoprostanes when compared to the placebo group [Citation349]. Also, in a study where a daily dose of 400 mg of ALA was administered, a decrease in levels of reactive oxygen metabolites measured by d-ROMs was reported, but no changes were observed in the levels of biological antioxidant potential [Citation350]. On the other hand, the administration of 600 mg/day of ALA intravenously for 3 weeks in patients with DM2 complicated by acute cerebral infarction, achieving an increase in SOD and GPX activity, a decrease in plasma MDA levels, and a reduction in the NIHSS score [Citation351]. Regarding metabolic and inflammatory parameters, blood lipids tend to improve after ALA administration [Citation348–351], along with a reduction in markers of inflammation [Citation348,Citation349,Citation351], blood glucose [Citation351,Citation352], and HbA1c levels [Citation352] in patients with DM2 ().

Other studies have been conducted to prevent cardiovascular damage in hemodialysis patients or those with CKD through supplementation with ALA. Khabbazi et al. administered 600 mg/day of ALA orally to patients with end-stage CKD, reporting a decrease in hs-CRP levels compared to the placebo group and an increase in HDL cholesterol levels compared to baseline levels. However, there were no significant differences in MDA levels, total antioxidant status, or other lipid profile parameters between the groups [Citation353]. These results are consistent with those reported by other two clinical trial who administered a combination of 600 mg/day of ALA with 666 IU/day of mixed tocopherols for 2 or 6 months, except for the inflammatory parameters, where no significant differences were found [Citation354,Citation355]. Other clinical studies in hemodialysis patients have been conducted, where the administration of 600 mg/day of oral ALA for 8 weeks demonstrated an increase in SOD activity, but no significant differences were observed in mean levels of albumin, hemoglobin, weight, or BMI [Citation356]. In contrast, the administration of the same dose of ALA for 6 months reported a decrease in levels of 8-OHdG, hs-CRP, TNF-α, and metabolic parameters [Citation357] ().

5.5. Coenzyme Q10

CoQ10 is a lipophilic molecule synthesized within the mitochondria of eukaryotic cells. It plays a crucial role as an electron carrier in the mitochondrial respiratory chain [Citation358]. The prospective use of CoQ10 supplementation in addressing tissue fibrosis, a factor associated with the age-related decline in the functionality of several organs, particularly the heart, is noteworthy. Clinical investigations have shown that CoQ10 supplementation can potentially reduce the extent of cardiovascular fibrosis experienced by older individuals, consequently enhancing cardiovascular performance [Citation359,Citation360]. Several clinical studies have evaluated the use of CoQ10 in metabolic or CVD, however, only a few have reported the effects on oxidative stress. In patients with coronary artery disease, the administration of 150 mg/day of CoQ10 for 12 weeks resulted in decreased levels of MDA and increased activity of the antioxidant enzymes CAT and SOD [Citation361]. Furthermore, the use of CoQ10 in hemodialysis patients decreased isofuran levels in one study, but not F2-isoprostane levels [Citation362], contrary to what was reported in another study where a decrease in F2-isoprostane levels was observed, but not in isofurans, cardiac biomarkers, and BP [Citation363] ().

ACEI, angiotensin-converting enzyme inhibitors; ACI, acute cerebral infarction; BMI, body mass index; BUN, blood urea nitrogen; CAT, catalase; CKD, chronic kidney disease; DM2, diabetes mellitus type 2; d-ROMs, reactive oxygen metabolites test; FBG, fasting blood glucose; GPX, glutathione peroxidase; HDL, high-density lipoprotein cholesterol; hs-CRP, highly sensitive C-reactive protein; IGT, impaired glucose tolerance; MDA, malondialdehyde; SBP, systolic blood pressure; SOD, superoxide dismutase; Tsat, transferrin saturation; 8-OHdG, 8-hydroxy-2’-deoxyguanosine; ΔRd,delta rate of glucose disappearance.

6. Reductive stress

Reductive stress is a concept that was first mentioned by Albert Wendel, a German biochemist known for his work on the role of glutathione in cellular redox reactions. It is essentially the opposite of oxidative stress and occurs when there is an alteration in redox control and signaling.

As well as oxidative stress, it can have adverse effects on cells and tissues and can affect various signaling pathways, disrupt the balance of ROS, and interfere with normal cellular functions. Excessive levels of reducing agents in cells can lead to reduce mitochondrial function, decrease cellular metabolism, activation of NF-κB pathway and a reduction in the formation of disulfide bonds, which are important for maintaining the structure and function of proteins. This process is particularly relevant in the context of antioxidant therapy, where excessive intake of them can potentially shift the cellular redox balance toward reductive stress, leading to unintended consequences on health and disease [Citation371].

7. Conclusion

In conclusion, aging implies a decrease in biological and metabolic activity, whose main pathophysiological mechanism is the alteration of redox signaling as a result of a decrease in our body's antioxidant defense. This leads to the appearance of various age-related diseases, such as neurodegenerative diseases, CVD, metabolic diseases, cancer, among others. These pathologies demand the use of medicines as the first line of treatment, which causes many older adults to fall into polypharmacy, has hidden costs and dangers, as well as negative effects, and is directly related to the development of frailty, which may compromise even more the body's antioxidant defense systems. This highlights the importance of careful medication management and regular assessment of drug regimens in older adults as well as the potential complementary toxic therapies of cancer for minimizing the adverse effects on oxidative balance.

In this sense, the understanding of the physiopathology of different age-related diseases has led to the development of different therapies based on healthy lifestyles and the consumption of natural antioxidants such as flavonoids, carotenoids, vitamin C, vitamin E, among others, as an option or complement to conventional pharmacological therapies, with the aim of reducing their adverse effects and contributing to the well-being of the older adult population. However, despite having evidence both in support and against the use of certain antioxidants for certain diseases, these therapies still lack the necessary evidence for widespread application in the population. Furthermore, the complexities associated with polypharmacy demand careful consideration when addressing oxidative stress in older adults. In this regard, healthcare professionals must pay special attention to potential drug-induced oxidative stress and pharmacological interactions to ensure optimal antioxidant support and minimize the negative effects of polypharmacy in the aging process. It is imperative to conduct further research to unravel the precise mechanisms underlying the interaction between oxidative stress, antioxidants, and polypharmacy. This will enable the development of personalized strategies aimed at maintaining optimal antioxidant balance in the aging population.

Author contributions

C.R.-S.: Analysis of published articles about oxidative stress-related diseases; V.P-G.: Studies on the state of the art in aging and its relationship with polypharmacy; J.L.-M.: Studies about antioxidants and drugs as a therapy in different diseases; T.G.-F.: Clinical effects of antioxidants in ageing; L.S.: Scientific study rationale; R.R.: Hypothesis design, fund raising. All authors have read and agreed to the published version of the manuscript.

Abbreviations
Aβ,=

amyloid-β;

ACE,=

angiotensin-converting enzyme;

AD,=

Alzheimer’s disease;

AGEs,=

advanced glycation end products;

AKI,=

acute kidney injury;

ALA,=

alpha-Lipoic acid;

AMI,=

acute myocardial infarction;

AMPK,=

AMP-activated protein kinase;

ANs,=

antineoplastic

ARE,=

antioxidant response elements;

ARBs,=

angiotensin-2 receptor blockers;

BMI,=

body mass index;

CAT,=

catalase;

CKD,=

chronic kidney disease;

CVD,=

cardiovascular diseases;

CoQ10,=

coenzyme Q10;

DM2,=

type 2 diabetes mellitus;

eNOS,=

endothelial nitric oxide synthase;

FRAP,=

ferric reducing antioxidant power;

GFR,=

glomerular filtration rate;

GLP-1,=

Glucagon-like peptide-1;

GLUT4,=

glucose transporter type 4

GPX,=

glutathione peroxidase;

GSH,=

gluthatione;

Hb1Ac,=

glycated hemoglobin;

HDL,=

high-density lipoprotein;

hs-CRP,=

high-sensitivity C-reactive protein;

HOMA-IR,=

Homeostatic Model Assessment of Insulin Resistance;

IL-6,=

interleukin-6;

IRI,=

ischemia-reperfusion injury;

LDL,=

low-density lipoprotein;

MCP-1,=

Monocyte Chemoattractant Protein 1;

MDA,=

malondialdehyde;

mPTP,=

mitochondrial permeability transition pore;

mtDNA,=

mitochondrial DNA;

NAC,=

N-acetylcysteine;

NF-κB,=

Nuclear factor kappa-light-chain-enhancer of activated B cells;

NMDA,=

N-methyl-D-aspartate;

NSAIDs,=

nonsteroidal anti-inflammatory drugs;

NO,=

nitric oxide;

NOS,=

nitric oxide synthase;

NOX,=

NADPH oxidase;

Nrf2,=

nuclear factor erythroid 2;

O2·_,=

superoxide anion radical;

ONOO−,=

peroxynitrite;

PARP,=

poly-ADP-ribose polymerase;

PCI,=

percutaneous coronary intervention;

PKC,=

protein kinase C;

PUFAs,=

polyunsaturated fatty acids;

RAAS,=

renin-angiotensin-aldosterone system;

RCTs,=

randomized controlled trials;

ROS,=

reactive oxygen species;

RNS,=

reactive nitrogen species;

SOD,=

superoxide dismutase;

SSRIs,=

selective serotonin reuptake inhibitors;

TBARS,=

thiobarbituric acid reactive substances;

OA,=

osteoarthritis;

8-OHdG,=

8-hydroxydeoxyguanosine;

Disclosure statement

No potential conflict of interest was reported by the author(s).

Data availability statement

Data sharing is not applicable to this article as no new data were created or analyzed in this study.

Additional information

Funding

This manuscript was funded by N◦ 1211850 of the Agencia Nacional de Investigación y Desarrollo (ANID). This institution did not participate in the design, literature review, or writing of the manuscript.

