Next Article in Journal
Exploring Synergistic Interactions between Natural Compounds and Conventional Chemotherapeutic Drugs in Preclinical Models of Lung Cancer
Previous Article in Journal
Cistus, Acacia, and Lemon verbena Valorization through Response Surface Methodology: Optimization Studies and Potential Application in the Pharmaceutical and Nutraceutical Industries
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Emerging Vistas for the Nutraceutical Withania somnifera in Inflammaging

1
PhytoVeda Pvt. Ltd., Mumbai 400 022, India
2
Viridis Biopharma Pvt. Ltd., Mumbai 400 022, India
3
College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2024, 17(5), 597; https://doi.org/10.3390/ph17050597
Submission received: 8 April 2024 / Revised: 26 April 2024 / Accepted: 3 May 2024 / Published: 7 May 2024
(This article belongs to the Section Natural Products)

Abstract

:
Inflammaging, a coexistence of inflammation and aging, is a persistent, systemic, low-grade inflammation seen in the geriatric population. Various natural compounds have been greatly explored for their potential role in preventing and treating inflammaging. Withania somnifera has been used for thousands of years in traditional medicine as a nutraceutical for its numerous health benefits including regenerative and adaptogenic effects. Recent preclinical and clinical studies on the role of Withania somnifera and its active compounds in treating aging, inflammation, and oxidative stress have shown promise for its use in healthy aging. We discuss the chemistry of Withania somnifera, the etiology of inflammaging and the protective role(s) of Withania somnifera in inflammaging in key organ systems including brain, lung, kidney, and liver as well as the mechanistic underpinning of these effects. Furthermore, we elucidate the beneficial effects of Withania somnifera in oxidative stress/DNA damage, immunomodulation, COVID-19, and the microbiome. We also delineate a putative protein–protein interaction network of key biomarkers modulated by Withania somnifera in inflammaging. In addition, we review the safety/potential toxicity of Withania somnifera as well as global clinical trials on Withania somnifera. Taken together, this is a synthetic review on the beneficial effects of Withania somnifera in inflammaging and highlights the potential of Withania somnifera in improving the health-related quality of life (HRQoL) in the aging population worldwide.

1. Introduction

Aging is a time-associated natural and complex phenomenon leading to deterioration of physiological functions. A prominent aspect of aging involves control and gradual increase in proinflammatory status, termed as inflammaging [1]. Inflammaging is a chronic, systemic, low-grade inflammation resulting from high levels of inflammation that is primarily observed in elderly patients [2,3]. The term inflammaging was first given by Franceschi et al. [4] and involves numerous molecular and cellular mechanisms, such as cellular senescence, immunosenescence, mitochondrial, and ubiquitin-proteasome system dysfunction, activation of inflammasome, and DNA damage response [4,5]. Inflammaging can also contribute to the progression of various disease including cardiovascular disease [6], neurodegenerative disorders [7,8], diabetic nephropathy and diabetic retinopathy in diabetes [9,10,11,12], and cancer [13].
Withania somnifera or Ashwagandha, also referred to as winter cherry or Indian ginseng, which belongs to the Solanaceae family, is a well-known herb having numerous health benefits [14,15]. It is generally found in arid regions of South Asia, Central Asia and Africa; moreover, it is commonly used in Ayurveda (the Traditional Medicinal System of India) [16,17]. Withania somnifera has been used for thousands of years as a ‘Rasayana’ (Rasayana is a holistic therapy that helps by suppressing the aging process by developing positive physical and mental health, boosting the immune system and maintaining youthfulness [18]), along with providing numerous health benefits [19]. Furthermore, Withania somnifera is referred to as ‘Avarada’ in Ayurvedic literature, which indicates regeneration or youthfulness [20]. It is also included among the medicinal plants in the World Health Organization’s (WHO) monographs [21]. Phytoconstituents in Withania somnifera are present throughout the plant such as roots, leaves, and berries; however, the root section of Withania somnifera has the highest concentration of preferred bioactives. Withanolides and Withaferins (steroidal lactones), with saponins and alkaloids being a few of the active biological constituents present in Withania somnifera [22]. Withania somnifera has various properties, such as modulation of the immune system [23], antioxidant [24], antiinflammation [25], cardiorespiratory endurance [26], neuroprotection [27], male infertility treatment [28], anticancer activity [29], and decreases cortisol levels, stress, anxiety, and blood pressure in chronic stress.
The exact underlying mechanism of inflammaging is not completely known. However, there are various factors and causes that may lead to inflammaging. Studies have reported that inflammaging develops due to several factors, including oxidative stress, immunosenescence, cell senescence, hormonal changes, chronic infections, psychological stress, and metabolic changes. Earlier studies have discussed phytoconstituents and dietary supplements, such as curcumin from turmeric, ellagic acid from pomegranate, and epigallocatechin-3-gallate (EGCG) from green tea [30]; Moringa oleifera [31]; sandalwood oil [32]; sulforaphane [33]; and vitamin K2-7 [34,35], along with their numerous health benefits. Furthermore, the roles of various phytocompounds such as organosulfur compounds [36], EGCG [37], curcumin [38], flavonoids [39], soy isoflavone [40], and γ-tocopherol [41] in the modulation of gene expression for cancer have been reported. In addition, the role of phytocompounds when administered in combination with, for example, sulforaphane and EGCG for colon and prostate cancer [42,43] and curcumin and sulforaphane for cancer chemoprevention has been elucidated earlier [44]. In this review, we discuss the etiology of inflammaging and the protective role(s) of Withania somnifera in various organ systems. In addition, we elucidate the mechanistic basis for the health-beneficial effects of Withania somnifera using a systems pharmacology approach. Finally, we discuss clinical trials on Withania somnifera to collate a synthetic review on Withania somnifera in inflammaging.

2. Chemistry of Withania somnifera

Withania somnifera contains numerous active compounds, such as steroidal lactones (withanolides), alkaloids, and sitoindosides. Steroidal lactones, including withanolides and withanosides, are major phytoconstituents of Withania somnifera. Withania somnifera also contains sitoindosides, which are derivatives of withanolides (glucose at carbon 27 of withanolides) [45]. A study by Ghosal et al. reported that sitoindosides have CNS and immunomodulatory effects [46]. Akhoon et al. [20] have demonstrated improvement in life expectancy using Withanolide A in Caenorhabditis elegans. Various alkaloids are present in Withania somnifera, including anahygrine, cuseohygrine, anaferine, and isopellentierine. Choline, which is an important nutrient required in proper functioning of various physiological processes, is present in Withania somnifera [47,48]. Withania somnifera also contains tannins [49], which are polyphenolic compounds having various uses, such as antioxidant, antiinflammatory, anticancer, and neuroprotective. Moreover, Withania somnifera also contains carbohydrates, fatty acids, and amino acids that play a significant role in maintaining daily complex biochemical processes [50]. The chemical structures of Withanolide A, Withaferin, 12-Deoxy-withastramonolide, Withanoside V, Withanone, Withanoside IV, Withanolide B, 27-Hydroxywithanone, Withanoside VI, and Physagulin-d have been illustrated in Figure 1.

3. Etiology of Inflammaging

The precise etiology and underlying mechanisms involved in development and progression of inflammaging is currently unknown. Recent studies suggest a complex process that when understood can provide strategies for diagnosis and therapy of inflammaging associated with numerous factors, including cellular senescence, immunosenescence, oxidative stress, mitochondrial dysfunction, epigenetic changes, inflammasome activation, metabolic changes, microbiome alterations, DNA damage, microbiome alterations, genetic predispositions, lifestyle, and chronic diseases. Cellular senescence, a complex process driven by genetic, epigenetic, and environmental factors, affects many types of somatic cells, leading to progression of aging and chronic diseases. Senescent cells promotes proinflammatory and matrix-degenerating substances and further accumulation of senescent cells results in development of inflammaging [51]. Immunosenescence is a gradual process of deterioration of the immune system with increase in age. It contributes to the progression of inflammaging due to weakening of immune system that leads to various pathological conditions, such as dysregulation of immunomodulation, accumulation of senescent cells, failure in recognizing pathogens, and increase in autoimmune conditions [52].

4. Withania somnifera in Inflammaging

Inflammaging progression is associated with numerous factors, and hence, it is difficult to understand the complex process and to represent the role of Withania somnifera in mitigating it due to limited evidence. Here, we have discussed the role of Withania somnifera in inflammaging and the potential mechanisms involved in inflammaging (as illustrated in Figure 2). Grunz-Borgmann et al. [53] performed an experiment to study the role of Withania somnifera water extract in aging kidney with chronic inflammation using a rat proximal tubular cell line (NRK-52E). Monocyte chemotactic protein-1, also known as CCL2, belongs to the category of C-C chemokines and possesses properties such as chemoattractant for T lymphocytes, leukocytes, and natural killer cells (NK). CCL2 is overexpressed in patients with chronic kidney disease [54]. CCL5 belongs to the chemokine family that regulates T-cells, monocytes, eosinophils, and basophils in inflammation. Overexpression of CCL2 and CCL5 can play a key role in progression of fibrosis in chronic kidney disease. Grunz-Borgmann et al. demonstrated that Withania somnifera water extract suppressed the gene expression of CCL2 and CCL5 via TNF-α/LPS stimulation by reducing the activity of NF-kB.

4.1. Withania somnifera in Aging

The elasticity and strength of the human skin and body are maintained by collagen and elastin. In aged skin, release of MMP-1 (an enzyme that degrades collagen) and intracellular ROS production is induced by TNF-α. Lee et al. [55] isolated withagenin A diglucoside from methanolic extract of Withania somnifera and investigated its functions in TNF-α stimulated human dermal fibroblasts. It was observed that withagenin A diglucoside inhibits intracellular ROS production that leads to suppression in secretion of MMP-1 and collapsing collagen type 1. Withagenin A diglucoside demonstrated inhibition of phosphorylation of MAPK, Akt, c-JUN, NF-kB, and cyclooxygenase-2 (COX-2) expression. NF-kB is involved in cellular proliferation as well as inflammation via regulating gene expression [56]. Serine-threonine kinases, known as mitogen-activated protein kinases, or MAPKs, enhance intracellular signaling related to a variety of cellular processes, such as cell division, proliferation, survival, and transformation [57]. Akt stimulates mammalian cell survival and proliferation, helping in the development of tumors, and it has been demonstrated recently that Akt activity increases with cellular senescence and that primary cultivated human endothelial cells live longer when Akt is inhibited. Constitutive activation of Akt stimulates a p53/p21-dependent pathway that results in dysfunctional endothelial cells and senescence-like interruption of cell proliferation. Akt’s unique function in controlling cellular lifetime may be a factor in many human conditions [58]. The enzyme COX-2 in the prostaglandin synthesis pathway may have a role in elderly people by causing oxidative damage and various stress responses [59]. Also, it was observed that withagenin A diglucoside inhibited expression of pro-inflammatory cytokines IL-6 and IL-8, which demonstrated the potential use of withagenin A diglucoside in skin aging. Dysregulation of collagenous tissues occurs because of collagen cross-linking in aging and hyperglycemia conditions that specifically affects renal, cardiovascular, and retinal tissues, which contributes to severe illness or even death.
Babu et al. [60] demonstrated that Withania somnifera can significantly reduce glycation, AGE, and cross-linking compared to metformin. The ethanolic extract of Withania somnifera demonstrated greater efficacy than the root powder, suggesting that ethanolic extract contains more phytocompounds. Withanolide A promotes health, lowers age-related physiological changes, and increases lifespan. Akhoon et al. [20] showed that Withanolide A has various neuroprotective properties, including antiamyloidogenic actions, reduction of α-synuclein aggregation, and neuroprotection via regulation of neuronal mediators such as acetylcholine. Also, it was demonstrated that Withanolide A promotes stress tolerance and extends lifespan via the insulin/insulin-like growth factor signaling pathway. Numerous clinical ailments, including sarcopenia, are associated with aging. This is caused by a disparity in synthesis and breakdown of myofibrillary protein synthesis (particularly myosin heavy chain (MyHC) adult form), as well as a loss in muscle regenerative potential. Salvadori et al. [61] investigated the efficacy of WST formulation containing Withania somnifera, Silybum marianum, and Trigonella foenum-graecum against MyHC-II degradation for the protection of C2C12 myoblasts. It was observed that WST upregulates Akt (protein kinase B)-dependent protein synthesis as well as p38 MAPK (p38 mitogen-activated protein kinase)/myogenin-dependent myoblast differentiation. p38 MAPK found in mammals plays significant roles in muscle physiology [62]. WST also maintains trophism in both C2C12 and young myotubes, as well as restores sarcopenic myotube size, developmental myoblast fusion, and MyHC expression.

Withania somnifera in Healthy Aging

Forkhead box protein O3A (FOXO3A) and sirtuin 3 (SIRT3) are some of the important biomarkers that play key roles in longevity by regulating gene expression [63,64]. Pradhan et al. [65] performed an experiment to study the expression of FOXO3A and SIRT3 in healthy aging in humans (473 subjects) and HEK-293 cells. A significant decrease in FOXO3A and SIRT3 was observed with an increase in the age of the patients. Furthermore, in vitro studies performed in stress-induced HEK-293 cells treated with Withania somnifera showed upregulation of FOXO3A and SIRT3. Kuchewar et al. [66] demonstrated that oral administration of Withania somnifera (500 mg capsule twice a day) for 6 months showed significant decrease in malondialdehyde (MDA) and increase in superoxide dismutase (SOD) levels. Superoxide dismutase forms the front line of defense against reactive oxygen species-mediated injury; induction in SOD activity in human aging may be a compensatory response of the individual to increased oxidative stress [67]. Furthermore, it was also suggested that Withania somnifera can help in preventing premature aging. Nuclear factor erythroid 2-related factor 2 (Nrf2) provides cytoprotective action by modulating gene expression of numerous antioxidant enzymes via binding to the promotor region of antioxidant response element (ARE). However, it has been observed that with an increase in age, there is a decrease in activity of Nrf2 [68]. To activate the Nrf2 for promoting healthy aging, Hybertson et al. [69] used a combination of Rosmarinus officinalis, Withania somnifera, and Sophora japonica extract powder in the ratio 15:5:2 and treated human hepatocellular carcinoma HepG2 cells. The phytochemical combination of Rosmarinus officinalis, Withania somnifera, and Sophora japonica promoted the activation of NRF2, suggesting their potential role in treating aging.
Cabey et al. [70] studied the role of Withania somnifera water extract using an in vivo model (Drosophila melanogaster) and an in vitro model (HepG2 cells). It was reported that water extract of Withania somnifera showed significant increase in NRF2 levels in HepG2-ARE (HepG2 cells expressing antioxidant response element by the application of firefly luciferase gene) cells. Also, the sniffer Drosophila model of oxidative stress showed improvement in locomotion when treated with water extract of Withania somnifera. A similar study was carried out in Drosophila melanogaster by Holvoet et al. [71], which suggested that water and ethanol extract of Withania somnifera showed improvement in stress-induced behavioral changes. Also, it was observed that water extract of Withania somnifera was more potent than ethanol extract. This could be due to the higher concentration of withanolides in water extract. It has been observed that there is an increase in anxiety and depression with age.
A study performed by Singh et al. [72] in middle aged wistar female albino rats (11–12 months old) demonstrated that intermittent fasting-dietary restriction along with supplementation of Withania somnifera and Tinospora cordifolia extract showed decrease in expression of molecular stress chaperones Hsp70, which is responsible for inducing stress. It has also been reported that an increase in stress levels can result in upregulation of inflammatory cytokines. Molecular chaperones (Hsp70) play a key role in protein damage prevention throughout aging, and their expression is required for longevity. Chemical stimulation of HSP production could, thus, be an important method in the future formulation of antiaging drugs [73]. Study of cytokines levels in rats showed downregulation of IL-1β, IL-6, Iba1, and TNF-α [72]. IL-6, TNF-α, and C-reactive protein levels significantly increase in the elderly population and are associated with physical and cognitive performance and can lead to mortality [74]. TNF-α may affect glucose metabolism but aging and obesity prevent normal inhibition of TNF-α production, contributing to decreased insulin sensitivity in older men [75]. In an aging population, beta cell functioning and its renewal was influenced by IL-1β signaling molecule. Increased levels of IL-1β correspond to increased disabilities and mortality rates in geriatric populations [76].
Male reproductive diseases associated with male sexual potency and erectile dysfunction can be promoted due to psychological stress. Withania somnifera has the potential to improve male sexual health by promoting improvement in stress management, healthy aging, and body immune system. Yadav et al. [77] performed a study in sexually sluggish male rats previously exposed to psychological stressors by administering Withania somnifera powder extract for 30 days. It was observed that Withania somnifera promoted the expression of cGMP, acetylcholine, nitric oxide, eNOS, and nNOS, which are associated with penile erectile facilitation. The incidence of penile erectile dysfunction increases with aging due to reduced nitric oxide synthesis, which is caused by decreased NOS expression and reduced endothelial NOS and neuronal NOS activity. This dysregulation leads to oxidative stress which results in an aged penis [78,79]. Also, there was an increase in serum levels of luteinizing hormone, follicle-stimulating hormone, and testosterone. The study concluded that Withania somnifera can be used as a potential phytochemical to improve sexual health in stressed, sexually sluggish male rats. Similarly, Gupta et al. [80] performed an experiment in 180 infertile male patients to study the role of Withania somnifera. Patients were administered with Withania somnifera powder extract (5 g/d for 3 months), which showed improvement in concentration of phenylalanine, histidine, glycerylphosphorylcholine, citrate, alanine, and lactate in seminal plasma.
Kukkemane et al. [81] performed an experiment to demonstrate the role of Withania somnifera against age-related suprachiasmatic nucleus changes. The experiment was performed in male wistar rats administered with 100 mg/kg of Withania somnifera leaf extract. Studies suggested that leaf extract of Withania somnifera can modulate core clock genes expression via SIRT1. SIRT1 is a circadian clock regulator, resulting in the restoration of rhythm/levels in the expression of clock genes. Such changes could restore the rhythmic production of Nrf2, a clock-controlled gene and master transcription factor that regulates antioxidant enzymes. Furthermore, naturally occurring antioxidant enzymes, as well as the possible antioxidants of Withania somnifera leaf extract, may neutralize reactive oxygen species. These results suggest that Withania somnifera leaf extract can alleviate age-related circadian disruption, perhaps leading to healthy aging and longevity.

4.2. Withania somnifera in Inflammation

Inflammation is a body’s immune system response to external stimuli by pathogens or injury that involves a complex biological process. There are various factors associated with inflammation, such as infections, chronic stress, environmental toxins, aging, genetics, hormones, autoimmune disorders, metabolic condition, etc. However, recent studies have suggested that various complex phenomena, such as accumulation of senescent cells, oxidative stress, dysfunction of mitochondria, hormonal changes, nutrition, and genetics due to aging, contribute to progression of inflammation. Sikandan et al. [82] investigated the antiinflammatory role of Withania somnifera water extract in human keratinocyte HaCaT cells and male C67BL/6J mice. It was found that Withania somnifera significantly suppressed inflammatory cytokines TNF-α, IL-1β, IL-6, IL-8, and IL-12 and overexpressed TGF-β1. Moreover, it was observed that Withania somnifera also inhibited NF-ΚB p65, c-Jun N-terminal kinase, and phosphorylation of p38. Also, suppression of TNF-α and upregulation of TGF- β1 were demonstrated in in vivo studies. IL-8 (a potent chemoattractant) is involved in recruiting immune cells to inflammation site [83]. Hence, increased IL-8 levels have been detected in several age-related complications mainly associated with inflammation [84]. Neuroinflammation promoted by prolonged activation of IL-12 and tumor growth factor-β1 are associated with neurodegenerative disorders such as Alzheimer’s disease and mild cognitive impairment [85,86].

4.2.1. Withania somnifera in Neuroinflammation

Neuroinflammation is associated with aging that can lead to various neurological disorders, including Alzheimer’s disease [87]. A study was performed by Gupta et al. [88] in wistar strain male albino rats to understand the underlying role of Withania somnifera leaf water extract in treating neuroinflammation. It was demonstrated that Withania somnifera leaf water extract suppressed cytokines associated with inflammation such as TNF-α, IL-1β, IL-6, reactive nitrogen species, and reactive oxygen species in the brain. Furthermore, it was observed that Withania somnifera leaf water extract suppressed NF-kB, p38, and JNK pathways. Similarly, another study performed by Gupta et al. [89] in primary microglial culture and mouse microglial BV-2 cell line showed suppression of IL-6, IL-1β, TNF-α, reactive nitrogen species, and a reactive oxygen species by downregulating NF-kB and AP1.
Withaferin A, an active constituent of Withania somnifera showed inhibition of NF-kB and REL A transcription factors. Also, overexpression of IKBKB and IKBKG (subunits of NF-kB) along with downregulation of JUN and STAT genes were observed in co-cultured cells of SH-SY5Y transfected with amyloid precursor protein plasmid (SH-APP) and immortalized CHME5 microglia cells [90]. Zhu et al. [91] performed an experiment in adult male C57BL/6 mice to investigate the role of withanolide A, a bioactive compound in Withania somnifera, in pilocarpine-induced status epilepticus. It was observed that withanolide A can suppress IL-1β and TNF in the hippocampus region of the mice. Furthermore, it was concluded withanolide A can provide neuroprotective effects by inhibiting neuroinflammation.
Microtubule associated protein 2 (MAP2) and growth associated protein 43 (GAP43) gene expression helps in maintenance of neuronal functions. An experiment carried out by Gupta et al. [92] in wistar strain male albino rats observed significant downregulation of MAP2 and GAP43 in LPS-mediated inflammation. Also, it was observed that Ashwagandha leaf water extract may protect neurons from neuroinflammation-associated degeneration by controlling the expression of microtubule associated protein 2 and growth associated protein 43. Amphetamine-related drug (+/−)-3,4-methylendioxymethamphetamine (MDMA) is used in treating neurological disorders. However, prolonged use of MDMA can lead to various disorders, including neuroinflammation. An experiment was performed in adult male C57BL/6J mice by Costa et al. [93], to determine potential benefits of Withania somnifera extract in suppressing MDMA-induced toxicity. It was concluded that animals treated with MDMA along with Withania somnifera extract prevented dopaminergic damage. The dopaminergic system in the brain can alter significantly with age, which may have an impact on cognitive performance. Dopamine levels in the human striatum can drop by up to 50% as people age, making the dopamine system particularly susceptible to the effects of aging [94].