References

  • United Nations. World social report 2023: leaving no one behind in an ageing world | UN DESA Publications [Internet]. 2023. Available from: https://desapublications.un.org/publications/world-social-report-2023-leaving-no-one-behind-ageing-world.
  • Chowdhury SR, Chandra Das D, Sunna TC, et al. Global and regional prevalence of multimorbidity in the adult population in community settings: a systematic review and meta-analysis. eClinicalMedicine. 2023;57:101860–101860. doi:10.1016/j.eclinm.2023.101860
  • Makovski TT, Schmitz S, Zeegers MP, et al. Multimorbidity and quality of life: systematic literature review and meta-analysis. Ageing Res Rev. 2019;53:100903–100903. doi:10.1016/j.arr.2019.04.005
  • López-Otín C, Blasco MA, Partridge L, et al. The hallmarks of aging. Cell. 2013;153:1194–1217. doi:10.1016/j.cell.2013.05.039
  • Delara M, Murray L, Jafari B, et al. Prevalence and factors associated with polypharmacy: a systematic review and meta-analysis. BMC Geriatr. 2022;22:601. doi:10.1186/s12877-022-03279-x
  • Midão L, Brochado P, Almada M, et al. Frailty status and polypharmacy predict all-cause mortality in community dwelling older adults in Europe. Int J Environ Res Public Health. 2021;18:3580. doi:10.3390/ijerph18073580
  • Doumat G, Daher D, Itani M, et al. The effect of polypharmacy on healthcare services utilization in older adults with comorbidities: a retrospective cohort study. BMC Prim Care. 2023;24:120. doi:10.1186/s12875-023-02070-0
  • Sinnott C, Mc Hugh S, Browne J, et al. GPs’ perspectives on the management of patients with multimorbidity: systematic review and synthesis of qualitative research. BMJ Open. 2013;3:e003610. doi:10.1136/bmjopen-2013-003610
  • O’Mahony D, Rochon PA. Prescribing cascades: we see only what we look for: we look for only what we know. Age Ageing. 2022;51:afac138. doi:10.1093/ageing/afac138
  • Kennedy BK, Berger SL, Brunet A, et al. Geroscience: linking aging to chronic disease. Cell. 2014;159:709–713. doi:10.1016/j.cell.2014.10.039
  • Van Der Graaf PH, Gabrielsson J. Pharmacokinetic–pharmacodynamic reasoning in drug discovery and early development. Future Med Chem. 2009;1:1371–1374. doi:10.4155/fmc.09.124
  • O’Connor A, O’Moráin C. Digestive function of the stomach. Dig Dis Basel Switz. 2014;32:186–191. doi:10.1159/000357848
  • Nandagopalan PA, Magdalene KF, Binu A. Effect of aging on the quantitative number of Brunner’s glands. J Clin Diagn Res JCDR. 2014;8:4–4.
  • Zhou R, Moench P, Heran C, et al. pH-Dependent dissolution in vitro and absorption in vivo of weakly basic drugs: development of a canine model. Pharm Res. 2005;22:188–192. doi:10.1007/s11095-004-1185-3
  • Pang J, Dalziel G, Dean B, et al. Pharmacokinetics and absorption of the anticancer agents dasatinib and GDC-0941 under various gastric conditions in dogs - reversing the effect of elevated gastric pH with betaine HCl. Mol Pharm. 2013;10:4024–4031. doi:10.1021/mp400356m
  • Phillips RJ, Kieffer EJ, Powley TL. Aging of the myenteric plexus: neuronal loss is specific to cholinergic neurons. Auton Neurosci Basic Clin. 2003;106:69–83. doi:10.1016/S1566-0702(03)00072-9
  • Bernard CE, Gibbons SJ, Gomez-Pinilla PJ, et al. Effect of age on the enteric nervous system of the human colon. Neurogastroenterol Motil Off J Eur Gastrointest Motil Soc. 2009;21:746-e46. https://pubmed-ncbi-nlm-nih-gov.uchile.idm.oclc.org/19220755/
  • Wiskur B, Greenwood-Van Meerveld B. The aging colon: the role of enteric neurodegeneration in constipation. Curr Gastroenterol Rep. 2010;12:507–512. doi:10.1007/s11894-010-0139-7
  • McLachlan AJ, Bath S, Naganathan V, et al. Clinical pharmacology of analgesic medicines in older people: impact of frailty and cognitive impairment. Br J Clin Pharmacol. 2011;71:351–364. doi:10.1111/j.1365-2125.2010.03847.x
  • Farage MA, Miller KW, Elsner P, et al. Characteristics of the aging skin. Adv Wound Care. 2013;2:5–10. doi:10.1089/wound.2011.0356
  • Miller MR. Structural and physiological age-associated changes in aging lungs. Semin Respir Crit Care Med. 2010;31:521–527. doi:10.1055/s-0030-1265893
  • Hochhegger B, de Meireles GP, Irion K, et al. O tórax e o envelhecimento: manifestações radiológicas. J Bras Pneumol. 2012;38:656–665. doi:10.1590/S1806-37132012000500016
  • Sprung J, Gajic O, Warner DO. Review article: age related alterations in respiratory function - anesthetic considerations. Can J Anesth. 2006;53:1244–1257. doi:10.1007/BF03021586
  • Ishaq A, Schröder J, Edwards N, et al. Dietary restriction ameliorates age-related increase in DNA damage,: senescence and inflammation in mouse adipose tissuey. J Nutr Health Aging. 2018;22:555–561. doi:10.1007/s12603-017-0968-2
  • Hughes VA, Roubenoff R, Wood M, et al. Anthropometric assessment of 10-y changes in body composition in the elderly. Am J Clin Nutr. 2004;80:475–482. doi:10.1093/ajcn/80.2.475
  • Lei SF, Liu MY, Chen XD, et al. Relationship of total body fatness and five anthropometric indices in Chinese aged 20–40 years: different effects of age and gender. Eur J Clin Nutr. 2006;60:511–518. doi:10.1038/sj.ejcn.1602345
  • Raguso CA, Kyle U, Kossovsky MP, et al. A 3-year longitudinal study on body composition changes in the elderly: role of physical exercise. Clin Nutr. 2006;25:573–580. doi:10.1016/j.clnu.2005.10.013
  • Mitchell WK, Williams J, Atherton P, et al. Sarcopenia: dynapenia, and the impact of advancing age on human skeletal muscle size and strength; a quantitative review. Front Physiol. 2012;3:260–260. doi:10.3389/fphys.2012.00260
  • Rodríguez-Julbe MC, Ramírez-Ronda CH, Arroyo E, et al. Antibiotics in older adults. P R Health Sci J. 2004;23:25–33.
  • Hanratty CG, McGlinchey P, Johnston GD, et al. Differential pharmacokinetics of digoxin in elderly patients. Drugs Aging. 2000;17:353–362. doi:10.2165/00002512-200017050-00003
  • Greenblatt DJ, Harmatz JS, Zhang Q, et al. Slow accumulation and elimination of diazepam and its active metabolite with extended treatment in the elderly. J Clin Pharmacol. 2021;61:193–203. doi:10.1002/jcph.1726
  • Cabrerizo S, Cuadras D, Gomez-Busto F, et al. Serum albumin and health in older people: review and meta analysis. Maturitas. 2015;81:17–27. doi:10.1016/j.maturitas.2015.02.009
  • Chudzik M, Maciązek-Jurczyk M, Pawełczak B, et al. Spectroscopic studies on the molecular ageing of serum albumin. Mol Basel Switz. 2016;22:E34–E34.
  • Silva-Fhon JR, Rojas-Huayta VM, Aparco-Balboa JP, et al. Sarcopenia and blood albumin: a systematic review with meta-analysis. Biomed Rev Inst Nac Salud. 2021;41:590–603.
  • Park HN, Song MJ, Choi YE, et al. LRG1 promotes ECM integrity by activating the TGF-β signaling pathway in fibroblasts. Int J Mol Sci. 2023;24:12445. doi:10.3390/ijms241512445
  • Grandison MK, Boudinot FD. Age-related changes in protein binding of drugs. Clin Pharmacokinet 2000. 2012;383:271–290.
  • Benet LZ, Hoener BA. Changes in plasma protein binding have little clinical relevance. Clin Pharmacol Ther. 2002;71:115–121. doi:10.1067/mcp.2002.121829
  • Warren A, Chaberek S, Ostrowski K, et al. Effects of old age on vascular complexity and dispersion of the hepatic sinusoidal network. Microcirculation. 2008;15:191–191. doi:10.1080/10739680701600856
  • Tajiri K, Shimizu Y. Liver physiology and liver diseases in the elderly. World J Gastroenterol. 2013;19:8459–8467. doi:10.3748/wjg.v19.i46.8459
  • Rattanacheeworn P, Kerr SJ, Kittanamongkolchai W, et al. Quantification of CYP3A and drug transporters activity in healthy young, healthy elderly and chronic kidney disease elderly patients by a microdose cocktail approach. Front Pharmacol. 2021;12:726669. doi:10.3389/fphar.2021.726669
  • Klotz U. Pharmacokinetics and drug metabolism in the elderly. Drug Metab Rev. 2009;41:67–76. doi:10.1080/03602530902722679
  • Long DA, Mu W, Price KL, et al. Blood vessels and the aging kidney. Nephron Exp Nephrol. 2005;101:e95–e99. doi:10.1159/000087146
  • Emamian SA, Nielsen MB, Pedersen JF, et al. Kidney dimensions at sonography: correlation with age,: sex, and habitus in 665 adult volunteers. AJR. Am J Roentgenol. 2013;160:83–86. doi:10.2214/ajr.160.1.8416654
  • Gourtsoyiannis N, Prassopoulos P, Cavouras D, et al. The thickness of the renal parenchyma decreases with age: a CT study of 360 patients. AJR. Am J Roentgenol. 2013;155:541–544. doi:10.2214/ajr.155.3.2117353
  • Roseman DA, Hwang SJ, Oyama-Manabe N, et al. Clinical associations of total kidney volume: the Framingham heart study. Nephrol Dial Transplant. 2017;32:1344–1350.
  • Zhou XJ, Rakheja D, Yu X, et al. The aging kidney. Kidney Int. 2008;74:710–720. doi:10.1038/ki.2008.319
  • Denic A, Lieske JC, Chakkera HA, et al. The substantial loss of nephrons in healthy human kidneys with aging. J Am Soc Nephrol. 2017;28:313–320. doi:10.1681/ASN.2016020154
  • Glassock RJ, Rule AD. The implications of anatomical and functional changes of the aging kidney: with an emphasis on the glomeruli. Kidney Int. 2012;82:270–277. doi:10.1038/ki.2012.65
  • Bolignano D, Mattace-Raso F, Sijbrands EJG, et al. The aging kidney revisited: a systematic review. Ageing Res Rev. 2014;14:65–80. doi:10.1016/j.arr.2014.02.003
  • Glassock RJ, Rule AD. Aging and the kidneys: anatomy, physiology and consequences for defining chronic kidney disease. Nephron. 2016;134:25–29. doi:10.1159/000445450
  • O’Sullivan ED, Hughes J, Ferenbach DA. Renal aging: causes and consequences. J Am Soc Nephrol JASN. 2017;28:407–420. doi:10.1681/ASN.2015121308
  • James MT, Hemmelgarn BR, Wiebe N, et al. Glomerular filtration rate, proteinuria, and the incidence and consequences of acute kidney injury: a cohort study. Lancet. 2010;376:2096–2103. doi:10.1016/S0140-6736(10)61271-8
  • López-Otín C, Blasco MA, Partridge L, et al. Hallmarks of aging: an expanding universe. Cell. 2023;186:243–278. doi:10.1016/j.cell.2022.11.001
  • Martínez de Toda I, Ceprián N, Díaz-Del Cerro E, et al. The role of immune cells in oxi-inflamm-aging. Cells. 2021;10:2974. doi:10.3390/cells10112974
  • Sies H, Berndt C, Jones DP. Oxidative stress. Annu Rev Biochem. 2017;86:715–748. doi:10.1146/annurev-biochem-061516-045037
  • Jones DP. Radical-free biology of oxidative stress. Am J Physiol Cell Physiol. 2008;295:C849–C868. doi:10.1152/ajpcell.00283.2008
  • Liu X, Hussain R, Mehmood K, et al. Mitochondrial-endoplasmic reticulum communication-mediated oxidative stress and autophagy. BioMed Res Int. 2022;2022.6459585. doi:10.1155/2022/6459585. PMID: 36164446.
  • Sies H. Chapter 13 - Oxidative stress: eustress and distress in redox homeostasis. In: Fink G, editor. Stress physiol biochem pathol. Düsseldorf, Germany: Academic Press; 2019. p. 153–163. [cited 2023 Nov 11]. Available from: https://www.sciencedirect.com/science/article/pii/B9780128131466000138.
  • Janaszak-Jasiecka A, Płoska A, Wierońska JM, et al. Endothelial dysfunction due to eNOS uncoupling: molecular mechanisms as potential therapeutic targets. Cell Mol Biol Lett. 2023;28:21. doi:10.1186/s11658-023-00423-2
  • Liu J, Kang R, Tang D. Signaling pathways and defense mechanisms of ferroptosis. FEBS J. 2022;289:7038–7050. doi:10.1111/febs.16059
  • Sies H. Total antioxidant capacity: Appraisal of a Concept12. J Nutr. 2007;137:1493–1495. doi:10.1093/jn/137.6.1493
  • Suzuki T, Yamamoto M. Stress-sensing mechanisms and the physiological roles of the Keap1–Nrf2 system during cellular stress. J Biol Chem. 2017;292:16817–16824. doi:10.1074/jbc.R117.800169
  • Kurutas EB. The importance of antioxidants which play the role in cellular response against oxidative/nitrosative stress: current state. Nutr J. 2016;151:1–22.
  • Lesnefsky EJ, Gudz TI, Moghaddas S, et al. Aging decreases electron transport complex III activity in heart interfibrillar mitochondria by alteration of the cytochrome c binding site. J Mol Cell Cardiol. 2001;33:37–47. doi:10.1006/jmcc.2000.1273
  • Miwa S, Kashyap S, Chini E, et al. Mitochondrial dysfunction in cell senescence and aging. J Clin Invest. 2022;132:e158447. doi:10.1172/JCI158447
  • Boengler K, Kosiol M, Mayr M, et al. Mitochondria and ageing: role in heart,: skeletal muscle and adipose tissue. J Cachexia Sarcopenia Muscle. 2017;8:349–369. doi:10.1002/jcsm.12178
  • Miwa S, Jow H, Baty K, et al. Low abundance of the matrix arm of complex I in mitochondria predicts longevity in mice. Nat Commun. 2014;5:3837–3837. doi:10.1038/ncomms4837
  • Espinoza SE, Guo H, Fedarko N, et al. Glutathione peroxidase enzyme activity in aging. J Gerontol A Biol Sci Med Sci. 2008;63:505–509. doi:10.1093/gerona/63.5.505
  • Aigner T, Fundel K, Saas J, et al. Large-scale gene expression profiling reveals major pathogenetic pathways of cartilage degeneration in osteoarthritis. Arthritis Rheum. 2006;54:3533–3544. doi:10.1002/art.22174
  • Ruiz-Romero C, Calamia V, Mateos J, et al. Mitochondrial dysregulation of osteoarthritic human articular chondrocytes analyzed by proteomics: a decrease in mitochondrial superoxide dismutase points to a redox imbalance. Mol Cell Proteomics MCP. 2008;8:172–189. doi:10.1074/mcp.M800292-MCP200
  • Scott JL, Gabrielides C, Davidson RK, et al. Superoxide dismutase downregulation in osteoarthritis progression and end-stage disease. Ann Rheum Dis. 2010;69:1502–1510. doi:10.1136/ard.2009.119966
  • Nissanka N, Moraes CT. Mitochondrial DNA damage and reactive oxygen species in neurodegenerative disease. FEBS Lett. 2018;592:728–742. doi:10.1002/1873-3468.12956
  • Bota DA, Davies KJA. Mitochondrial Lon protease in human disease and aging: including an etiologic classification of Lon-related diseases and disorders. Free Radic Biol Med. 2016;100:188–198. doi:10.1016/j.freeradbiomed.2016.06.031
  • Ahola S, Langer T, Macvicar T, et al. Mitochondrial proteolysis and metabolic control. Cold Spring Harb Perspect Biol. 2019;11:a033936–a033936. doi:10.1101/cshperspect.a033936
  • Sebastián D, Sorianello E, Segalés J, et al. Mfn2 deficiency links age-related sarcopenia and impaired autophagy to activation of an adaptive mitophagy pathway. EMBO J. 2016;35:1677–1693. doi:10.15252/embj.201593084
  • Chun SK, Lee S, Flores-Toro J, et al. Loss of sirtuin 1 and mitofusin 2 contributes to enhanced ischemia/reperfusion injury in aged livers. Aging Cell. 2018;17:e12761–e12761. doi:10.1111/acel.12761
  • Medala VK, Gollapelli B, Dewanjee S, et al. Mitochondrial dysfunction,: mitophagy, and role of dynamin-related protein 1 in Alzheimer’s disease. J Neurosci Res. 2021;99:1120–1135. doi:10.1002/jnr.24781
  • Alexeyev MF. Is there more to aging than mitochondrial DNA and reactive oxygen species? FEBS J. 2009;276:5768–5787. doi:10.1111/j.1742-4658.2009.07269.x
  • Wolf AM. MtDNA mutations and aging—not a closed case after all? Signal Transduct Target Ther. 2021;6.56.
  • Lagouge M, Argmann C, Gerhart-Hines Z, et al. Resveratrol improves mitochondrial function and protects against metabolic disease by activating SIRT1 and PGC-1α. Cell. 2006;127:1109–1122. doi:10.1016/j.cell.2006.11.013
  • Lira VA, Benton CR, Yan Z, et al. PGC-1α regulation by exercise training and its influences on muscle function and insulin sensitivity. Am J Physiol - Endocrinol Metab. 2010;299.E145-61.
  • Joseph AM, Hood DA. Relationships between exercise, mitochondrial biogenesis and type 2 diabetes. Diabetes Phys Act. 2014;60:48–61.
  • Hou CY, Tain YL, Yu HR, et al. The effects of resveratrol in the treatment of metabolic syndrome. Int J Mol Sci. 2019;20:535. doi:10.3390/ijms20030535
  • Rodop BB, Erbaş O. Mitochondrial dysfunction in aging and disease: the role of nutrition. 2023. Available from: https://www.researchgate.net/publication/371071283.
  • Das SK, Vasudevan DM. Alcohol-induced oxidative stress. Life Sci. 2007;81:177–187. doi:10.1016/j.lfs.2007.05.005
  • Horinouchi T, Higashi T, Mazaki Y, et al. Carbonyl compounds in the gas phase of cigarette mainstream smoke and their pharmacological properties. Biol Pharm Bull. 2016;39:909–914. doi:10.1248/bpb.b16-00025
  • Goel R, Bitzer Z, Reilly SM, et al. Variation in free radical yields from U.S. marketed cigarettes. Chem Res Toxicol. 2017;30:1038–1045. doi:10.1021/acs.chemrestox.6b00359
  • Sánchez-Rodríguez MA, Zacarías-Flores M, Correa-Muñoz E, et al. Oxidative stress risk is increased with a sedentary lifestyle during aging in Mexican women. Oxid Med Cell Longev. 2021;2021:9971765–9971765. doi:10.1155/2021/9971765
  • Calabrese V, Cornelius C, Mancuso C, et al. Cellular stress response: a novel target for chemoprevention and nutritional neuroprotection in aging, neurodegenerative disorders and longevity. Neurochem Res. 2008;33:2444–2471. doi:10.1007/s11064-008-9775-9
  • Calabrese V, Cornelius C, Dinkova-Kostova AT, et al. Vitagenes,: cellular stress response, and acetylcarnitine: relevance to hormesis. BioFactors Oxf Engl. 2009;35:146–160. doi:10.1002/biof.22
  • Son TG, Camandola S, Mattson MP. Hormetic dietary phytochemicals. Neuromolecular Med. 2008;10:236–246. doi:10.1007/s12017-008-8037-y
  • Calabrese EJ. Another California milestone: the first application of Hormesis in litigation and regulation. Int J Toxicol. 2008;27:31–33. doi:10.1080/10915810701876554
  • Iqbal AM, Jamal SF. Essential Hypertension. Treasure Island (FL): StatPearls Publishing; 2023; [cited 2023 Jun 14]. Available from: http://www.ncbi.nlm.nih.gov/books/NBK539859/.
  • Muntner P, Carey RM, Gidding S, et al. Potential US population impact of the 2017 ACC/AHA high blood pressure guideline. Circulation. 2018;137:109–118. doi:10.1161/CIRCULATIONAHA.117.032582
  • Whelton PK, Carey RM, Aronow WS, et al. ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA guideline for the prevention, detection, evaluation, and management of high blood pressure in adults: executive summary: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Hypertension. 2018;71:1269–1324. doi:10.1161/HYP.0000000000000066
  • World Health Organization. Hypertension [Internet]. 2023. Available from: https://www.who.int/news-room/fact-sheets/detail/hypertension.
  • Franco C, Sciatti E, Favero G, et al. Essential hypertension and oxidative stress: novel future perspectives. Int J Mol Sci. 2022;23(22):14489. Available from: /pmc/articles/PMC9692622/.
  • do Vale GT, Tirapelli CR. Are reactive oxygen species important mediators of vascular dysfunction? Curr Hypertens Rev. 2020;16:163–165. doi:10.2174/15734021OTgxdMTQaTcVY
  • Pan Y, Qiao QY, Pan LH, et al. Losartan reduces insulin resistance by inhibiting oxidative stress and enhancing insulin signaling transduction. Exp Clin Endocrinol Diabetes Off J Ger Soc Endocrinol Ger Diabetes Assoc. 2015;123:170–177.
  • Rivasi G, Rafanelli M, Mossello E, et al. Drug-related orthostatic hypotension: beyond anti-hypertensive medications. Drugs Aging. 2020;37:725–738. doi:10.1007/s40266-020-00796-5
  • Lapi F, Azoulay L, Yin H, et al. Concurrent use of diuretics, angiotensin converting enzyme inhibitors, and angiotensin receptor blockers with non-steroidal anti-inflammatory drugs and risk of acute kidney injury: nested case-control study. Br Med J. 2013;346:e8525. doi:10.1136/bmj.e8525
  • Morris EJ, Brown JD, Manini TM, et al. Differences in health-related quality of life among adults with a potential dihydropyridine calcium channel blocker-loop diuretic prescribing cascade. Drugs Aging. 2021;38:625–632. doi:10.1007/s40266-021-00868-0
  • Scheltens P, De Strooper B, Kivipelto M, et al. Alzheimer’s disease. Lancet Lond Engl. 2021;397:1577–1577. doi:10.1016/S0140-6736(20)32205-4
  • Alzheimer’s Association. Alzheimer’s disease facts and figures. Alzheimers Dement J Alzheimers Assoc. 2022;18:700–789. doi:10.1002/alz.12638
  • Andreone BJ, Lacoste B, Gu C. Neuronal and vascular interactions. Annu Rev Neurosci. 2015;38:25–46. doi:10.1146/annurev-neuro-071714-033835
  • Yoon JH, Seo Y, Jo YS, et al. Brain lipidomics: from functional landscape to clinical significance. Sci Adv. 2022;8:eadc9317. doi:10.1126/sciadv.adc9317
  • Cutler RG, Kelly J, Storie K, et al. Involvement of oxidative stress-induced abnormalities in ceramide and cholesterol metabolism in brain aging and Alzheimer’s disease. Proc Natl Acad Sci U S A. 2004;101:2070–2070. doi:10.1073/pnas.0305799101
  • Shoeb M, Ansari NH, Srivastava SK, et al. 4-hydroxynonenal in the pathogenesis and progression of human diseases. Curr Med Chem. 2014;21:230–230. doi:10.2174/09298673113209990181
  • Singh M, Dang TN, Arseneault M, et al. Role of by-products of lipid oxidation in Alzheimer’s disease brain: a focus on acrolein. J Alzheimers Dis JAD. 2010;21:741–756. doi:10.3233/JAD-2010-100405
  • Zhao Y, Zhao B. Oxidative stress and the pathogenesis of Alzheimer’s disease. Oxid Med Cell Longev. 2013:316523. Available from: /pmc/articles/PMC3745981/
  • Butterfield DA, Kanski J. Brain protein oxidation in age-related neurodegenerative disorders that are associated with aggregated proteins. Mech Ageing Dev. 2001;122:945–962. doi:10.1016/S0047-6374(01)00249-4
  • Ahmed N, Ahmed U, Thornalley PJ, et al. Protein glycation: oxidation and nitration adduct residues and free adducts of cerebrospinal fluid in Alzheimer’s disease and link to cognitive impairment. J Neurochem. 2005;92:255–263. doi:10.1111/j.1471-4159.2004.02864.x
  • Al-Hilaly YK, Williams TL, Stewart-Parker M, et al. A central role for dityrosine crosslinking of amyloid-β in Alzheimer’s disease. Acta Neuropathol Commun. 2013;1:83. doi:10.1186/2051-5960-1-83
  • La Penna G, Hureau C, Andreussi O, et al. Identifying: by first-principles simulations, Cu[amyloid-β] species making Fenton-type reactions in Alzheimer’s disease. J Phys Chem B. 2013;117:16455–16467. doi:10.1021/jp410046w