4.2.2. Withania somnifera in Lung Inflammation

Lung aging occurs due to various factors, including improper intercellular communication, ECM dysregulation, genomic instability, epigenetic modification, telomere attrition, proteostasis loss, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and deregulated nutrient-sensing. These factors can contribute in the progression of lung inflammation [95]. Kaur et al. [96] demonstrated an increase in the level of IL-10 and a decrease in TNF-α and NF-kB in lung homogenate of monocrotaline-induced Sprague Dawley rats treated with Withania somnifera root powder. Decreasing levels of IL-10 is closely associated to aging-related inflammation, because it facilitates reducing the inflammation of the skeletal muscles [97].

4.2.3. Withania somnifera in Kidney Inflammation

Inflammation and premature aging are a few of the major contributing factors in the progression of chronic kidney disease [98]. Chen et al. [99] treated male C57BL/6J mice model of unilateral ureteral obstruction (UUO) with Withaferin A. Levels of inflammatory markers p-NF-kB-p65, IL-1β, and COX-2 were suppressed in UUO animals treated with Withaferin A for 14 days.

4.2.4. Withania somnifera in Liver Inflammation

The aging process often contributes to modulation in mitochondrial function and nutrient sensing pathways that can lead to inflammation and cellular senescence [100]. Xia et al. [101] investigated the role of Withaferin A in fulminant hepatitis (FH) in liver. In this experiment, C57BL/6J mice were administered with D-galacosamine (GaLN)/lipopolysaccharide (LPS) to induce FH. Withaferin A inhibited hepatic NLRP3 and upregulated the expression of NRF2. Furthermore, it also suppressed the release of hepatic IL-1β, IL-6, and TNF-α. These results suggested that Withaferin A can be used in the treatment of fulminant hepatitis as an immunoregulator. Aging-related NLRP3 inflammasome activation and hyperactivation have been directly linked to inflammatory reactions and cellular damage. Indeed, research has indicated that the activation of the NLRP3 inflammasome leads to intracellular inflammation, which is implicated in the process of aging [102]. Liver X receptors (LXR) are cholesterol-sensing nuclear receptors that regulate metabolism and transportation of lipids. Furthermore, they suppress inflammation by transrepression mechanism [103]. Also, it has been reported that LXR-α negatively modulates NF-kB signaling pathway. Shiragannavar et al. [104] studied the mechanistic role of Withaferin A in HepG2, Hep3B, and Huh-7 hepatocellular carcinoma cells in modulating expression of various genes. It was demonstrated that Withaferin A activated LXR-α that led to suppression of NF-kB, eventually leading to the attenuation of inflammation. Sirtuin 3 (SIRT3) is a deacetylase enzyme that plays a critical role in developing numerous inflammatory diseases, such as liver fibrosis. Gu et al. [105] revealed that Withaferin A activated SIRT3, which suppressed liver fibrosis via upregulating the activity of antioxidant enzymes such as catalase (CAT), superoxide dismutase, and glutathione peroxidase (GPx). Indeed, GPx plays a critical role in the reduction of hydrogen peroxide that contributes to aging by activating NF-κB-related inflammatory signaling and tissue damage [106]. Devkar et al. [107] investigated the hepatoprotective effect of Withania somnifera in acetaminophen-overdosed adult male albino wistar rats. Animals were treated with 750 mg/kg of acetaminophen for 14 days preceded by administration of withanolide rich fraction isolated from Withania somnifera root methanolic extract. Results indicated significant suppression of inflammatory markers such as IL-1β, TNF-α, COX-2, and iNOS in animals treated with withanolide rich fraction. Hence, it was concluded that withanolide rich fraction can have the potential to exert hepatoprotective action by suppressing oxidative stress and inflammation.
Although there is limited clinical evidence on the role(s) of Withania somnifera in neuroinflammation, lung inflammation, kidney inflammation, and liver inflammation, the above-mentioned preclinical studies exploring its effects on these inflammatory conditions offer valuable insights. These studies lay the groundwork for conducting better-informed clinical research to investigate the potential therapeutic benefits of Withania somnifera.

5. Inflammaging and Oxidative Stress: Protective Role of Withania somnifera

Oxidative stress is a state due to imbalance in reactive oxygen species (ROS) and plays a critical role in progression of inflammaging. ROS being highly reactive in nature contributes in inflammaging by damaging cellular components such as DNA, protein, and lipids, causing endoplasmic reticulum stress, and activating inflammatory pathways and senescence-associated secretory phenotype (SASP). Bakar et al. [108] performed an experiment to determine the role of Withaferin A in male C57BL/6J obese mice associated with oxidative stress and inflammation. It was demonstrated that Withaferin A suppresses oxidative stress by maintaining the levels of superoxide dismiutase, glutathione peroxidase, glutathione (GSH), thiobarbituric acid reactive species (TBARS), and catalase. Catalase is essential for maintaining cellular homeostasis because of its capacity to break down hydrogen peroxide, the absence or dysregulation of which is associated with several age-related complications [109]. The antioxidant glutathione, which helps with drug and xenobiotic detoxification strengthens anomalies of the liver, diminishes the complications of diabetes, and protects against viral infections [110]. The role of Withaferin A in cerulein-induced acute pancreatitis was studied by Tiruveedi et al. [111] in adult male Swiss mice. Withaferin A suppressed myeloperodixase (MPO), a peroxidase enzyme and nitrotyrosine and upregulated Nrf2 in acute pancreatitis mice indicating its potential role as an antioxidant. MPO levels are positively correlated with type 2 diabetes, a major risk factor for cardiovascular illnesses and mortality, whereas reduced MPO levels are associated with improved survival in the elderly [112]. To determine the role and underlying mechanism of Withania somnifera in rheumatoid arthritis, Khan et al. [113] performed a study in male Wistar albino rats. This rodent model with rheumatoid arthritis was treated with Withania somnifera water extract and evaluated for various parameters including the measurement of inflammatory cytokines and antioxidant markers. Furthermore, antioxidant superoxide dismutase activity was increased whereas catalase and glutathione peroxidase were suppressed indicating the antioxidant property of Withania somnifera.

6. Inflammaging and DNA Damage: Protective Role of Withania somnifera

Human cells, during their lifespan, undergo DNA damage, which is defined as changes in molecular structure of genomic DNA [114]. DNA damage occurs due to various endogenous (DNA replication errors, spontaneous base deamination, abasic sites, oxidative DNA damage, DNA methylation) and exogenous factors (ionizing radiation, ultraviolet radiation, chemical agents such as alkylating agents, toxins, environmental stresses, DNA damage response) [115]. Numerous theories of DNA damage and DNA repair have suggested that DNA damage can lead to the development of aging and contribute to inflammaging [116,117]. DNA damage response is a complex phenomenon that leads to coordination of cell cycle arrest and activation of repair mechanisms to prevent replication of damaged DNA. Persistent DNA damage response can lead to cell senescence and contribute to chronic inflammation. Senescence-associated secretory phenotype is activated due to chronic DNA damage response and senescent cells lead to release of various proinflammatory cytokines and chemokines, which can contribute to the progression of inflammaging. Also, the activation of immune response due to cell senescence with damaged DNA can further contribute to the progression of inflammaging. Furthermore, chronic inflammation can increase DNA damage by generating ROS and reactive nitrogen species (RNS). The impaired cell function due to accumulation of damaged DNA will further increase proinflammatory cytokines production and continuously maintain chronic inflammatory state. Oxidative stress is one of the major causes of DNA damage and Withania somnifera has been used as an antioxidant agent in treatment of numerous diseases. Hence, Withania somnifera can be used as a potential candidate for suppression of DNA damage that may help in prevention of inflammaging.

7. Inflammaging and Immunomodulation: Protective Role of Withania somnifera

Immunomodulation is a process that involves the regulation of immune system, which can play a critical role in mitigating inflammaging. Immunomodulation is performed by maintaining the level of inflammatory response, regulation of immune cells (macrophages, B cells, and T cells), cytokine regulation, and suppression of immunosenescence effect. Withania somnifera plays an important role in immunomodulation, with clinical evidence suggesting the role of Withania somnifera for the same [118,119]. Also, Withania somnifera helps in increasing the levels of white blood cells that helps in the development of human immune defense. Furthermore, it is used in treating arthritis [120]. Withania somnifera suppresses delayed type hypersensitivity and increases the macrophage phagocytic activity. Also, Withania somnifera increases the nitric oxide signaling in macrophages by stimulating nitric oxide synthase enzyme activity [121]. Tharakan et al. [122] reported the immunomodulatory effect of Withania somnifera in a randomized placebo-controlled double-blinded with an open-label extension study for 30 days in 24 adult male and female healthy volunteers administered with 60 mg of Withania somnifera extract. It was reported that Withania somnifera improved immunity by demonstrating significant increase in IgA, IgM, IgG, IgG2, IgG3, IgG4, IFN-γ, IL-4, CD45+, CD3+, CD4+, CD8+, CD19+, and NK cells. Immunoglobulins, a group of glycoproteins, play a vital role in the immune system. However, the level of IgM (which plays a key role in immune response to pathogens) significantly decreases with age while the level of IgA and IgG increases [123]. IFN-γ and IL-4 are key components of the immune response. IFN-γ promotes cellular immunity against intracellular infections, whereas IL-4 promotes humoral immunity and allergy responses. The balance of these cytokines is critical for immunological homeostasis and proper response to varied immune stressors. IFN-γ and IL-4 dysregulation is linked to a range of immunological disorders, including infections, autoimmune, and allergy problems [124]. Changes in CD45 expression and activity have been observed with aging. CD45 expression increases in healthy aged individuals compared to younger individuals [125]. Priyanka et al. [126] reported suppression of IL-6 suggesting immuno-stimulant properties of Withania somnifera in stress-induced equines in a study carried out for 21 days. A study was performed by Mikolai et al. [127] in which five volunteers were administered with 6 mL Withania somnifera root extract for 96 h. Expression of CD4 on CD3+ T cells and activation of CD56+ NK cells after 96 h in peripheral blood samples was observed. The protective effect of Withania somnifera was investigated in albino mice by Ziauddin et al. [128], who showed that Withania somnifera can be used an immunomodulatory agent to protect against harmful effects of myelosuppressive drugs.

8. Inflammaging and Microbiome: Protective Role of Withania somnifera

The microbiome plays an important role in human health. The microbiome and host human go through numerous physiological changes in aging. These changes can contribute to regulating the immune system, thereby influencing inflammation. Variations in the composition, function, phenotype, diversity, and metabolic output of the microbiome are significant age-dependent alterations that can significantly affect the homeostasis and immune balance. These shifts in the microbiome can contribute to inflammaging, leading to an increased inflammatory responses and excessive adverse effects. In 2015, Clark et al. [129] investigated the impact of microbiota on Drosophila, focusing on heightened intestinal permeability, inflammation, and mortality. Another study in 2016 by Conley et al. [130] explored microbial dysbiosis associated with aging in mice, leading to increased intestinal permeability and subsequent systemic inflammation.
Toll-like receptor protein (TLR4), which is present on the surface of various cells, including the gut, plays a critical role in the innate immune system. Danger-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns (PAMPs) activate TLR4. Lipopolysaccharides (LPS) present on the surface of cell wall of bacteria activate TRL4. In the case of aged patients, TLR4 gets continuously activated due to the intake of bacterial products. Purushotham et al. reported that active phytocompounds of Withania somnifera inhibited TLR-4-activated innate immunity signaling pathways, including MAPK and NF-κB [131].

9. Inflammaging and COVID-19: The Protective Role of Withania somnifera

The severe acute respiratory syndrome coronavirus (COVID-19) pandemic that led to the disruption of normal life severely affected the elderly population. According to the Centers for Disease Control and Prevention, more than 80% of deaths caused by COVID-19 were above the age of 60 years [132]. Inflammaging contributes to various age-associated disorders that increase the risk of COVID-19 disease [133]. These disorders include diabetes, neuropathic degeneration, and rheumatoid arthritis. Furthermore, COVID-19 worsens when it co-exists with these diseases and can also become fatal [133,134,135,136]. Inflammaging can also contribute to the progression of cytokine storm, which is a complex of acute systemic inflammatory symptoms, elevated cytokine levels, and secondary organ dysfunction [137]. SARS-CoV-2 can be triggered by the presence of pro-inflammatory environments in patients suffering from inflammaging. NF-κB is a crucial regulator of inflammation that can contribute to exaggerating the inflammatory response in older people who have inflammation when they contract the virus. Hence, targeting NF-κB signaling may be used as a potential therapeutic approach for COVID-19. Downregulation of NF-κB can help in reducing the severity of cytokine storm. Numerous preclinical studies suggest the role of Withania somnifera in downregulation of NF-κB [53,82,138,139]. Also, the level of pro-inflammatory cytokines, including TNF-α and IL-6, that are observed in cytokine storm can be regulated by Withania somnifera suggesting the potential role of Withania somnifera in managing disease progression associated with COVID-19. Interferon (IFN) proteins play a critical role in immune response. They exist in different forms, i.e., IFN-α, IFN-β, IFN-γ, and IFN-λ, depending on the bodily innate immune response to viral infections. There is dysregulation of IFN response in elderly people including patients suffering from inflammaging that can lead to severe harmful conditions in patients with COVID-19. A study performed by Davis et al. [140] demonstrated enhanced levels of IFN-γ in mice treated with Withania somnifera root extract, suggesting their potential role.
Angiotensin converting enzyme (ACE2) plays a critical role in the regulation of blood pressure, sodium, and fluid balance. The spike protein of SARS-CoV-2 binds to the ACE2 receptor and facilitates the progression of COVID-19. In the renin-angiotensin-aldosterone system, the ACE2 receptor plays an important role in the regulation of inflammation. Angiotensin II is converted to angiotensin 1-7 by ACE2, which reduces the proinflammatory effects including IL-6, TNF-α, and IL-8 by inhibiting P38 mitogen-activated protein kinase (MAPK)-NF-kB pathway. ACE2 has antiinflammatory properties which protects the lungs from injury during acute respiratory distress syndrome (ARDS). Severe COVID-19 cytokine release during SARS-CoV-2 infection is partially triggered by increased angiotensin II, possibly as a result of TNF-α overproduction [141]. Indeed, the underlying mechanism is not completely understood. There are studies which suggest that Withania somnifera can modulate ACE2 that can be used as a potential approach for treating COVID-19; however, more research is required to prove the role of Withania somnifera in modulation of ACE2 in COVID-19 [142]. In Figure 3, we have depicted the molecular pathways for Withania somnifera in inflammaging that are modulated by COVID-19 and the microbiome.
The present discussion that Withania somnifera may potentially play a protective role in mitigating the detrimental effects of inflammaging during COVID-19 is, indeed, speculative given the absence of data at the time of writing. Further preclinical and clinical studies are necessary to validate this proposition.
In addition, we have constructed a protein–protein interaction network, illustrated in Figure 4, for inflammaging modulated by Withania somnifera. A protein–protein interaction network was constructed using Withania somnifera-modulated proteins obtained from the literature, which was filtered using the STRING database and illustrated using Cytoscape 3.10.1. This biological network, which is focused on Withania somnifera-modulated proteins linked to inflammaging, unveils potential targets and key Withania somnifera-regulated pathways, offering valuable insights into therapeutic avenues for age-related inflammation. Furthermore, we have summarized various preclinical studies implicating health-beneficial effects of Withania somnifera in inflammation and aging in Table 1 for the benefit of the reader.

10. Safety and Toxicity of Withania somnifera

Recent investigations suggest the efficacy of Withania somnifera in numerous diseases. Clinical trial studies have been performed to determine the safety and efficacy of Withania somnifera. A pilot study was performed by Sharma et al. [161] with the objective to investigate the safety and efficacy of Withania somnifera in 25 hypothyroid patients. Studies demonstrated no significant changes between the placebo and Withania somnifera-treated groups indicating the safety of Withania somnifera. Likewise, Verma et al. [162] performed a study to investigate safety and tolerability in 80 healthy volunteers. Withania somnifera root extracts were administered orally 300 mg twice daily for 8 weeks. Studies reported no significant change in body weight, body temperature, body mass index, respiratory rate, systolic and diastolic blood pressure, hematological and biochemical parameters, and thyroid hormone, suggesting that Withania somnifera root extract is well-tolerated in healthy male and female volunteers for 8 weeks. Langade et al. [163] demonstrated safety of Withania somnifera in healthy volunteers and insomnia patients treated with Withania somnifera root extract capsules 300 mg twice daily for 8 weeks.

11. Clinical Studies on Withania somnifera

There are various global clinical trials carried out on Withania somnifera, including aging and safety studies. The opportunities and challenges in resource-limited clinical trials have been discussed elsewhere [164]. Herein, in Table 2, we have summarized the various clinical trials adapted from www.clinicaltrials.gov (accessed on 26 December 2023) [165], Australia New Zealand Clinical Trial Registry www.anzctr.org.au/ (accessed on 26 December 2023) [166], and International Clinical Trial Registry Platform www.who.int/clinical-trials-registry-platform (accessed on 26 December 2023) [167] for the benefit of the reader. However, there are limited results available from clinical studies, and almost none of these trial results have been subjected to peer review, thus underscoring their uncertain status.

12. Conclusions and Future Perspectives

Withania somnifera, a traditional Ayurvedic medicine, has numerous health benefits, including its abilities as an antioxidant and antiinflammatory agent. There are numerous preclinical and clinical studies attempting to shed light on Withania somnifera’s potential significance in the treatment of inflammation, neurological disorders, aging, healthy aging, and fertility problems. However, conclusive evidence regarding its therapeutic potential is yet to be established in clinical studies. The aging-associated phenomenon known as inflammaging, prevalent among the elderly, results from multifaceted factors. Withania somnifera has the potential to treat inflammaging, as there is considerable evidence that demonstrates its efficacy. Withania somnifera possesses antioxidant properties that help retard the progression of inflammaging. Furthermore, the antiinflammatory property of Withania somnifera contributes to modulating the chronic low-grade inflammation characteristic of this aging-related disorder. Additionally, antiaging effects, involvement in DNA damage repair mechanisms, and potential effects on the microbiome are factors that suggest a key role for Withania somnifera in inflammaging. However, there is limited preclinical and clinical evidence to firmly support this hypothesis. Hence, more studies are required to be carried out to understand the therapeutic efficacy and mechanism of Withania somnifera in treating inflammaging. Herein, we have discussed the etiology of inflammaging and the chemistry of Withania somnifera. Furthermore, we have illustrated the biological pathways involved in inflammaging that are regulated by Withania somnifera and its active compounds which may help in providing insights into the potential role(s) of Withania somnifera in inflammaging. Also, a protein–protein interaction network was generated in order to have a systems pharmacology perspective of key proteins modulated by Withania somnifera and to help identify key biomarkers as well as provide potential targets for treating inflammaging. This review underscores the potential health-beneficial effects of Withania somnifera in inflammaging and provides hope to improve the health-related quality of life (HRQoL) in aging.

Author Contributions

V.B.: Writing—Original draft preparation, Software; G.G.: Writing—Tables; S.A.: Software, Visualization, Drawing—Figures; M.G.—Revision; D.M.: Writing—Reviewing and Editing, Funding acquisition; S.N.: Conceptualization, Supervision, Project Administration, Writing—Reviewing and Editing. All authors have read and agreed to the published version of the manuscript.

Funding

The author(s) declare that no financial support was received for the research and authorship of this article. The APC was funded by PhytoVeda Pvt. Ltd., Mumbai, India.

Acknowledgments

The authors thank PhytoVeda Pvt. Ltd. for their support and Viridis Biopharma Pvt. Ltd. for electronic access to scientific databases.

Conflicts of Interest

V.B., G.G., S.A., D.M. and S.N. are employees of PhytoVeda Pvt. Ltd. and Viridis Biopharma Pvt. Ltd., Mumbai, India. M.G. declares no conflict of interest.