  • Chen K, Jiang X, Wu M, et al. Ferroptosis, a potential therapeutic target in Alzheimer’s disease. Front Cell Dev Biol. 2021;9:704298. Available from: https://pubmed-ncbi-nlm-nih-gov.uchile.idm.oclc.org/34422824/.
  • Calabrese V, Mancuso C, Calvani M, et al. Nitric oxide in the central nervous system: neuroprotection versus neurotoxicity. Nat Rev Neurosci. 2007;8:766–775. doi:10.1038/nrn2214
  • Picón-Pagès P, Garcia-Buendia J, Muñoz FJ. Functions and dysfunctions of nitric oxide in brain. Biochim Biophys Acta Mol Basis Dis. 2019;1865:1949–1967. doi:10.1016/j.bbadis.2018.11.007
  • Chachlaki K, Prevot V. Nitric oxide signalling in the brain and its control of bodily functions. Br J Pharmacol. 2020;177:5437–5458. doi:10.1111/bph.14800
  • Pomilio AB, Vitale AA, Lazarowski AJ. COVID-19 and Alzheimer’s disease: neuroinflammation: oxidative stress, ferroptosis, and mechanisms involved. Curr Med Chem. 2023;30(35):3993–4031.
  • Phillips MI, de Oliveira EM. Brain renin angiotensin in disease. J Mol Med Berl Ger. 2008;86:715–722. doi:10.1007/s00109-008-0331-5
  • Ababei D-C, Bild V, Macadan I, et al. Therapeutic implications of renin-angiotensin system modulators in Alzheimer’s dementia. Pharmaceutics. 2023;15:2290. doi:10.3390/pharmaceutics15092290
  • Hu W, Li Y, Zhao Y, et al. Telmisartan and Rosuvastatin synergistically ameliorate dementia and cognitive impairment in older hypertensive patients with apolipoprotein E genotype. Front Aging Neurosci. 2020;12:154. doi:10.3389/fnagi.2020.00154
  • Liu C-H, Sung P-S, Li Y-R, et al. Telmisartan use and risk of dementia in type 2 diabetes patients with hypertension: a population-based cohort study. PLoS Med. 2021;18:e1003707. doi:10.1371/journal.pmed.1003707
  • Sharma K. Cholinesterase inhibitors as Alzheimer’s therapeutics (Review). Mol Med Rep. 2019;20:1479–1487.
  • Sangaleti CT, Katayama KY, De Angelis K, et al. The cholinergic drug galantamine alleviates oxidative stress alongside anti-inflammatory and cardio-metabolic effects in subjects with the metabolic syndrome in a randomized trial. Front Immunol. 2021;12:613979. doi:10.3389/fimmu.2021.613979
  • Kundo NK, Manik MIN, Biswas K, et al. Identification of polyphenolics from Loranthus globosus as potential inhibitors of cholinesterase and oxidative stress for Alzheimer’s disease treatment. BioMed Res Int. 2021:9154406.
  • Althobaiti YS. Development of memantine as a drug for Alzheimer’s disease: a review of preclinical and clinical studies. Trop J Pharm Res. 2020;19:1535–1540. doi:10.4314/tjpr.v19i7.28
  • Matsunaga S, Kishi T, Nomura I, et al. The efficacy and safety of memantine for the treatment of Alzheimer’s disease. Expert Opin Drug Saf. 2018;17:1053–1061. doi:10.1080/14740338.2018.1524870
  • Guo J, Wang Z, Liu R, et al. Memantine, donepezil, or combination therapy-what is the best therapy for Alzheimer’s disease? A network meta-analysis. Brain Behav. 2020;10(11):e01831.
  • Press D, Buss S. Treatment of Alzheimer disease. Cholinesterase Inhib Treat Dement. 2021. Available from: https://www.uptodate.com/contents/treatment-of-alzheimer-disease
  • Livingston G, Huntley J, Sommerlad A, et al. Dementia prevention, intervention, and care: 2020 report of the Lancet Commission. Lancet Lond Engl. 2020;396:413–446. doi:10.1016/S0140-6736(20)30367-6
  • Allen KD, Thoma LM, Golightly YM. Epidemiology of osteoarthritis. Osteoarthritis Cartilage. 2022;30:184–195. doi:10.1016/j.joca.2021.04.020
  • Johnson VL, Hunter DJ. The epidemiology of osteoarthritis. Best Pract Res Clin Rheumatol. 2014;28:5–15. doi:10.1016/j.berh.2014.01.004
  • Hawker GA, King LK. The burden of osteoarthritis in older adults. Clin Geriatr Med. 2022;38:181–192. doi:10.1016/j.cger.2021.11.005
  • Coryell PR, Diekman BO, Loeser RF. Mechanisms and therapeutic implications of cellular senescence in osteoarthritis. Nat Rev Rheumatol. 2021;17:47–57. doi:10.1038/s41584-020-00533-7
  • Franceschi C, Garagnani P, Parini P, et al. Inflammaging: a new immune–metabolic viewpoint for age-related diseases. Nat Rev Endocrinol. 2018;14:576–590. doi:10.1038/s41574-018-0059-4
  • Jeon OH, David N, Campisi J, et al. Senescent cells and osteoarthritis: a painful connection. J Clin Invest. 2018;128:1229–1237. doi:10.1172/JCI95147
  • Delmonico MJ, Harris TB, Visser M, et al. Longitudinal study of muscle strength, quality, and adipose tissue infiltration. Am J Clin Nutr. 2009;90:1579–1579. doi:10.3945/ajcn.2009.28047
  • Sellam J, Berenbaum F. Is osteoarthritis a metabolic disease? Joint Bone Spine. 2013;80:568–573. doi:10.1016/j.jbspin.2013.09.007
  • Courties A, Gualillo O, Berenbaum F, et al. Metabolic stress-induced joint inflammation and osteoarthritis. Osteoarthritis Cartilage. 2015;23:1955–1965. doi:10.1016/j.joca.2015.05.016
  • Loeser RF, Collins JA, Diekman BO. Ageing and the pathogenesis of osteoarthritis. Nat Rev Rheumatol. 2016;12:412–412. doi:10.1038/nrrheum.2016.65
  • Gossan N, Zeef L, Hensman J, et al. The circadian clock in murine chondrocytes regulates genes controlling key aspects of cartilage homeostasis. Arthritis Rheum. 2013;65:2334–2345. doi:10.1002/art.38035
  • Jones DP. Redox theory of aging. Redox Biol. 2015;5:71–71. doi:10.1016/j.redox.2015.03.004
  • Nemoto S, Finkel T. Redox regulation of forkhead proteins through a p66shc-dependent signaling pathway. Science. 2002;295:2450–2452. doi:10.1126/science.1069004
  • Mazumder S, De R, Debsharma S, et al. Indomethacin impairs mitochondrial dynamics by activating the PKCζ-p38-DRP1 pathway and inducing apoptosis in gastric cancer and normal mucosal cells. J Biol Chem. 2019;294:8238–8258. doi:10.1074/jbc.RA118.004415
  • Mazumder S, De R, Sarkar S, et al. Selective scavenging of intra-mitochondrial superoxide corrects diclofenac-induced mitochondrial dysfunction and gastric injury: a novel gastroprotective mechanism independent of gastric acid suppression. Biochem Pharmacol. 2016;121:33–51. doi:10.1016/j.bcp.2016.09.027
  • Koike M, Nojiri H, Ozawa Y, et al. Mechanical overloading causes mitochondrial superoxide and SOD2 imbalance in chondrocytes resulting in cartilage degeneration. Sci Rep. 2015;5:11722.
  • Hanachi N, Charef N, Baghiani A, et al. Comparison of xanthine oxidase levels in synovial fluid from patients with rheumatoid arthritis and other joint inflammations. Saudi Med J. 2009;30:1422–1425.
  • Yin W, Park JI, Loeser RF. Oxidative stress inhibits insulin-like growth factor-I induction of chondrocyte proteoglycan synthesis through differential regulation of phosphatidylinositol 3-Kinase-Akt and MEK-ERK MAPK signaling pathways. J Biol Chem. 2009;284:31972–31981. doi:10.1074/jbc.M109.056838
  • Wood ST, Long DL, Reisz JA, et al. Cysteine-mediated redox regulation of cell signaling in chondrocytes stimulated with fibronectin fragments. Arthritis Rheumatol Hoboken NJ. 2016;68:117–126. doi:10.1002/art.39326
  • Collins JA, Wood ST, Nelson KJ, et al. Oxidative stress promotes peroxiredoxin hyperoxidation and attenuates pro-survival signaling in aging chondrocytes. J Biol Chem. 2016;291:6641–6654. doi:10.1074/jbc.M115.693523
  • Miao Y, Chen Y, Xue F, et al. Contribution of ferroptosis and GPX4’s dual functions to osteoarthritis progression. EBioMedicine. 2022;76:103847 doi:10.1016/j.ebiom.2022.103847.
  • Bennell KL, Hunter DJ, Hinman RS. Management of osteoarthritis of the knee. Br Med J. 2012;345:e493410.1136/bmj.e4934.
  • McAlindon TE, Bannuru RR, Sullivan MC, et al. OARSI guidelines for the non-surgical management of knee osteoarthritis. Osteoarthritis Cartilage. 2014;22:363–388. doi:10.1016/j.joca.2014.01.003
  • Kompel AJ, Roemer FW, Murakami AM, et al. Intra-articular corticosteroid injections in the hip and knee: perhaps not as safe as we thought? Radiology. 2019;293:656–663. doi:10.1148/radiol.2019190341
  • Sandoval-Acuña C, Lopez-Alarcón C, Aliaga ME, et al. Inhibition of mitochondrial complex I by various non-steroidal anti-inflammatory drugs and its protection by quercetin via a coenzyme Q-like action. Chem Biol Interact. 2012;199:18–28. doi:10.1016/j.cbi.2012.05.006
  • Shin SJ, Noh CK, Lim SG, et al. Non-steroidal anti-inflammatory drug-induced enteropathy. Intest Res. 2017;15:446–455. doi:10.5217/ir.2017.15.4.446
  • Watanabe T, Fujiwara Y, Chan FKL. Current knowledge on non-steroidal anti-inflammatory drug-induced small-bowel damage: a comprehensive review. J Gastroenterol. 2020;55:481–495. doi:10.1007/s00535-019-01657-8
  • Baigent C, Bhala N, Emberson J, et al. Vascular and upper gastrointestinal effects of non-steroidal anti-inflammatory drugs: meta-analyses of individual participant data from randomised trials. Lancet Lond Engl. 2013;382:769–779. doi:10.1016/S0140-6736(13)60900-9
  • Huerta C, Castellsague J, Varas-Lorenzo C, et al. Nonsteroidal anti-inflammatory drugs and risk of ARF in the general population. Am J Kidney Dis. 2005;45:531–539. doi:10.1053/j.ajkd.2004.12.005
  • Ory MG, Anderson LA, Friedman DB, et al. Cancer prevention among adults aged 45-64 years: setting the stage. Am J Prev Med. 2014;46:(3 Suppl 1):S1–6. doi:10.1016/j.amepre.2013.10.027.
  • Pilleron S, Sarfati D, Janssen-Heijnen M, et al. Global cancer incidence in older adults, 2012 and 2035: a population-based study. Int J Cancer. 2019;144:49–58. doi:10.1002/ijc.31664
  • Novak J, Goldberg A, Dharmarajan K, et al. Polypharmacy in older adults with cancer undergoing radiotherapy: a review. J Geriatr Oncol. 2022;13:778–778. doi:10.1016/j.jgo.2022.02.007
  • Zabłocka-Słowińska K, Płaczkowska S, Skórska K, et al. Oxidative stress in lung cancer patients is associated with altered serum markers of lipid metabolism. PLoS One. 2019;14:e0215246. doi:10.1371/journal.pone.0215246
  • Desouki MM, Kulawiec M, Bansal S, et al. Cross talk between mitochondria and superoxide generating NADPH oxidase in breast and ovarian tumors. Cancer Biol Ther. 2005;4:1367–1373. doi:10.4161/cbt.4.12.2233
  • Kawahara T, Kohjima M, Kuwano Y, et al. Helicobacter pylori lipopolysaccharide activates Rac1 and transcription of NADPH oxidase Nox1 and its organizer NOXO1 in guinea pig gastric mucosal cells. Am J Physiol - Cell Physiol. 2005;288(2):C450–7. doi:10.1152/ajpcell.00319.2004.
  • Fukuyama M, Rokutan K, Sano T, et al. Overexpression of a novel superoxide-producing enzyme, NADPH oxidase 1, in adenoma and well differentiated adenocarcinoma of the human colon. Cancer Lett. 2005;221:97–104. doi:10.1016/j.canlet.2004.08.031
  • Block K, Gorin Y, New DD, et al. The NADPH oxidase subunit p22phox inhibits the function of the tumor suppressor protein tuberin. Am J Pathol. 2010;176:2447–2455. doi:10.2353/ajpath.2010.090606
  • Shimada K, Fujii T, Anai S, et al. ROS generation via NOX4 and its utility in the cytological diagnosis of urothelial carcinoma of the urinary bladder. BMC Urol. 2011;11:1–12. doi:10.1186/1471-2490-11-1