References

  1. Xia, S.; Zhang, X.; Zheng, S.; Khanabdali, R.; Kalionis, B.; Wu, J.; Wan, W.; Tai, X. An Update on Inflamm-Aging: Mechanisms, Prevention, and Treatment. J. Immunol. Res. 2016, 2016, 8426874. [Google Scholar] [CrossRef] [PubMed]
  2. Antuña, E.; Cachán-Vega, C.; Bermejo-Millo, J.C.; Potes, Y.; Caballero, B.; Vega-Naredo, I.; Coto-Montes, A.; Garcia-Gonzalez, C. Inflammaging: Implications in Sarcopenia. Int. J. Mol. Sci. 2022, 23, 15039. [Google Scholar] [CrossRef] [PubMed]
  3. Li, X.; Li, C.; Zhang, W.; Wang, Y.; Qian, P.; Huang, H. Inflammation and Aging: Signaling Pathways and Intervention Therapies. Sig. Transduct. Target. Ther. 2023, 8, 239. [Google Scholar] [CrossRef] [PubMed]
  4. Franceschi, C.; Bonafè, M.; Valensin, S.; Olivieri, F.; De Luca, M.; Ottaviani, E.; De Benedictis, G. Inflamm-aging: An Evolutionary Perspective on Immunosenescence. Ann. N. Y. Acad. Sci. 2000, 908, 244–254. [Google Scholar] [CrossRef]
  5. Franceschi, C.; Garagnani, P.; Parini, P.; Giuliani, C.; Santoro, A. Inflammaging: A New Immune–Metabolic Viewpoint for Age-Related Diseases. Nat. Rev. Endocrinol. 2018, 14, 576–590. [Google Scholar] [CrossRef] [PubMed]
  6. Ferrucci, L.; Fabbri, E. Inflammageing: Chronic Inflammation in Ageing, Cardiovascular Disease, and Frailty. Nat. Rev. Cardiol. 2018, 15, 505–522. [Google Scholar] [CrossRef] [PubMed]
  7. Mohd Sahardi, N.F.N.; Makpol, S. Suppression of Inflamm-Aging by Moringa Oleifera and Zingiber Officinale Roscoe in the Prevention of Degenerative Diseases: A Review of Current Evidence. Molecules 2023, 28, 5867. [Google Scholar] [CrossRef] [PubMed]
  8. Nainu, F.; Salim, E.; Asri, R.M.; Hori, A.; Kuraishi, T. Neurodegenerative Disorders and Sterile Inflammation: Lessons from a Drosophila Model. J. Biochem. 2019, 166, 213–221. [Google Scholar] [CrossRef] [PubMed]
  9. Basudkar, V.; Ajgaonkar, S.; Mehta, D.; Nair, S. Current Clinical Insights into circRNAs and Signal Transduction in Diabetic Nephropathy. Diabet. Nephrop. 2023, 3, 58–67. [Google Scholar] [CrossRef]
  10. Fernandes, R.; Viana, S.D.; Nunes, S.; Reis, F. Diabetic Gut Microbiota Dysbiosis as an Inflammaging and Immunosenescence Condition That Fosters Progression of Retinopathy and Nephropathy. Biochim. Biophys. Acta (BBA) Mol. Basis Dis. 2019, 1865, 1876–1897. [Google Scholar] [CrossRef]
  11. Li, J.; Lv, J.; Cao, X.; Zhang, H.; Tan, Y.; Chu, T.; Zhao, L.; Liu, Z.; Ren, Y. Gut Microbiota Dysbiosis as an Inflammaging Condition That Regulates Obesity-Related Retinopathy and Nephropathy. Front. Microbiol. 2022, 13, 1040846. [Google Scholar] [CrossRef] [PubMed]
  12. Pandey, A.; Ajgaonkar, S.; Jadhav, N.; Saha, P.; Gurav, P.; Panda, S.; Mehta, D.; Nair, S. Current Insights into miRNA and lncRNA Dysregulation in Diabetes: Signal Transduction, Clinical Trials and Biomarker Discovery. Pharmaceuticals 2022, 15, 1269. [Google Scholar] [CrossRef]
  13. Zhang, Q.; Jazwinski, S.M. A Novel Strategy to Model Age-Related Cancer for Elucidation of the Role of Th17 Inflammaging in Cancer Progression. Cancers 2022, 14, 5185. [Google Scholar] [CrossRef] [PubMed]
  14. Mandlik, D.S.; Namdeo, A.G. Pharmacological Evaluation of Ashwagandha Highlighting Its Healthcare Claims, Safety, and Toxicity Aspects. J. Diet. Suppl. 2021, 18, 183–226. [Google Scholar] [CrossRef] [PubMed]
  15. Mikulska, P.; Malinowska, M.; Ignacyk, M.; Szustowski, P.; Nowak, J.; Pesta, K.; Szeląg, M.; Szklanny, D.; Judasz, E.; Kaczmarek, G.; et al. Ashwagandha (Withania somnifera)—Current Research on the Health-Promoting Activities: A Narrative Review. Pharmaceutics 2023, 15, 1057. [Google Scholar] [CrossRef]
  16. Lopresti, A.L.; Smith, S.J.; Malvi, H.; Kodgule, R. An Investigation into the Stress-Relieving and Pharmacological Actions of an Ashwagandha (Withania somnifera) Extract: A Randomized, Double-Blind, Placebo-Controlled Study. Medicine 2019, 98, e17186. [Google Scholar] [CrossRef]
  17. Speers, A.B.; Cabey, K.A.; Soumyanath, A.; Wright, K.M. Effects of Withania somnifera (Ashwagandha) on Stress and the Stress—Related Neuropsychiatric Disorders Anxiety, Depression, and Insomnia. Curr. Neuropharmacol. 2021, 19, 1468–1495. [Google Scholar] [CrossRef] [PubMed]
  18. Chincholikar, M. Concept of Rasayan and Common Rasayana. Available online: http://ccras.nic.in/content/concept-rasayan-and-common-rasayana (accessed on 5 January 2024).
  19. Paul, S.; Chakraborty, S.; Anand, U.; Dey, S.; Nandy, S.; Ghorai, M.; Saha, S.C.; Patil, M.T.; Kandimalla, R.; Proćków, J.; et al. Withania somnifera (L.) Dunal (Ashwagandha): A Comprehensive Review on Ethnopharmacology, Pharmacotherapeutics, Biomedicinal and Toxicological Aspects. Biomed. Pharmacother. 2021, 143, 112175. [Google Scholar] [CrossRef] [PubMed]
  20. Akhoon, B.A.; Pandey, S.; Tiwari, S.; Pandey, R. Withanolide A Offers Neuroprotection, Ameliorates Stress Resistance and Prolongs the Life Expectancy of Caenorhabditis Elegans. Exp. Gerontol. 2016, 78, 47–56. [Google Scholar] [CrossRef]
  21. KrishnaRaju, A.V.; Somepalli, V.; Thanawala, S.; Shah, R. Efficacy and Anti-Inflammatory Activity of Ashwagandha Sustained-Release Formulation on Depression and Anxiety Induced by Chronic Unpredictable Stress: In Vivo and in Vitro Studies. JEP 2023, 15, 291–305. [Google Scholar] [CrossRef]
  22. Singh, N.; Bhalla, M.; de Jager, P.; Gilca, M. An Overview on Ashwagandha: A Rasayana (Rejuvenator) of Ayurveda. Afr. J. Tradit. Complement. Altern. Med. 2011, 8, 208–213. [Google Scholar] [CrossRef] [PubMed]
  23. Singh, M.; Jayant, K.; Singh, D.; Bhutani, S.; Poddar, N.K.; Chaudhary, A.A.; Khan, S.-U.-D.; Adnan, M.; Siddiqui, A.J.; Hassan, M.I.; et al. Withania somnifera (L.) Dunal (Ashwagandha) for the Possible Therapeutics and Clinical Management of SARS-CoV-2 Infection: Plant-Based Drug Discovery and Targeted Therapy. Front. Cell. Infect. Microbiol. 2022, 12, 933824. [Google Scholar] [CrossRef] [PubMed]
  24. Ahmed, W.; Mofed, D.; Zekri, A.-R.; El-Sayed, N.; Rahouma, M.; Sabet, S. Antioxidant Activity and Apoptotic Induction as Mechanisms of Action of Withania somnifera (Ashwagandha) against a Hepatocellular Carcinoma Cell Line. J. Int. Med. Res. 2018, 46, 1358–1369. [Google Scholar] [CrossRef] [PubMed]
  25. Sharma, P.K.; Kumar, L.; Goswami, Y.; Pujani, M.; Dikshit, M.; Tandon, R. The Aqueous Root Extract of Withania somnifera Ameliorates LPS-Induced Inflammatory Changes in the in Vitro Cell-Based and Mice Models of Inflammation. Front. Pharmacol. 2023, 14, 1139654. [Google Scholar] [CrossRef] [PubMed]
  26. Tiwari, S.; Gupta, S.K.; Pathak, A.K. A Double-Blind, Randomized, Placebo-Controlled Trial on the Effect of Ashwagandha (Withania somnifera Dunal.) Root Extract in Improving Cardiorespiratory Endurance and Recovery in Healthy Athletic Adults. J. Ethnopharmacol. 2021, 272, 113929. [Google Scholar] [CrossRef] [PubMed]
  27. Kuboyama, T.; Tohda, C.; Komatsu, K. Effects of Ashwagandha (Roots of Withania somnifera) on Neurodegenerative Diseases. Biol. Pharm. Bull. 2014, 37, 892–897. [Google Scholar] [CrossRef] [PubMed]
  28. Sengupta, P.; Agarwal, A.; Pogrebetskaya, M.; Roychoudhury, S.; Durairajanayagam, D.; Henkel, R. Role of Withania somnifera (Ashwagandha) in the Management of Male Infertility. Reprod. BioMedicine Online 2018, 36, 311–326. [Google Scholar] [CrossRef] [PubMed]
  29. Mehta, V.; Chander, H.; Munshi, A. Mechanisms of Anti-Tumor Activity of Withania somnifera (Ashwagandha). Nutr. Cancer 2021, 73, 914–926. [Google Scholar] [CrossRef]
  30. Shah, D.; Gandhi, M.; Kumar, A.; Cruz-Martins, N.; Sharma, R.; Nair, S. Current Insights into Epigenetics, Noncoding RNA Interactome and Clinical Pharmacokinetics of Dietary Polyphenols in Cancer Chemoprevention. Crit. Rev. Food Sci. Nutr. 2021, 63, 1755–1791. [Google Scholar] [CrossRef]
  31. Sodvadiya, M.; Patel, H.; Mishra, A.; Nair, S. Emerging Insights into Anticancer Chemopreventive Activities of Nutraceutical Moringa Oleifera: Molecular Mechanisms, Signal Transduction and In Vivo Efficacy. Curr. Pharmacol. Rep. 2020, 6, 38–51. [Google Scholar] [CrossRef]
  32. Jain, R.; Nair, S. Sandalwood Oil for the Chemoprevention of Skin Cancer: Mechanistic Insights, Anti-Inflammatory, and In Vivo Anticancer Potential. Curr. Pharmacol. Rep. 2019, 5, 345–358. [Google Scholar] [CrossRef]
  33. Nair, S.; Ah-Ng Tony, K. Pharmacometrics of Nutraceutical Sulforaphane and Its Implications in Prostate Cancer Prevention. J. Chin. Phram. Sci. 2016, 25, 12–22. [Google Scholar] [CrossRef]
  34. Jadhav, N.; Ajgaonkar, S.; Saha, P.; Gurav, P.; Pandey, A.; Basudkar, V.; Gada, Y.; Panda, S.; Jadhav, S.; Mehta, D.; et al. Molecular Pathways and Roles for Vitamin K2-7 as a Health-Beneficial Nutraceutical: Challenges and Opportunities. Front. Pharmacol. 2022, 13, 896920. [Google Scholar] [CrossRef] [PubMed]
  35. Vaidya, R.; Godse, C.; Jadhav, S.; Saha, P.; Ajgaonkar, S.; Pandey, A.; Gurav, P.; Jadhav, N.; Mehta, D.; Nair, S. An Intrinsic Need for Vitamin K2-7 Supplementation: A Narrative Review of K2-7 and Peripheral Neuropathy. BJSTR 2022, 42, 33679–33687. [Google Scholar] [CrossRef]
  36. Chen, C.; Pung, D.; Leong, V.; Hebbar, V.; Shen, G.; Nair, S.; Li, W.; Tony Kong, A.-N. Induction of Detoxifying Enzymes by Garlic Organosulfur Compounds through Transcription Factor Nrf2: Effect of Chemical Structure and Stress Signals. Free. Radic. Biol. Med. 2004, 37, 1578–1590. [Google Scholar] [CrossRef]
  37. Shen, G.; Xu, C.; Hu, R.; Jain, M.R.; Nair, S.; Lin, W.; Yang, C.S.; Chan, J.Y.; Kong, A.-N.T. Comparison of (−)-Epigallocatechin-3-Gallate Elicited Liver and Small Intestine Gene Expression Profiles Between C57BL/6J Mice and C57BL/6J/Nrf2 (−/−) Mice. Pharm. Res. 2005, 22, 1805–1820. [Google Scholar] [CrossRef] [PubMed]
  38. Shen, G.; Xu, C.; Hu, R.; Jain, M.R.; Gopalkrishnan, A.; Nair, S.; Huang, M.-T.; Chan, J.Y.; Kong, A.-N.T. Modulation of Nuclear Factor E2-Related Factor 2–Mediated Gene Expression in Mice Liver and Small Intestine by Cancer Chemopreventive Agent Curcumin. Mol. Cancer Ther. 2006, 5, 39–51. [Google Scholar] [CrossRef]
  39. Gopalakrishnan, A.; Xu, C.-J.; Nair, S.S.; Chen, C.; Hebbar, V.; Kong, A.-N.T. Modulation of Activator Protein-1 (AP-1) and MAPK Pathway by Flavonoids in Human Prostate Cancer PC3 Cells. Arch. Pharm. Res. 2006, 29, 633–644. [Google Scholar] [CrossRef]
  40. Barve, A.; Khor, T.O.; Nair, S.; Lin, W.; Yu, S.; Jain, M.R.; Chan, J.Y.; Kong, A.-N. Pharmacogenomic Profile of Soy Isoflavone Concentrate in the Prostate of Nrf2 Deficient and Wild-Type Mice. J. Pharm. Sci. 2008, 97, 4528–4545. [Google Scholar] [CrossRef]
  41. Barve, A.; Khor, T.O.; Nair, S.; Reuhl, K.; Suh, N.; Reddy, B.; Newmark, H.; Kong, A.-N. γ-Tocopherol-Enriched Mixed Tocopherol Diet Inhibits Prostate Carcinogenesis in TRAMP Mice. Int. J. Cancer 2009, 124, 1693–1699. [Google Scholar] [CrossRef]
  42. Nair, S.; Barve, A.; Khor, T.-O.; Shen, G.; Lin, W.; Chan, J.Y.; Cai, L.; Kong, A.-N. Regulation of Nrf2- and AP-1-Mediated Gene Expression by Epigallocatechin-3-Gallate and Sulforaphane in Prostate of Nrf2-Knockout or C57BL/6J Mice and PC-3 AP-1 Human Prostate Cancer Cells. Acta Pharmacol. Sin. 2010, 31, 1223–1240. [Google Scholar] [CrossRef] [PubMed]
  43. Nair, S.; Hebbar, V.; Shen, G.; Gopalakrishnan, A.; Khor, T.O.; Yu, S.; Xu, C.; Kong, A.-N. Synergistic Effects of a Combination of Dietary Factors Sulforaphane and (−) Epigallocatechin-3-Gallate in HT-29 AP-1 Human Colon Carcinoma Cells. Pharm. Res. 2008, 25, 387–399. [Google Scholar] [CrossRef] [PubMed]
  44. Fuentes, F.; Gomez, Y.; Paredes-Gonzalez, X.; Barve, A.; Nair, S.; Yu, S. Nrf2-Mediated Antioxidant and Detoxifying Enzyme Induction by a Combination of Curcumin and Sulforaphane. J. Chin. Pharm. Sci. 2016, 25, 559–569. [Google Scholar]
  45. Mishra, L.-C.; Singh, B.B.; Dagenais, S. Scientific Basis for the Therapeutic Use of Withania somnifera (Ashwagandha): A Review. Altern. Med. Rev. 2000, 5, 334–346. [Google Scholar]
  46. Ghosal, S.; Lal, J.; Srivastava, R.; Bhattacharya, S.K.; Upadhyay, S.N.; Jaiswal, A.K.; Chattopadhyay, U. Immunomodulatory and CNS Effects of Sitoindosides IX and X, Two New Glycowithanolides from Withania somnifera. Phytother. Res. 1989, 3, 201–206. [Google Scholar] [CrossRef]
  47. Singh, M.; Manoj, J.; Sahu, D.; Kumar, Y. Augmentation of Meat Quality Attributes of Broilers by Dietary Supplementation of Selenium and Ashwagandha. Int. J. Livest. Res. 2020, 1, 73–78. [Google Scholar] [CrossRef]
  48. Wallace, T.C.; Blusztajn, J.K.; Caudill, M.A.; Klatt, K.C.; Natker, E.; Zeisel, S.H.; Zelman, K.M. Choline: The Underconsumed and Underappreciated Essential Nutrient. Nutr. Today 2018, 53, 240–253. [Google Scholar] [CrossRef]
  49. Ahmed, H.A.; El-Darier, S.M. Phytochemistry, Allelopathy and Anticancer Potentiality of Withania somnifera (L.) Dunal (Solanaceae). Braz. J. Biol. 2024, 84, e263815. [Google Scholar] [CrossRef] [PubMed]
  50. Bhople, S.; Singh, M. Effect of Iron Enrichment on Textural Properties of Rice Based Ashwagandha (Withania somnifera) Fortified Extruded Snacks. Chem. Sci. Rev. Lett. 2017, 6, 1468–1475. [Google Scholar]
  51. Lagoumtzi, S.M.; Chondrogianni, N. Senolytics and Senomorphics: Natural and Synthetic Therapeutics in the Treatment of Aging and Chronic Diseases. Free. Radic. Biol. Med. 2021, 171, 169–190. [Google Scholar] [CrossRef]
  52. Fulop, T.; Larbi, A.; Dupuis, G.; Le Page, A.; Frost, E.H.; Cohen, A.A.; Witkowski, J.M.; Franceschi, C. Immunosenescence and Inflamm-Aging As Two Sides of the Same Coin: Friends or Foes? Front. Immunol. 2018, 8, 1960. [Google Scholar] [CrossRef] [PubMed]
  53. Grunz-Borgmann, E.; Mossine, V.; Fritsche, K.; Parrish, A.R. Ashwagandha Attenuates TNF-α- and LPS-Induced NF-κB Activation and CCL2 and CCL5 Gene Expression in NRK-52E Cells. BMC Complement. Altern. Med. 2015, 15, 434. [Google Scholar] [CrossRef]
  54. Pivari, F.; Mingione, A.; Piazzini, G.; Ceccarani, C.; Ottaviano, E.; Brasacchio, C.; Dei Cas, M.; Vischi, M.; Cozzolino, M.G.; Fogagnolo, P.; et al. Curcumin Supplementation (Meriva®) Modulates Inflammation, Lipid Peroxidation and Gut Microbiota Composition in Chronic Kidney Disease. Nutrients 2022, 14, 231. [Google Scholar] [CrossRef] [PubMed]
  55. Lee, S.; Choi, Y.J.; Lee, S.; Kang, K.S.; Jang, T.S.; Kim, K.H. Protective Effects of Withagenin A Diglucoside from Indian Ginseng (Withania somnifera) against Human Dermal Fibroblast Damaged by TNF-α Stimulation. Antioxidants 2022, 11, 2248. [Google Scholar] [CrossRef] [PubMed]
  56. Ko, Y.S.; Pyo, J.-S.; Cho, W.J. Roles of NF-κB Activation in Benign Prostatic Hyperplasia and Association between NF-κB and HIF-1α. Pathol. Res. Pract. 2022, 237, 154021. [Google Scholar] [CrossRef] [PubMed]
  57. Kim, E.K.; Choi, E.-J. Pathological Roles of MAPK Signaling Pathways in Human Diseases. Biochim. Biophys. Acta (BBA) Mol. Basis Dis. 2010, 1802, 396–405. [Google Scholar] [CrossRef] [PubMed]
  58. Minamino, T.; Miyauchi, H.; Tateno, K.; Kunieda, T.; Komuro, I. Akt-Induced Cellular Senescence: Implication for Human Disease. Cell Cycle 2004, 3, 449–451. [Google Scholar] [CrossRef] [PubMed]
  59. Zdanov, S.; Bernard, D.; Debacq-Chainiaux, F.; Martien, S.; Gosselin, K.; Vercamer, C.; Chelli, F.; Toussaint, O.; Abbadie, C. Normal or Stress-Induced Fibroblast Senescence Involves COX-2 Activity. Exp. Cell Res. 2007, 313, 3046–3056. [Google Scholar] [CrossRef]
  60. Babu, P.V.A.; Gokulakrishnan, A.; Dhandayuthabani, R.; Ameethkhan, D.; Kumar, C.V.P.; Ahamed, M.I.N. Protective Effect of Withania somnifera (Solanaceae) on Collagen Glycation and Cross-Linking. Comp. Biochem. Physiol. Part B Biochem. Mol. Biol. 2007, 147, 308–313. [Google Scholar] [CrossRef]