  • Jitschin R, Hofmann AD, Bruns H, et al. Mitochondrial metabolism contributes to oxidative stress and reveals therapeutic targets in chronic lymphocytic leukemia. Blood. 2014;123:2663–2672. doi:10.1182/blood-2013-10-532200
  • Hart PC, Mao M, De Abreu ALP, et al. MnSOD upregulation sustains the Warburg effect via mitochondrial ROS and AMPK-dependent signalling in cancer. Nat Commun. 2015;6:6053.
  • Foo BJ-A, Eu JQ, Hirpara JL, et al. Interplay between mitochondrial metabolism and cellular redox state dictates cancer cell survival. Oxid Med Cell Longev. 2021;2021:1341604.
  • Burdick AD, Davis JW, Liu KJ, et al. Benzo(a)pyrene quinones increase cell proliferation, generate reactive oxygen species, and transactivate the epidermal growth factor receptor in breast epithelial cells. Cancer Res. 2003;63:7825–7833.
  • Sarsour EH, Venkataraman S, Kalen AL, et al. Manganese superoxide dismutase activity regulates transitions between quiescent and proliferative growth. Aging Cell. 2008;7:405–417. doi:10.1111/j.1474-9726.2008.00384.x
  • Sieber OM, Heinimann K, Tomlinson IPM. Genomic instability - the engine of tumorigenesis? Nat Rev Cancer. 2003;3:701–708. doi:10.1038/nrc1170
  • Cooke MS, Evans MD, Dizdaroglu M, et al. Oxidative DNA damage: mechanisms: mutation, and disease. FASEB J. 2003;17:1195–1214. doi:10.1096/fj.02-0752rev
  • Moeller BJ, Cao Y, Li CY, et al. Radiation activates HIF-1 to regulate vascular radiosensitivity in tumors: role of reoxygenation: free radicals, and stress granules. Cancer Cell. 2004;5:429–441. doi:10.1016/S1535-6108(04)00115-1
  • Xu RH, Pelicano H, Zhou Y, et al. Inhibition of glycolysis in cancer cells: a novel strategy to overcome drug resistance associated with mitochondrial respiratory defect and hypoxia. Cancer Res. 2005;65:613–621. doi:10.1158/0008-5472.613.65.2
  • Yoo NJ, Kim HR, Kim YR, et al. Somatic mutations of the KEAP1 gene in common solid cancers. Histopathology. 2012;60:943–952. doi:10.1111/j.1365-2559.2012.04178.x
  • Sajadimajd S, Khazaei M. Oxidative stress and cancer: the role of Nrf2. Curr Cancer Drug Targets. 2018;18:538–557. doi:10.2174/1568009617666171002144228
  • Crociani O, Marzi I, Cipolleschi MG, et al. The unveiling of the Warburg effect and the inscribed innovative approach to a radical non toxic anticancer therapy. Cell Cycle Georget Tex. 2018;17:288–297. doi:10.1080/15384101.2017.1403679
  • Schiliro C, Firestein BL. Mechanisms of metabolic reprogramming in cancer cells supporting enhanced growth and proliferation. Cells. 2021;10:1056. doi:10.3390/cells10051056
  • Chen H, Yan Z, Wu S, et al. A glutathione-responsive polyphenol - constructed nanodevice for double roles in apoptosis and ferroptosis. Colloids Surf B Biointerfaces. 2021;205:111902. doi:10.1016/j.colsurfb.2021.111902
  • Ghigo A, Li M, Hirsch E. New signal transduction paradigms in anthracycline-induced cardiotoxicity. Biochim Biophys Acta BBA - Mol Cell Res. 2016;1863:1916–1925. doi:10.1016/j.bbamcr.2016.01.021
  • Stephens C, Andrade RJ, Lucena MI. Mechanisms of drug-induced liver injury. Curr Opin Allergy Clin Immunol. 2014;14:286–292. doi:10.1097/ACI.0000000000000070
  • Baumgart SJ, Haendler B. Drug-induced liver injury: cascade of events leading to cell death, apoptosis or necrosis. Int J Mol Sci. 2017;18:E1018–E1018. doi:10.3390/ijms18051017
  • Staff NP, Grisold A, Grisold W, et al. Chemotherapy-induced peripheral neuropathy: a current review. Ann Neurol. 2017;81:772–781. doi:10.1002/ana.24951
  • Ren X, Keeney JTR, Miriyala S, et al. The triangle of death of neurons: oxidative damage, mitochondrial dysfunction, and loss of choline-containing biomolecules in brains of mice treated with doxorubicin. Advanced insights into mechanisms of chemotherapy induced cognitive impairment (“chemobrain”) involving TNF-α. Free Radic Biol Med. 2018;134:1–8. doi:10.1016/j.freeradbiomed.2018.12.029
  • Herrmann J. Adverse cardiac effects of cancer therapies: cardiotoxicity and arrhythmia. Nat Rev Cardiol. 2020;17:474–502. doi:10.1038/s41569-020-0348-1
  • McElroy T, Allen AR. A bibliometric review of publications on oxidative stress and chemobrain: 1990-2019. Antioxid Basel Switz. 2020;9:E439–E439. doi:10.3390/antiox9050439
  • Man Q, Deng Y, Li P, et al. Licorice ameliorates cisplatin-induced hepatotoxicity through antiapoptosis, antioxidative stress, anti-inflammation, and acceleration of metabolism. Front Pharmacol. 2020;11:563750–563750. doi:10.3389/fphar.2020.563750
  • Fabiani I, Aimo A, Grigoratos C, et al. Oxidative stress and inflammation: determinants of anthracycline cardiotoxicity and possible therapeutic targets. Heart Fail Rev. 2020;26:881–890. doi:10.1007/s10741-020-10063-9
  • Pantazi D, Tselepis AD. Cardiovascular toxic effects of antitumor agents: pathogenetic mechanisms. Thromb Res. 2022;213:S95–102. doi:10.1016/j.thromres.2021.12.017
  • Li G, Ding K, Qiao Y, et al. Flavonoids regulate inflammation and oxidative stress in cancer. Molecules. 2020;25(23):5628. Available from: /pmc/articles/PMC7729519/.
  • Lee SG, Kim B, Yang Y, et al. Berry anthocyanins suppress the expression and secretion of proinflammatory mediators in macrophages by inhibiting nuclear translocation of NF-κB independent of NRF2-mediated mechanism. J Nutr Biochem. 2014;25:404–411. doi:10.1016/j.jnutbio.2013.12.001
  • Edirisinghe I, Banaszewski K, Cappozzo J, et al. Effect of black currant anthocyanins on the activation of endothelial nitric oxide synthase (eNOS) in vitro in human endothelial cells. J Agric Food Chem. 2011;59:8616–8624. doi:10.1021/jf201116y
  • World Health Organization. Obesity [Internet]. Available from: https://www.who.int/health-topics/obesity#tab = tab_1.
  • Chooi YC, Ding C, Magkos F. The epidemiology of obesity. Metabolism. 2019;92:6–10. doi:10.1016/j.metabol.2018.09.005
  • Colleluori G, Villareal DT. Aging: obesity, sarcopenia and the effect of diet and exercise intervention. Exp Gerontol. 2021;155:111561. doi:10.1016/j.exger.2021.111561
  • Cruz-Jentoft AJ, Bahat G, Bauer J, et al. Sarcopenia: revised European consensus on definition and diagnosis. Age Ageing. 2019;48:16–31. doi:10.1093/ageing/afy169
  • Le Lay S, Simard G, Martinez MC, et al. Oxidative stress and metabolic pathologies: from an adipocentric point of view. Oxid Med Cell Longev. 2014;2014:908539. Available from: /pmc/articles/PMC4131099/.
  • Khalil A, Berrougui H. Mechanism of action of resveratrol in lipid metabolism and atherosclerosis. Clin. Lipidol. 2017;4:527–531. doi:10.2217/clp.09.53
  • Meneses MJ, Silvestre R, Sousa-Lima I, et al. Paraoxonase-1 as a regulator of glucose and lipid homeostasis: impact on the onset and progression of metabolic disorders. Int J Mol Sci. 2019;20. doi:10.3390/ijms20164049
  • Hansel B, Giral P, Nobecourt E, et al. Metabolic syndrome is associated with elevated oxidative stress and dysfunctional dense high-density lipoprotein particles displaying impaired antioxidative activity. J Clin Endocrinol Metab. 2004;89:4963–4971. doi:10.1210/jc.2004-0305
  • Holvoet P. Relations between metabolic syndrome: oxidative stress and inflammation and cardiovascular disease. Verh K Acad Geneeskd Belg. 2008;70:193–219.
  • Maingrette F, Renier G. Leptin increases lipoprotein lipase secretion by macrophages: involvement of oxidative stress and protein kinase C. Diabetes. 2003;52:2121–2128. doi:10.2337/diabetes.52.8.2121
  • Chang C-C, Sia K-C, Chang J-F, et al. Participation of lipopolysaccharide in hyperplasic adipose expansion: involvement of NADPH oxidase/ROS/p42/p44 MAPK-dependent Cyclooxygenase-2. J Cell Mol Med. 2022;26:3850–3861. doi:10.1111/jcmm.17419
  • Silvester AJ, Aseer KR, Yun JW. Dietary polyphenols and their roles in fat browning. J Nutr Biochem. 2019;64:1–12. doi:10.1016/j.jnutbio.2018.09.028
  • Qiu H, Schlegel V. Impact of nutrient overload on metabolic homeostasis. Nutr Rev. 2018;76:693–707. doi:10.1093/nutrit/nuy023
  • Bhatti JS, Bhatti GK, Reddy PH. Mitochondrial dysfunction and oxidative stress in metabolic disorders - a step towards mitochondria based therapeutic strategies. Biochim Biophys Acta Mol Basis Dis. 2017;1863:1066–1077. doi:10.1016/j.bbadis.2016.11.010
  • Bouchez C, Devin A. Mitochondrial biogenesis and mitochondrial reactive oxygen species (ROS): a complex relationship regulated by the cAMP/PKA signaling pathway. Cells. 2019;8:287. doi:10.3390/cells8040287
  • Mulero MC, Huxford T, Ghosh G. NF-κB, IκB, and IKK: integral components of immune system signaling. Adv Exp Med Biol. 2019;1172:207–226. doi:10.1007/978-981-13-9367-9_10
  • Oliveira-Marques V, Marinho HS, Cyrne L, et al. Role of hydrogen peroxide in NF-kappaB activation: from inducer to modulator. Antioxid Redox Signal. 2009;11:2223–2243. doi:10.1089/ars.2009.2601
  • Martínez-Martínez E, Jurado-López R, Valero-Muñoz M, et al. Leptin induces cardiac fibrosis through galectin-3, mTOR and oxidative stress: potential role in obesity. J Hypertens. 2014;32:1104–1114. discussion 1114. doi:10.1097/HJH.0000000000000149
  • Xu X, Huang X, Zhang L, et al. Adiponectin protects obesity-related glomerulopathy by inhibiting ROS/NF-κB/NLRP3 inflammation pathway. BMC Nephrol. 2021;22(1):218. doi:10.1186/s12882-021-02391-1.
  • Mitrou P, Boutati E, Lambadiari V, et al. Insulin resistance in hyperthyroidism: the role of IL6 and TNF alpha. Eur J Endocrinol. 2010;162:121–126. doi:10.1530/EJE-09-0622
  • Bjørklund G, Peana M, Pivina L, et al. Iron deficiency in obesity and after bariatric surgery. Biomolecules. 2021;11(5):613. doi:10.3390/biom11050613.
  • González-Domínguez Á, Visiedo-García FM, Domínguez-Riscart J, et al. Iron metabolism in obesity and metabolic syndrome. Int J Mol Sci. 2020;21:5529. doi:10.3390/ijms21155529
  • Sombra LRS, Anastasopoulou C Pharmacologic Therapy for Obesity. [Updated 2022 Aug 29] In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2023 Jan. Available from: https://www-ncbi-nlm-nih-gov.uchile.idm.oclc.org/books/NBK562269/.
  • Kawanami D, Takashi Y. GLP-1 receptor agonists in diabetic kidney disease: from clinical outcomes to mechanisms. Front Pharmacol. 2020;11:967. doi:10.3389/fphar.2020.00967
  • Mehdi SF, Pusapati S, Anwar MS, et al. Glucagon-like peptide-1: a multi-faceted anti-inflammatory agent. Front Immunol. 2023;14:1148209. doi:10.3389/fimmu.2023.1148209
  • Hayes JD, Dinkova-Kostova AT. The Nrf2 regulatory network provides an interface between redox and intermediary metabolism. Trends Biochem Sci. 2014;39:199–218. doi:10.1016/j.tibs.2014.02.002
  • Dinkova-Kostova AT, Abramov AY. The emerging role of Nrf2 in mitochondrial function. Free Radic Biol Med. 2015;88:179–188. doi:10.1016/j.freeradbiomed.2015.04.036
  • Mangmool S, Hemplueksa P, Parichatikanond W, et al. Epac is required for GLP-1R-mediated inhibition of oxidative stress and apoptosis in cardiomyocytes. Mol Endocrinol Baltim Md. 2015;29:583–596. doi:10.1210/me.2014-1346