  61. Salvadori, L.; Mandrone, M.; Manenti, T.; Ercolani, C.; Cornioli, L.; Lianza, M.; Tomasi, P.; Chiappalupi, S.; Di Filippo, E.S.; Fulle, S.; et al. Identification of Withania somnifera-Silybum Marianum-Trigonella Foenum-Graecum Formulation as a Nutritional Supplement to Contrast Muscle Atrophy and Sarcopenia. Nutrients 2020, 13, 49. [Google Scholar] [CrossRef]
  62. Brennan, C.M.; Emerson, C.P.; Owens, J.; Christoforou, N. P38 MAPKs—Roles in Skeletal Muscle Physiology, Disease Mechanisms, and as Potential Therapeutic Targets. JCI Insight 2021, 6, e149915. [Google Scholar] [CrossRef] [PubMed]
  63. Pradhan, R.; Yadav, S.K.; Prem, N.N.; Bhagel, V.; Pathak, M.; Shekhar, S.; Gaikwad, S.; Dwivedi, S.N.; Bal, C.S.; Dey, A.B.; et al. Serum FOXO3A: A Ray of Hope for Early Diagnosis of Alzheimer’s Disease. Mech. Ageing Dev. 2020, 190, 111290. [Google Scholar] [CrossRef] [PubMed]
  64. Salih, D.A.; Brunet, A. FoxO Transcription Factors in the Maintenance of Cellular Homeostasis during Aging. Curr. Opin. Cell Biol. 2008, 20, 126–136. [Google Scholar] [CrossRef]
  65. Pradhan, R.; Kumar, R.; Shekhar, S.; Rai, N.; Ambashtha, A.; Banerjee, J.; Pathak, M.; Dwivedi, S.N.; Dey, S.; Dey, A.B. Longevity and Healthy Ageing Genes FOXO3A and SIRT3: Serum Protein Marker and New Road Map to Burst Oxidative Stress by Withania somnifera. Exp. Gerontol. 2017, 95, 9–15. [Google Scholar] [CrossRef] [PubMed]
  66. Kuchewar, V.; Borkar, M.; Nisargandha, M. Evaluation of Antioxidant Potential of Rasayana Drugs in Healthy Human Volunteers. Ayu 2014, 35, 46–49. [Google Scholar] [CrossRef] [PubMed]
  67. Rizvi, S.I.; Maurya, P. Alterations in Antioxidant Enzymes During Aging in Humans. Mol. Biotechnol. 2007, 37, 58–61. [Google Scholar] [CrossRef] [PubMed]
  68. Bruns, D.R.; Drake, J.C.; Biela, L.M.; Peelor, F.F.; Miller, B.F.; Hamilton, K.L. Nrf2 Signaling and the Slowed Aging Phenotype: Evidence from Long-Lived Models. Oxidative Med. Cell. Longev. 2015, 2015, 732596. [Google Scholar] [CrossRef] [PubMed]
  69. Hybertson, B.M.; Gao, B.; Bose, S.; McCord, J.M. Phytochemical Combination PB125 Activates the Nrf2 Pathway and Induces Cellular Protection against Oxidative Injury. Antioxidants 2019, 8, 119. [Google Scholar] [CrossRef] [PubMed]
  70. Cabey, K.; Long, D.M.; Law, A.; Gray, N.E.; McClure, C.; Caruso, M.; Lak, P.; Wright, K.M.; Stevens, J.F.; Maier, C.S.; et al. Withania somnifera and Centella Asiatica Extracts Ameliorate Behavioral Deficits in an In Vivo Drosophila Melanogaster Model of Oxidative Stress. Antioxidants 2022, 11, 121. [Google Scholar] [CrossRef]
  71. Holvoet, H.; Long, D.M.; Law, A.; McClure, C.; Choi, J.; Yang, L.; Marney, L.; Poeck, B.; Strauss, R.; Stevens, J.F.; et al. Withania somnifera Extracts Promote Resilience against Age-Related and Stress-Induced Behavioral Phenotypes in Drosophila Melanogaster; a Possible Role of Other Compounds besides Withanolides. Nutrients 2022, 14, 3923. [Google Scholar] [CrossRef]
  72. Singh, H.; Kaur, T.; Manchanda, S.; Kaur, G. Intermittent Fasting Combined with Supplementation with Ayurvedic Herbs Reduces Anxiety in Middle Aged Female Rats by Anti-Inflammatory Pathways. Biogerontology 2017, 18, 601–614. [Google Scholar] [CrossRef]
  73. Calderwood, S.K.; Murshid, A.; Prince, T. The Shock of Aging: Molecular Chaperones and the Heat Shock Response in Longevity and Aging—A Mini-Review. Gerontology 2009, 55, 550–558. [Google Scholar] [CrossRef] [PubMed]
  74. Puzianowska-Kuźnicka, M.; Owczarz, M.; Wieczorowska-Tobis, K.; Nadrowski, P.; Chudek, J.; Slusarczyk, P.; Skalska, A.; Jonas, M.; Franek, E.; Mossakowska, M. Interleukin-6 and C-Reactive Protein, Successful Aging, and Mortality: The PolSenior Study. Immun. Ageing 2016, 13, 21. [Google Scholar] [CrossRef]
  75. Kirwan, J.P.; Krishnan, R.K.; Weaver, J.A.; Del Aguila, L.F.; Evans, W.J. Human Aging Is Associated with Altered TNF-α Production during Hyperglycemia and Hyperinsulinemia. Am. J. Physiol. Endocrinol. Metab. 2001, 281, E1137–E1143. [Google Scholar] [CrossRef]
  76. Böni-Schnetzler, M.; Méreau, H.; Rachid, L.; Wiedemann, S.J.; Schulze, F.; Trimigliozzi, K.; Meier, D.T.; Donath, M.Y. IL-1beta Promotes the Age-Associated Decline of Beta Cell Function. iScience 2021, 24, 103250. [Google Scholar] [CrossRef] [PubMed]
  77. Yadav, A.; Mishra, R.K. Withania somnifera Ameliorates Sexual Arousal and Impotence in Stressed Sexually Sluggish Male Rats by Modulating Neurotransmitters and NO/cGMP/PDE5α Pathway. J. Ethnopharmacol. 2024, 318, 116971. [Google Scholar] [CrossRef]
  78. Musicki, B.; Burnett, A.L. eNOS Function and Dysfunction in the Penis. Exp. Biol. Med. 2006, 231, 154–165. [Google Scholar] [CrossRef] [PubMed]
  79. Dashwood, M.R.; Crump, A.; Shi-Wen, X.; Loesch, A. Identification of Neuronal Nitric Oxide Synthase (nNOS) in Human Penis: A Potential Role of Reduced Neuronally-Derived Nitric Oxide in Erectile Dysfunction. Curr. Pharm. Biotechnol. 2011, 12, 1316–1321. [Google Scholar] [CrossRef] [PubMed]
  80. Gupta, A.; Mahdi, A.A.; Shukla, K.K.; Ahmad, M.K.; Bansal, N.; Sankhwar, P.; Sankhwar, S.N. Efficacy of Withania somnifera on Seminal Plasma Metabolites of Infertile Males: A Proton NMR Study at 800MHz. J. Ethnopharmacol. 2013, 149, 208–214. [Google Scholar] [CrossRef]
  81. Kukkemane, K.; Jagota, A. Therapeutic Effects of Hydro-Alcoholic Leaf Extract of Withania somnifera on Age-Induced Changes in Daily Rhythms of Sirt1, Nrf2 and Rev-Erbα in the SCN of Male Wistar Rats. Biogerontology 2020, 21, 593–607. [Google Scholar] [CrossRef]
  82. Sikandan, A.; Shinomiya, T.; Nagahara, Y. Ashwagandha Root Extract Exerts Anti-inflammatory Effects in HaCaT Cells by Inhibiting the MAPK/NF-κB Pathways and by Regulating Cytokines. Int. J. Mol. Med. 2018, 42, 425–434. [Google Scholar] [CrossRef] [PubMed]
  83. Bickel, M. The Role of Interleukin-8 in Inflammation and Mechanisms of Regulation. J. Periodontol. 1993, 64, 456–460. [Google Scholar] [PubMed]
  84. Rea, I.M.; Gibson, D.S.; McGilligan, V.; McNerlan, S.E.; Alexander, H.D.; Ross, O.A. Age and Age-Related Diseases: Role of Inflammation Triggers and Cytokines. Front. Immunol. 2018, 9, 586. [Google Scholar] [CrossRef] [PubMed]
  85. Lin, E.; Kuo, P.-H.; Liu, Y.-L.; Yang, A.C.; Tsai, S.-J. Association and Interaction Effects of Interleukin-12 Related Genes and Physical Activity on Cognitive Aging in Old Adults in the Taiwanese Population. Front. Neurol. 2019, 10, 1065. [Google Scholar] [CrossRef] [PubMed]
  86. Fessel, J. Ineffective Levels of Transforming Growth Factors and Their Receptor Account for Old Age Being a Risk Factor for Alzheimer’s Disease. Alzheimers Dement. 2019, 5, 899–905. [Google Scholar] [CrossRef] [PubMed]
  87. Andronie-Cioara, F.L.; Ardelean, A.I.; Nistor-Cseppento, C.D.; Jurcau, A.; Jurcau, M.C.; Pascalau, N.; Marcu, F. Molecular Mechanisms of Neuroinflammation in Aging and Alzheimer’s Disease Progression. Int. J. Mol. Sci. 2023, 24, 1869. [Google Scholar] [CrossRef] [PubMed]
  88. Gupta, M.; Kaur, G. Withania somnifera as a Potential Anxiolytic and Anti-Inflammatory Candidate Against Systemic Lipopolysaccharide-Induced Neuroinflammation. Neuromol Med 2018, 20, 343–362. [Google Scholar] [CrossRef]
  89. Gupta, M.; Kaur, G. Aqueous Extract from the Withania somnifera Leaves as a Potential Anti-Neuroinflammatory Agent: A Mechanistic Study. J. Neuroinflamm. 2016, 13, 193. [Google Scholar] [CrossRef]
  90. Atluri, V.S.R.; Tiwari, S.; Rodriguez, M.; Kaushik, A.; Yndart, A.; Kolishetti, N.; Yatham, M.; Nair, M. Inhibition of Amyloid-Beta Production, Associated Neuroinflammation, and Histone Deacetylase 2-Mediated Epigenetic Modifications Prevent Neuropathology in Alzheimer’s Disease in Vitro Model. Front. Aging Neurosci. 2020, 11, 342. [Google Scholar] [CrossRef]