  • Fernández-Millán E, Martín MA, Goya L, et al. Glucagon-like peptide-1 improves beta-cell antioxidant capacity via extracellular regulated kinases pathway and Nrf2 translocation. Free Radic Biol Med. 2016;95:16–26. doi:10.1016/j.freeradbiomed.2016.03.002
  • Tsao CW, Aday AW, Almarzooq ZI, et al. Heart disease and stroke statistics-2022 update: a report from the American Heart Association. Circulation. 2022;145:E153–E639.
  • Kattoor AJ, Pothineni NVK, Palagiri D, et al. Oxidative stress in atherosclerosis. Curr Atheroscler Rep. 2017;19:42. doi:10.1007/s11883-017-0678-6
  • Ford TJ, Corcoran D, Berry C. Stable coronary syndromes: pathophysiology: diagnostic advances and therapeutic need. Heart Br Card Soc. 2018;104:284–292.
  • King TC. Cell injury, cellular responses to injury, and cell death. Elseviers Integr Pathol. 2007: 1–20.
  • Sanada S, Komuro I, Kitakaze M. Pathophysiology of myocardial reperfusion injury: preconditioning,: postconditioning, and translational aspects of protective measures. Am J Physiol Heart Circ Physiol. 2011;301. doi:10.1152/ajpheart.00553.2011
  • Kalogeris T, Baines CP, Krenz M, et al. Cell biology of ischemia/reperfusion injury. Int Rev Cell Mol Biol. 2012;298:229–229. doi:10.1016/B978-0-12-394309-5.00006-7
  • Yellon DM, Hausenloy DJ. Myocardial reperfusion injury [Internet]. Massachusetts Medical Society; 2007 [cited 2023 May 1]. Available from: https://www.nejm.org/doi/pdf/10.1056NEJMra071667.
  • Algoet M, Janssens S, Himmelreich U, et al. Myocardial ischemia-reperfusion injury and the influence of inflammation. Trends Cardiovasc Med. 2023;33:357–366. doi:10.1016/j.tcm.2022.02.005
  • Suski JM, Lebiedzinska M, Bonora M, et al. Relation between mitochondrial membrane potential and ROS formation. Methods Mol Biol Clifton NJ. 2012;810:183–205. doi:10.1007/978-1-61779-382-0_12
  • Wu MY, Yiang GT, Liao WT, et al. Current mechanistic concepts in ischemia and reperfusion injury. Cell Physiol Biochem. 2018;46:1650–1667. doi:10.1159/000489241
  • Neri M, Fineschi V, Paolo M, et al. Cardiac oxidative stress and inflammatory cytokines response after myocardial infarction. Curr Vasc Pharmacol. 2015;13:26–36. doi:10.2174/15701611113119990003
  • Zhao ZQ, Nakamura M, Wang NP, et al. Dynamic progression of contractile and endothelial dysfunction and infarct extension in the late phase of reperfusion. J Surg Res. 2000;94:133–144. doi:10.1006/jsre.2000.6029
  • Incalza MA, D’Oria R, Natalicchio A, et al. Oxidative stress and reactive oxygen species in endothelial dysfunction associated with cardiovascular and metabolic diseases. Vascul Pharmacol. 2018;100:1–19. doi:10.1016/j.vph.2017.05.005
  • Luo S, Lei H, Qin H, et al. Molecular mechanisms of endothelial NO synthase uncoupling. Curr Pharm Des. 2014;20:3548–3553. doi:10.2174/13816128113196660746
  • Garrido AM, Griendling KK. NADPH oxidases and angiotensin II receptor signaling. Mol Cell Endocrinol. 2009;302:148–158. doi:10.1016/j.mce.2008.11.003
  • Tibaut M, Mekis D, Petrovic D. Pathophysiology of myocardial infarction and acute management strategies. Cardiovasc Hematol Agents Med Chem. 2017;14:150–159. doi:10.2174/1871525714666161216100553
  • Sima P, Vannucci L, Vetvicka V. Atherosclerosis as autoimmune disease. Ann Transl Med. 2018;6:116. doi:10.21037/atm.2018.02.02
  • Sato A, Ueda C, Kimura R, et al. Angiotensin II induces the aggregation of native and oxidized low-density lipoprotein. Eur Biophys J EBJ. 2018;47:1–9. doi:10.1007/s00249-017-1208-8
  • Yeh HL, Kuo LT, Sung FC, et al. Association between polymorphisms of antioxidant gene (MnSOD, CAT, and GPx1) and risk of coronary artery disease. BioMed Res Int. 2018;2018:5086869. doi:10.1155/2018/5086869.
  • Förstermann U, Xia N, Li H. Roles of vascular oxidative stress and nitric oxide in the pathogenesis of atherosclerosis. Circ Res. 2017;120:713–735. doi:10.1161/CIRCRESAHA.116.309326
  • Singh A, Museedi AS, Grossman SA. Acute coronary syndrome. StatPearls [Internet]. 2022; Available from: https://www-ncbi-nlm-nih-gov.uchile.idm.oclc.org/books/NBK459157/.
  • Steinhubl SR. Why have antioxidants failed in clinical trials? Am J Cardiol. 2008;101(10A):14D–19D. doi:10.1016/j.amjcard.2008.02.003.
  • Rodrigo R, Prieto JC, Aguayo R, et al. Joint cardioprotective effect of vitamin C and other antioxidants against reperfusion injury in patients with acute myocardial infarction undergoing percutaneous coronary intervention. Molecules. 2021;26:5702. doi:10.3390/molecules26185702
  • Ahmed S, Ahmed N, Rungatscher A, et al. Cocoa flavonoids reduce inflammation and oxidative stress in a myocardial ischemia-reperfusion experimental model. Antioxid Basel Switz. 2020;9:167. doi:10.3390/antiox9020167
  • ElSayed NA, Aleppo G, Aroda VR, et al. Classification and diagnosis of diabetes: standards of care in diabetes—2023. Diabetes Care. 2022;46:S19–S40. doi:10.2337/dc23-S002
  • International Diabetes Federation. IDF diabetes atlas 2021 | IDF Diabetes Atlas [Internet]. 2022. Available from: https://diabetesatlas.org/atlas/tenth-edition/.
  • Papachristoforou E, Lambadiari V, Maratou E, et al. Association of glycemic indices (hyperglycemia, glucose variability, and hypoglycemia) with oxidative stress and diabetic complications. J Diabetes Res. 2020;2020:7489795. doi:10.1155/2020/7489795.
  • Kang Q, Yang C. Oxidative stress and diabetic retinopathy: molecular mechanisms: pathogenetic role and therapeutic implications. Redox Biol. 2020;37:101799. doi:10.1016/j.redox.2020.101799.
  • Darenskaya MA, Kolesnikova LI, Kolesnikov SI. Oxidative stress: pathogenetic role in diabetes mellitus and its complications and therapeutic approaches to correction. Bull Exp Biol Med. 2021;171:136–149. doi:10.47056/0365-9615-2021-171-2-136-149
  • Kiss L, Szabó C. The pathogenesis of diabetic complications: the role of DNA injury and poly(ADP-ribose) polymerase activation in peroxynitrite-mediated cytotoxicity. Mem Inst Oswaldo Cruz. 2005;100(Suppl 1):29–37. doi:10.1590/S0074-02762005000900007
  • Khalid M, Petroianu G, Adem A. Advanced glycation end products and diabetes mellitus: mechanisms and perspectives. Biomolecules. 2022;12(4):542. doi:10.3390/biom12040542.
  • Baghaie L, Bunsick DA, Szewczuk MR. Insulin receptor signaling in health and disease. Biomolecules. 2023;13:807–807. doi:10.3390/biom13050807
  • Howes KA, Liu Y, Dunaief JL, et al. Receptor for advanced glycation end products and age-related macular degeneration. Invest Ophthalmol Vis Sci. 2004;45:3713–3720. doi:10.1167/iovs.04-0404
  • Moldogazieva NT, Mokhosoev IM, Mel’Nikova TI, et al. Oxidative stress and advanced lipoxidation and glycation end products (ALEs and AGEs) in aging and age-related diseases. Oxid Med Cell Longev. 2019;2019:3085756. doi:10.1155/2019/3085756.
  • Lorenzi M. The polyol pathway as a mechanism for diabetic retinopathy: attractive, elusive, and resilient. Exp Diabetes Res. 2007;2007:61038. doi:10.1155/2007/61038.
  • Cho S-J, Roman G, Yeboah F, et al. The road to advanced glycation end products: a mechanistic perspective. Curr Med Chem. 2007;14:1653–1671. doi:10.2174/092986707780830989
  • Inoguchi T, Li P, Umeda F, et al. High glucose level and free fatty acid stimulate reactive oxygen species production through protein kinase C–dependent activation of NAD(P)H oxidase in cultured vascular cells. Diabetes. 2000;49:1939–1945. doi:10.2337/diabetes.49.11.1939
  • Faramoushi M, Amir Sasan R, Sari Sarraf V, et al. Cardiac fibrosis and down regulation of GLUT4 in experimental diabetic cardiomyopathy are ameliorated by chronic exposures to intermittent altitude. J Cardiovasc Thorac Res. 2016;8:26–33. doi:10.15171/jcvtr.2016.05
  • Pacher P, Szabó C. Role of poly(ADP-ribose) polymerase-1 activation in the pathogenesis of diabetic complications: endothelial dysfunction, as a common underlying theme. Antioxid Redox Signal. 2005;7:1568–1580. doi:10.1089/ars.2005.7.1568
  • Halim M, Halim A. The effects of inflammation, aging and oxidative stress on the pathogenesis of diabetes mellitus (type 2 diabetes). Diabetes Metab Syndr Clin Res Rev. 2019;13:1165–1172. doi:10.1016/j.dsx.2019.01.040
  • Luc K, Schramm-Luc A, Guzik TJ, et al. Oxidative stress and inflammatory markers in prediabetes and diabetes. J Physiol Pharmacol Off J Pol Physiol Soc. 2019;70(6):809–824. doi:10.26402/jpp.2019.6.01.
  • Krogh-Madsen R, Møller K, Dela F, et al. Effect of hyperglycemia and hyperinsulinemia on the response of IL-6, TNF-alpha, and FFAs to low-dose endotoxemia in humans. Am J Physiol Endocrinol Metab. 2004;286(5):E766–72. doi:10.1152/ajpendo.00468.2003.
  • Akbari M, Hassan-Zadeh V. IL-6 signalling pathways and the development of type 2 diabetes. Inflammopharmacology. 2018;26:685–698. doi:10.1007/s10787-018-0458-0
  • Yaribeygi H, Sathyapalan T, Atkin SL, et al. Molecular mechanisms linking oxidative stress and diabetes mellitus. Oxid Med Cell Longev. 2020;2020:8609213. doi:10.1155/2020/8609213
  • Zhang P, Li T, Wu X, et al. Oxidative stress and diabetes: antioxidative strategies. Front Med. 2020;14:583–600. doi:10.1007/s11684-019-0729-1
  • Fonseca SG, Gromada J, Urano F. Endoplasmic reticulum stress and pancreatic β-cell death. Trends Endocrinol Metab TEM. 2011;22:266–274.
  • Torres-Castro I, Arroyo-Camarena ÚD, Martínez-Reyes CP, et al. Human monocytes and macrophages undergo M1-type inflammatory polarization in response to high levels of glucose. Immunol Lett. 2016;176:81–89. doi:10.1016/j.imlet.2016.06.001
  • Alexopoulos AS, Kahkoska AR, Pate V, et al. Deintensification of treatment with sulfonylurea and insulin after severe hypoglycemia among older adults with diabetes. JAMA Netw Open. 2021;4(11):e2132215. doi:10.1001/jamanetworkopen.2021.32215.
  • Nam YH, Brensinger CM, Bilker WB, et al. Serious hypoglycemia and use of warfarin in combination with sulfonylureas or metformin. Clin Pharmacol Ther. 2019;105:210–218. doi:10.1002/cpt.1146
  • Wang G, Wang Y, Yang Q, et al. Metformin prevents methylglyoxal-induced apoptosis by suppressing oxidative stress in vitro and in vivo. Cell Death Dis. 2022;13(1):29. doi:10.1038/s41419-021-04478-x.
  • Katila N, Bhurtel S, Park PH, et al. Metformin attenuates rotenone-induced oxidative stress and mitochondrial damage via the AKT/Nrf2 pathway. Neurochem Int. 2021;148:105120. doi:10.1016/j.neuint.2021.105120.
  • Zhao X, Liu L, Jiang Y, et al. Protective effect of metformin against hydrogen peroxide-induced oxidative damage in human retinal pigment epithelial (RPE) cells by enhancing autophagy through activation of AMPK pathway. Oxid Med Cell Longev. 2020;2020:2524174. doi:10.1155/2020/2524174.
  • Padki MM, Stambler I. Encycl Gerontol Popul Aging. Targeting Aging with Metformin (TAME). Springer, Cham; 2021. p. 4098–4910. doi:10.1007/978-3-030-22009-9_400.
  • Mohammed I, Hollenberg MD, Ding H, et al. A critical review of the evidence that metformin is a putative anti-aging drug that enhances healthspan and extends lifespan. Front Endocrinol. 2021;12:718942.
  • Apostolova N, Iannantuoni F, Gruevska A, et al. Mechanisms of action of metformin in type 2 diabetes: effects on mitochondria and leukocyte-endothelium interactions. Redox Biol. 2020;34:101517. 10.1016/j.redox.2020.101517.
  • Zenebe Y, Akele B, W/Selassie M, et al. Prevalence and determinants of depression among old age: a systematic review and meta-analysis. Ann Gen Psychiatry. 2021;20:55. doi:10.1186/s12991-021-00375-x
  • Inoue K, Beekley J, Goto A, et al. Depression and cardiovascular disease events among patients with type 2 diabetes: A systematic review and meta-analysis with bias analysis. J Diabetes Complications. 2020;34:107710. doi:10.1016/j.jdiacomp.2020.107710
  • American Psychiatric Association. Practice guideline for the treatment of patients with major depressive disorder. Arlington (VA): American Psychiatric Association; 2010.
  • Tedeschini E, Levkovitz Y, Iovieno N, et al. Efficacy of antidepressants for late-life depression: a meta-analysis and meta-regression of placebo-controlled randomized trials. J Clin Psychiatry. 2011;72:1660–1668. doi:10.4088/JCP.10r06531
  • Calvi A, Fischetti I, Verzicco I, et al. Antidepressant drugs effects on blood pressure. Front Cardiovasc Med. 2021;8:704281. doi:10.3389/fcvm.2021.704281
  • Mercurio M, de Filippis R, Spina G, et al. The use of antidepressants is linked to bone loss: a systematic review and metanalysis. Orthop Rev. 2022;14:38564.
  • Miguel C, Albuquerque E. Drug interaction in psycho-oncology: antidepressants and antineoplastics. Pharmacology. 2011;88:333–339. doi:10.1159/000334738
  • Byeon E, Park JC, Hagiwara A, et al. Two antidepressants fluoxetine and sertraline cause growth retardation and oxidative stress in the marine rotifer Brachionus koreanus. Aquat Toxicol Amst Neth. 2020;218:105337. doi:10.1016/j.aquatox.2019.105337
  • Khanzode SD, Dakhale GN, Khanzode SS, et al. Oxidative damage and major depression: the potential antioxidant action of selective serotonin re-uptake inhibitors. Redox Rep Commun Free Radic Res. 2003;8:365–370.
  • Kovesdy CP. Epidemiology of chronic kidney disease: an update 2022. Kidney Int Suppl. 2022;12:7–11. doi:10.1016/j.kisu.2021.11.003
  • Bello AK, Okpechi IG, Osman MA, et al. Epidemiology of haemodialysis outcomes. Nat Rev Nephrol. 2022;18:378–395. doi:10.1038/s41581-022-00542-7
  • Canaud B, Tong L, Tentori F, et al. Clinical practices and outcomes in elderly hemodialysis patients: results from the dialysis outcomes and practice patterns study (DOPPS). Clin J Am Soc Nephrol CJASN. 2011;6:1651–1662. doi:10.2215/CJN.03530410
  • Vodošek Hojs N, Bevc S, Ekart R, et al. Oxidative stress markers in chronic kidney disease with emphasis on diabetic nephropathy. Antioxidants. 2020;9:925. doi:10.3390/antiox9100925
  • Irazabal MV, Torres VE. Reactive oxygen species and redox signaling in chronic kidney disease. Cells. 2020;9:1342. doi:10.3390/cells9061342
  • Handelman GJ, Walter MF, Adhikarla R, et al. Elevated plasma F2-isoprostanes in patients on long-term hemodialysis. Kidney Int. 2001;59:1960–1966. doi:10.1046/j.1523-1755.2001.0590051960.x
  • Tarng DC, Huang TP, Wei YH, et al. 8-hydroxy-2’-deoxyguanosine of leukocyte DNA as a marker of oxidative stress in chronic hemodialysis patients. Am J Kidney Dis Off J Natl Kidney Found. 2000;36:934–944. doi:10.1053/ajkd.2000.19086
  • Shimoike T, Inoguchi T, Umeda F, et al. The meaning of serum levels of advanced glycosylation end products in diabetic nephropathy. Metabolism. 2000;49:1030–1035. doi:10.1053/meta.2000.7738
  • Dozio E, Caldiroli L, Molinari P, et al. Accelerated AGEing: the impact of advanced glycation end products on the prognosis of chronic kidney disease. Antioxidants. 2023;12:584. doi:10.3390/antiox12030584
  • Kato H, Watanabe H, Imafuku T, et al. Advanced oxidation protein products contribute to chronic kidney disease-induced muscle atrophy by inducing oxidative stress via CD36/NADPH oxidase pathway. J Cachexia Sarcopenia Muscle. 2021;12:1832–1847. doi:10.1002/jcsm.12786
  • Ravarotto V, Bertoldi G, Innico G, et al. The pivotal role of oxidative stress in the pathophysiology of cardiovascular-renal remodeling in kidney disease. Antioxidants. 2021;10:1041. doi:10.3390/antiox10071041
  • Satou R, Cypress MW, Woods TC, et al. Blockade of sodium-glucose cotransporter 2 suppresses high glucose-induced angiotensinogen augmentation in renal proximal tubular cells. Am J Physiol - Ren Physiol. 2020;318:F67–F75. doi:10.1152/ajprenal.00402.2019
  • Zaibi N, Li P, Xu S-Z. Protective effects of dapagliflozin against oxidative stress-induced cell injury in human proximal tubular cells. PLoS One. 2021;16:e0247234. doi:10.1371/journal.pone.0247234
  • Hasan R, Lasker S, Hasan A, et al. Canagliflozin ameliorates renal oxidative stress and inflammation by stimulating AMPK–Akt–eNOS pathway in the isoprenaline-induced oxidative stress model. Sci Rep. 2020;10:14659. doi:10.1038/s41598-020-71599-2
  • Das NA, Carpenter AJ, Belenchia A, et al. Empagliflozin reduces high glucose-induced oxidative stress and miR-21-dependent TRAF3IP2 induction and RECK suppression, and inhibits human renal proximal tubular epithelial cell migration and epithelial-to-mesenchymal transition. Cell Signal. 2020;68:109506. doi:10.1016/j.cellsig.2019.109506
  • Fang R, Chen J, Long J, et al. Empagliflozin improves kidney senescence induced by d-galactose by reducing sirt1-mediated oxidative stress. Biogerontology. 2023;24:771–782. doi:10.1007/s10522-023-10038-x
  • Semo D, Obergassel J, Dorenkamp M, et al. The sodium-glucose co-transporter 2 (SGLT2) inhibitor empagliflozin reverses hyperglycemia-induced monocyte and endothelial dysfunction primarily through glucose transport-independent but redox-dependent mechanisms. J Clin Med. 2023;12:1356. doi:10.3390/jcm12041356
  • Plantinga Y, Ghiadoni L, Magagna A, et al. Supplementation with vitamins C and E improves arterial stiffness and endothelial function in essential hypertensive patients. Am J Hypertens. 2007;20:392–397. doi:10.1016/j.amjhyper.2006.09.021
  • Tessier DM, Khalil A, Trottier L, et al. Effects of vitamin C supplementation on antioxidants and lipid peroxidation markers in elderly subjects with type 2 diabetes. Arch Gerontol Geriatr. 2009;48:67–72. doi:10.1016/j.archger.2007.10.005
  • Mazloom Z, Hejazi N, Dabbaghmanesh M-H, et al. Effect of vitamin C supplementation on postprandial oxidative stress and lipid profile in type 2 diabetic patients. Pak J Biol Sci PJBS. 2011;14:900–904. doi:10.3923/pjbs.2011.900.904
  • Ulker S, McKeown PP, Bayraktutan U. Vitamins reverse endothelial dysfunction through regulation of eNOS and NAD(P)H oxidase activities. Hypertens Dallas Tex 1979. 2003;41:534–539.
  • Boonthongkaew C, Tong-Un T, Kanpetta Y, et al. Vitamin C supplementation improves blood pressure and oxidative stress after acute exercise in patients with poorly controlled type 2 diabetes mellitus: a randomized,: placebo-controlled, cross-over study. Chin J Physiol. 2021;64:16–23. doi:10.4103/cjp.cjp_95_20
  • Rodrigo R, Prat H, Passalacqua W, et al. Decrease in oxidative stress through supplementation of vitamins C and E is associated with a reduction in blood pressure in patients with essential hypertension. Clin Sci Lond Engl 1979. 2008;114:625–634.
  • Hajjar IM, George V, Sasse EA, et al. A randomized: double-blind, controlled trial of vitamin C in the management of hypertension and lipids. Am J Ther. 2002;9:289–293. doi:10.1097/00045391-200207000-00005
  • Guan Y, Dai P, Wang H. Effects of vitamin C supplementation on essential hypertension: a systematic review and meta-analysis. Medicine (Baltimore. 2020;99:e19274. doi:10.1097/MD.0000000000019274
  • Sato K, Dohi Y, Kojima M, et al. Effects of ascorbic acid on ambulatory blood pressure in elderly patients with refractory hypertension. Arzneimittelforschung. 2006;56:535–540.
  • Mason SA, Della Gatta PA, Snow RJ, et al. Ascorbic acid supplementation improves skeletal muscle oxidative stress and insulin sensitivity in people with type 2 diabetes: findings of a randomized controlled study. Free Radic Biol Med. 2016;93:227–238. doi:10.1016/j.freeradbiomed.2016.01.006
  • El-Aal AA, El-Ghffar EAA, Ghali AA, et al. The effect of vitamin C and/or E supplementations on type 2 diabetic adult males under metformin treatment: a single-blinded randomized controlled clinical trial. Diabetes Metab Syndr. 2018;12:483–489. doi:10.1016/j.dsx.2018.03.013
  • Mason SA, Rasmussen B, van Loon LJC, et al. Ascorbic acid supplementation improves postprandial glycaemic control and blood pressure in individuals with type 2 diabetes: findings of a randomized cross-over trial. Diabetes Obes Metab. 2019;21:674–682. doi:10.1111/dom.13571
  • Ellulu MS, Rahmat A, Patimah I, et al. Effect of vitamin C on inflammation and metabolic markers in hypertensive and/or diabetic obese adults: a randomized controlled trial. Drug Des Devel Ther. 2015;9:3405–3412. doi:10.2147/DDDT.S83144
  • Fraga CG, Croft KD, Kennedy DO, et al. The effects of polyphenols and other bioactives on human health. Food Funct. 2019;10:514–528. doi:10.1039/C8FO01997E
  • Serban M-C, Sahebkar A, Zanchetti A, et al. Effects of quercetin on blood pressure: a systematic review and meta-analysis of randomized controlled trials. J Am Heart Assoc. 2016;5:e002713.
  • Brüll V, Burak C, Stoffel-Wagner B, et al. Effects of a quercetin-rich onion skin extract on 24 h ambulatory blood pressure and endothelial function in overweight-to-obese patients with (pre-)hypertension: a randomised double-blinded placebo-controlled cross-over trial. Br J Nutr. 2015;114:1263–1277. doi:10.1017/S0007114515002950
  • Egert S, Bosy-Westphal A, Seiberl J, et al. Quercetin reduces systolic blood pressure and plasma oxidised low-density lipoprotein concentrations in overweight subjects with a high-cardiovascular disease risk phenotype: a double-blinded, placebo-controlled cross-over study. Br J Nutr. 2009;102:1065–1074. doi:10.1017/S0007114509359127
  • Breuss JM, Atanasov AG, Uhrin P. Resveratrol and its effects on the vascular system. Int J Mol Sci. 2019;20:1523. doi:10.3390/ijms20071523
  • Sweeney M, Burns G, Sturgeon N, et al. The effects of berry polyphenols on the gut microbiota and blood pressure: a systematic review of randomized clinical trials in humans. Nutrients. 2022;14:2263. doi:10.3390/nu14112263
  • Vetrani C, Vitale M, Bozzetto L, et al. Association between different dietary polyphenol subclasses and the improvement in cardiometabolic risk factors: evidence from a randomized controlled clinical trial. Acta Diabetol. 2018;55:149–153. doi:10.1007/s00592-017-1075-x
  • Mokgalaboni K, Ntamo Y, Ziqubu K, et al. Curcumin supplementation improves biomarkers of oxidative stress and inflammation in conditions of obesity, type 2 diabetes and NAFLD: updating the status of clinical evidence. Food Funct. 2021;12:12235–12249. doi:10.1039/D1FO02696H
  • Witte AV, Kerti L, Margulies DS, et al. Effects of resveratrol on memory performance: hippocampal functional connectivity, and glucose metabolism in healthy older adults. J Neurosci. 2014;34:7862–7870. doi:10.1523/JNEUROSCI.0385-14.2014
  • Mahjabeen W, Khan DA, Mirza SA. Role of resveratrol supplementation in regulation of glucose hemostasis: inflammation and oxidative stress in patients with diabetes mellitus type 2: A randomized, placebo-controlled trial. Complement Ther Med. 2022;66:102819. doi:10.1016/j.ctim.2022.102819
  • Du C, Smith A, Avalos M, et al. Blueberries improve pain, gait performance, and inflammation in individuals with symptomatic knee osteoarthritis. Nutrients. 2019;11:290. doi:10.3390/nu11020290
  • Hussain SA, Marouf BH, Ali ZS, et al. Efficacy and safety of co-administration of resveratrol with meloxicam in patients with knee osteoarthritis: a pilot interventional study. Clin Interv Aging. 2018;13:1621–1630. doi:10.2147/CIA.S172758
  • Ávila-Gálvez MÁ, González-Sarrías A, Martínez-Díaz F, et al. Disposition of dietary polyphenols in breast cancer patients’ tumors, and their associated anticancer activity: the particular case of curcumin. Mol Nutr Food Res. 2021;65:2100163. doi:10.1002/mnfr.202100163
  • Huhn S, Beyer F, Zhang R, et al. Effects of resveratrol on memory performance, hippocampus connectivity and microstructure in older adults - a randomized controlled trial. NeuroImage. 2018;174:177–190. doi:10.1016/j.neuroimage.2018.03.023
  • Moussa C, Hebron M, Huang X, et al. Resveratrol regulates neuro-inflammation and induces adaptive immunity in Alzheimer’s disease. J Neuroinflammation. 2017;14:1. doi:10.1186/s12974-016-0779-0
  • Baum L, Lam CWK, Cheung SK-K, et al. Six-month randomized, placebo-controlled, double-blind, pilot clinical trial of curcumin in patients with Alzheimer disease. J Clin Psychopharmacol. 2008;28:110–113. doi:10.1097/jcp.0b013e318160862c
  • Kumar P, Liu C, Suliburk J, et al. Supplementing glycine and N-acetylcysteine (GlyNAC) in older adults improves glutathione deficiency, oxidative stress, mitochondrial dysfunction, inflammation, physical function, and aging hallmarks: a randomized clinical trial. J Gerontol A Biol Sci Med Sci. 2023;78:75–89. doi:10.1093/gerona/glac135
  • Hashemi G, Mirjalili M, Basiri Z, et al. A pilot study to evaluate the effects of oral N-acetyl cysteine on inflammatory and oxidative stress biomarkers in rheumatoid arthritis. Curr Rheumatol Rev. 2019;15:246–253. doi:10.2174/1573403X14666180926100811
  • Esalatmanesh K, Jamali A, Esalatmanesh R, et al. Effects of N-acetylcysteine supplementation on disease activity: oxidative stress, and inflammatory and metabolic parameters in rheumatoid arthritis patients: a randomized double-blind placebo-controlled trial. Amino Acids. 2022;54:433–440. doi:10.1007/s00726-022-03134-8
  • Batooei M, Tahamoli-Roudsari A, Basiri Z, et al. Evaluating the effect of oral N-acetylcysteine as an adjuvant treatment on clinical outcomes of patients with rheumatoid arthritis: a randomized, double blind clinical trial. Rev Recent Clin Trials. 2018;13:132–138. doi:10.2174/1574887113666180307151937
  • Adair JC, Knoefel JE, Morgan N. Controlled trial of N-acetylcysteine for patients with probable Alzheimer’s disease. Neurology. 2001;57:1515–1517. doi:10.1212/WNL.57.8.1515
  • Remington R, Chan A, Paskavitz J, et al. Efficacy of a vitamin/nutriceutical formulation for moderate-stage to later-stage Alzheimer’s disease: a placebo-controlled pilot study. Am J Alzheimers Dis Dementias. 2009;24:27–33. doi:10.1177/1533317508325094
  • Chan A, Paskavitz J, Remington R, et al. Efficacy of a vitamin/nutriceutical formulation for early-stage Alzheimer’s disease: a 1-year, open-label pilot study with an 16-month caregiver extension. Am J Alzheimers Dis Dementias. 2009;23:571–585. doi:10.1177/1533317508325093
  • Remington R, Bechtel C, Larsen D, et al. A phase II randomized clinical trial of a nutritional formulation for cognition and mood in Alzheimer’s disease. J Alzheimers Dis JAD. 2015;45:395–405. doi:10.3233/JAD-142499
  • Skvarc DR, Dean OM, Byrne LK, et al. The effect of N-acetylcysteine (NAC) on human cognition – a systematic review. Neurosci Biobehav Rev. 2017;78:44–56. doi:10.1016/j.neubiorev.2017.04.013
  • Fasipe B, Faria A, Laher I. Potential for novel therapeutic uses of alpha lipoic acid. Curr Med Chem. 2023;30:3942–3954. doi:10.2174/0929867329666221006115329
  • Rezaei Zonooz S, Hasani M, Morvaridzadeh M, et al. Effect of alpha-lipoic acid on oxidative stress parameters: a systematic review and meta-analysis. J Funct Foods. 2021;87:104774. doi:10.1016/j.jff.2021.104774
  • Jeffrey S, Isaac Samraj P, Sundara Raj B. Therapeutic benefits of alpha-lipoic acid supplementation in diabetes mellitus: a narrative review. J Diet Suppl. 2022;19:566–586. doi:10.1080/19390211.2021.2020387
  • Derosa G, D’Angelo A, Romano D, et al. A clinical trial about a food supplement containing α-lipoic acid on oxidative stress markers in type 2 diabetic patients. Int J Mol Sci. 2016;17:1802. doi:10.3390/ijms17111802
  • Mendoza-Núñez VM, García-Martínez BI, Rosado-Pérez J, et al. The effect of 600 mg alpha-lipoic acid supplementation on oxidative stress, inflammation, and RAGE in older adults with type 2 diabetes mellitus. Oxid Med Cell Longev. 2019;2019:3276958.
  • Gianturco V, Bellomo A, D’ottavio E, et al. Impact of therapy with α-lipoic acid (ALA) on the oxidative stress in the controlled niddm: a possible preventive way against the organ dysfunction? Arch Gerontol Geriatr. 2009;49:129–133. doi:10.1016/j.archger.2009.09.022
  • Zhao L, Hu F-X. α-Lipoic acid treatment of aged type 2 diabetes mellitus complicated with acute cerebral infarction. Eur Rev Med Pharmacol Sci. 2014;18:3715–3719.
  • Porasuphatana S, Suddee S, Nartnampong A, et al. Glycemic and oxidative status of patients with type 2 diabetes mellitus following oral administration of alpha-lipoic acid: a randomized double-blinded placebo-controlled study. Asia Pac J Clin Nutr. 2012;21:12–21.
  • Khabbazi T, Mahdavi R, Safa J, et al. Effects of alpha-lipoic acid supplementation on inflammation: oxidative stress, and serum lipid profile levels in patients with end-stage renal disease on hemodialysis. J Ren Nutr. 2012;22:244–250. doi:10.1053/j.jrn.2011.06.005
  • Ramos LF, Kane J, McMonagle E, et al. Effects of combination tocopherols and alpha lipoic acid therapy on oxidative stress and inflammatory biomarkers in chronic kidney disease. J Ren Nutr Off J Counc Ren Nutr Natl Kidney Found. 2011;21:211–218.
  • Himmelfarb J, Ikizler TA, Ellis C, et al. Provision of antioxidant therapy in hemodialysis (PATH): a randomized clinical trial. J Am Soc Nephrol JASN. 2014;25:623–633. doi:10.1681/ASN.2013050545
  • Mahdavi R, Khabbazi T, Safa J. Alpha lipoic acid supplementation improved antioxidant enzyme activities in hemodialysis patients. Int J Vitam Nutr Res Int Z Vitam- Ernahrungsforschung J Int Vitaminol Nutr. 2019;89:161–167. doi:10.1024/0300-9831/a000552
  • Abdel Hamid DZ, Nienaa YA, Mostafa TM. Alpha-lipoic acid improved anemia: erythropoietin resistance, maintained glycemic control, and reduced cardiovascular risk in diabetic patients on hemodialysis: a multi-center prospective randomized controlled study. Eur Rev Med Pharmacol Sci. 2022;26:2313–2329.
  • Díaz-Casado ME, Quiles JL, Barriocanal-Casado E, et al. The paradox of coenzyme Q10 in aging. Nutrients. 2019;11:2221. doi:10.3390/nu11092221
  • Hargreaves I, Heaton RA, Mantle D. Disorders of human coenzyme Q10 metabolism: an overview. Int J Mol Sci. 2020;21:6695. doi:10.3390/ijms21186695
  • Hargreaves IP, Mantle D. Coenzyme Q10 supplementation in fibrosis and aging. Adv Exp Med Biol. 2019;1178:103–112. doi:10.1007/978-3-030-25650-0_6
  • Lee B-J, Huang Y-C, Chen S-J, et al. Coenzyme Q10 supplementation reduces oxidative stress and increases antioxidant enzyme activity in patients with coronary artery disease. Nutrition. 2012;28:250–255. doi:10.1016/j.nut.2011.06.004
  • Yeung CK, Billings FT, Claessens AJ, et al. Coenzyme Q10 dose-escalation study in hemodialysis patients: safety, tolerability, and effect on oxidative stress. BMC Nephrol. 2015;16:183. doi:10.1186/s12882-015-0178-2
  • Rivara MB, Yeung CK, Robinson-Cohen C, et al. Effect of coenzyme Q10 on biomarkers of oxidative stress and cardiac function in hemodialysis patients: the CoQ10 biomarker trial. Am J Kidney Dis Off J Natl Kidney Found. 2017;69:389–399. doi:10.1053/j.ajkd.2016.08.041
  • The Heart Outcomes Prevention Evaluation Study Investigators. Vitamin E supplementation and cardiovascular events in high-risk patients. N Engl J Med. 2000;342:154–160. doi:10.1056/NEJM200001203420302
  • Lee I-M, Cook NR, Gaziano JM, et al. Vitamin E in the primary prevention of cardiovascular disease and cancer: the Women’s Health Study: a randomized controlled trial. JAMA. 2005;294:56–65. doi:10.1001/jama.294.1.56
  • Nascimento MM, Suliman ME, Silva M, et al. Effect of oral N-acetylcysteine treatment on plasma inflammatory and oxidative stress markers in peritoneal dialysis patients: a placebo-controlled study. Perit Dial Int J Int Soc Perit Dial. 2010;30:336–342. doi:10.3747/pdi.2009.00073
  • Renke M, Tylicki L, Rutkowski P, et al. The effect of N-acetylcysteine on blood pressure and markers of cardiovascular risk in non-diabetic patients with chronic kidney disease: a placebo-controlled, randomized, cross-over study. Med Sci Monit Int Med J Exp Clin Res. 2010;16:PI13–PI18.
  • Zhang Y, Han P, Wu N, et al. Amelioration of lipid abnormalities by α-lipoic acid through antioxidative and anti-inflammatory effects. Obesity. 2011;19:1647–1653. doi:10.1038/oby.2011.121
  • Bobe G, Michels AJ, Zhang W-J, et al. A randomized controlled trial of long-term (R)-α-lipoic acid supplementation promotes weight loss in overweight or obese adults without altering baseline elevated plasma triglyceride concentrations. J Nutr. 2020;150:2336–2345. doi:10.1093/jn/nxaa203
  • Ahmadi A, Mazooji N, Roozbeh J, et al. Effect of alpha-lipoic acid and vitamin E supplementation on oxidative stress,: inflammation, and malnutrition in hemodialysis patients. Iran J Kidney Dis. 2013;7:461–467.
  • Pérez-Torres I, Guarner-Lans V, Rubio-Ruiz ME. Reductive stress in inflammation-associated diseases and the pro-oxidant effect of antioxidant agents. Int J Mol Sci. 2017;18:2098. doi:10.3390/ijms18102098