  91. Zhu, J.; Park, S.; Jeong, K.H.; Kim, W.-J. Withanolide-A Treatment Exerts a Neuroprotective Effect via Inhibiting Neuroinflammation in the Hippocampus after Pilocarpine-Induced Status Epilepticus. Epilepsy Res. 2020, 165, 106394. [Google Scholar] [CrossRef]
  92. Gupta, M.; Kaur, G. Withania somnifera (L.) Dunal Ameliorates Neurodegeneration and Cognitive Impairments Associated with Systemic Inflammation. BMC Complement. Altern. Med. 2019, 19, 217. [Google Scholar] [CrossRef] [PubMed]
  93. Costa, G.; Serra, M.; Maccioni, R.; Casu, M.A.; Kasture, S.B.; Acquas, E.; Morelli, M. Withania somnifera Influences MDMA-Induced Hyperthermic, Cognitive, Neurotoxic and Neuroinflammatory Effects in Mice. Biomed. Pharmacother. 2023, 161, 114475. [Google Scholar] [CrossRef] [PubMed]
  94. Berry, A.S.; Shah, V.D.; Baker, S.L.; Vogel, J.W.; O’Neil, J.P.; Janabi, M.; Schwimmer, H.D.; Marks, S.M.; Jagust, W.J. Aging Affects Dopaminergic Neural Mechanisms of Cognitive Flexibility. J. Neurosci. 2016, 36, 12559–12569. [Google Scholar] [CrossRef] [PubMed]
  95. Faniyi, A.A.; Hughes, M.J.; Scott, A.; Belchamber, K.B.R.; Sapey, E. Inflammation, Ageing and Diseases of the Lung: Potential Therapeutic Strategies from Shared Biological Pathways. Br. J Pharmacol. 2022, 179, 1790–1807. [Google Scholar] [CrossRef] [PubMed]
  96. Kaur, G.; Singh, N.; Samuel, S.S.; Bora, H.K.; Sharma, S.; Pachauri, S.D.; Dwivedi, A.K.; Siddiqui, H.H.; Hanif, K. Withania somnifera Shows a Protective Effect in Monocrotaline-Induced Pulmonary Hypertension. Pharm. Biol. 2015, 53, 147–157. [Google Scholar] [CrossRef] [PubMed]
  97. IL-10 Prevents Aging-associated Inflammation and Insulin Resistance in Skeletal Muscle—Dagdeviren—2017—The FASEB Journal—Wiley Online Library. Available online: https://faseb.onlinelibrary.wiley.com/doi/abs/10.1096/fj.201600832R (accessed on 25 April 2024).
  98. Ebert, T.; Pawelzik, S.-C.; Witasp, A.; Arefin, S.; Hobson, S.; Kublickiene, K.; Shiels, P.G.; Bäck, M.; Stenvinkel, P. Inflammation and Premature Ageing in Chronic Kidney Disease. Toxins 2020, 12, 227. [Google Scholar] [CrossRef] [PubMed]
  99. Chen, C.-M.; Chung, Y.-P.; Liu, C.-H.; Huang, K.-T.; Guan, S.-S.; Chiang, C.-K.; Wu, C.-T.; Liu, S.-H. Withaferin A Protects against Endoplasmic Reticulum Stress-Associated Apoptosis, Inflammation, and Fibrosis in the Kidney of a Mouse Model of Unilateral Ureteral Obstruction. Phytomedicine 2020, 79, 153352. [Google Scholar] [CrossRef] [PubMed]
  100. Hunt, N.J.; Kang, S.W.; Lockwood, G.P.; Le Couteur, D.G.; Cogger, V.C. Hallmarks of Aging in the Liver. Comput. Struct. Biotechnol. J. 2019, 17, 1151–1161. [Google Scholar] [CrossRef] [PubMed]
  101. Xia, Y.; Wang, P.; Yan, N.; Gonzalez, F.J.; Yan, T. Withaferin A Alleviates Fulminant Hepatitis by Targeting Macrophage and NLRP3. Cell Death Dis. 2021, 12, 174. [Google Scholar] [CrossRef]
  102. The Role of NLRP3 Inflammasome in Aging and Age-Related Diseases—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/38317229/ (accessed on 25 April 2024).
  103. Im, S.-S.; Osborne, T.F. Liver X Receptors in Atherosclerosis and Inflammation. Circ. Res. 2011, 108, 996–1001. [Google Scholar] [CrossRef]
  104. Shiragannavar, V.D.; Gowda, N.G.S.; Kumar, D.P.; Mirshahi, F.; Santhekadur, P.K. Withaferin A Acts as a Novel Regulator of Liver X Receptor-α in HCC. Front. Oncol. 2021, 10, 628506. [Google Scholar] [CrossRef] [PubMed]
  105. Gu, J.; Chen, C.; Wang, J.; Chen, T.; Yao, W.; Yan, T.; Liu, Z. Withaferin A Exerts Preventive Effect on Liver Fibrosis through Oxidative Stress Inhibition in a Sirtuin 3-Dependent Manner. Oxidative Med. Cell. Longev. 2020, 2020, 2452848. [Google Scholar] [CrossRef] [PubMed]
  106. Espinoza, S.E.; Guo, H.; Fedarko, N.; DeZern, A.; Fried, L.P.; Xue, Q.-L.; Leng, S.; Beamer, B.; Walston, J.D. Glutathione Peroxidase Enzyme Activity in Aging. J. Gerontol. Biol. Sci. Med. Sci. 2008, 63, 505–509. [Google Scholar] [CrossRef]
  107. Devkar, S.T.; Kandhare, A.D.; Zanwar, A.A.; Jagtap, S.D.; Katyare, S.S.; Bodhankar, S.L.; Hegde, M.V. Hepatoprotective Effect of Withanolide-Rich Fraction in Acetaminophen-Intoxicated Rat: Decisive Role of TNF-α, IL-1β, COX-II and iNOS. Pharm. Biol. 2016, 54, 2394–2403. [Google Scholar] [CrossRef] [PubMed]
  108. Abu Bakar, M.H.; Azmi, M.N.; Shariff, K.A.; Tan, J.S. Withaferin A Protects Against High-Fat Diet–Induced Obesity Via Attenuation of Oxidative Stress, Inflammation, and Insulin Resistance. Appl. Biochem. Biotechnol. 2019, 188, 241–259. [Google Scholar] [CrossRef] [PubMed]
  109. Role of Catalase in Oxidative Stress- and Age-Associated Degenerative Diseases. Available online: https://www.hindawi.com/journals/omcl/2019/9613090/ (accessed on 25 April 2024).
  110. Weschawalit, S.; Thongthip, S.; Phutrakool, P.; Asawanonda, P. Glutathione and Its Antiaging and Antimelanogenic Effects. Clin. Cosmet. Investig. Dermatol. 2017, 10, 147–153. [Google Scholar] [CrossRef] [PubMed]
  111. Tiruveedi, V.L.; Bale, S.; Khurana, A.; Godugu, C. Withaferin A, a Novel Compound of Indian Ginseng (Withania somnifera), Ameliorates Cerulein-Induced Acute Pancreatitis: Possible Role of Oxidative Stress and Inflammation: Withaferin a Ameliorates Acute Pancreatitis. Phytother. Res. 2018, 32, 2586–2596. [Google Scholar] [CrossRef] [PubMed]
  112. Giovannini, S.; Onder, G.; Leeuwenburgh, C.; Carter, C.; Marzetti, E.; Russo, A.; Capoluongo, E.; Pahor, M.; Bernabei, R.; Landi, F. Myeloperoxidase Levels and Mortality in Frail Community-Living Elderly Individuals. J. Gerontol. Biol. Sci. Med. Sci. 2010, 65A, 369–376. [Google Scholar] [CrossRef] [PubMed]
  113. Khan, M.A.; Ahmed, R.S.; Chandra, N.; Arora, V.K.; Ali, A. In Vivo, Extract from Withania somnifera Root Ameliorates Arthritis via Regulation of Key Immune Mediators of Inflammation in Experimental Model of Arthritis. AIAAMC 2019, 18, 55–70. [Google Scholar] [CrossRef]
  114. Lu, Y.; Liu, Y.; Yang, C. Evaluating In Vitro DNA Damage Using Comet Assay. JoVE 2017, 128, e56450. [Google Scholar] [CrossRef]
  115. Chatterjee, N.; Walker, G.C. Mechanisms of DNA Damage, Repair, and Mutagenesis. Env. Mol Mutagen 2017, 58, 235–263. [Google Scholar] [CrossRef] [PubMed]
  116. Maynard, S.; Fang, E.F.; Scheibye-Knudsen, M.; Croteau, D.L.; Bohr, V.A. DNA Damage, DNA Repair, Aging, and Neurodegeneration. Cold Spring Harb. Perspect. Med. 2015, 5, a025130. [Google Scholar] [CrossRef]
  117. White, R.R.; Milholland, B.; de Bruin, A.; Curran, S.; Laberge, R.-M.; van Steeg, H.; Campisi, J.; Maslov, A.Y.; Vijg, J. Controlled Induction of DNA Double-Strand Breaks in the Mouse Liver Induces Features of Tissue Ageing. Nat. Commun. 2015, 6, 6790. [Google Scholar] [CrossRef] [PubMed]
  118. Chengappa, K.N.R.; Brar, J.S.; Gannon, J.M.; Schlicht, P.J. Adjunctive Use of a Standardized Extract of Withania somnifera (Ashwagandha) to Treat Symptom Exacerbation in Schizophrenia: A Randomized, Double-Blind, Placebo-Controlled Study. J. Clin. Psychiatry 2018, 79, 17m11826. [Google Scholar] [CrossRef] [PubMed]
  119. Chopra, A.; Srikanth, N.; Patwardhan, B. Withania somnifera as a Safer Option to Hydroxychloroquine in the Chemoprophylaxis of COVID-19: Results of Interim Analysis. Complement. Ther. Med. 2021, 62, 102768. [Google Scholar] [CrossRef] [PubMed]
  120. Sivan, A.; Vani, G.P.A.; Singh, C.; Purvia, R.P.; Adlakha, M. A Review Article on Aswagandha (Withania somnifera)—The Natural Immunity Booster. IRJAY 2022, 05, 104–109. [Google Scholar] [CrossRef]
  121. Tiwari, R.; Chakraborty, S.; Saminathan, M.; Dhama, K.; Singh, S. Ashwagandha (Withania somnifera): Role in Safegaurding Health, Immunomodulatory Effects, Combating Infections and Therapeutic Applications: A Review. J. Biol. Sci. 2014, 14, 77–94. [Google Scholar] [CrossRef]
  122. Tharakan, A.; Shukla, H.; Benny, I.R.; Tharakan, M.; George, L.; Koshy, S. Immunomodulatory Effect of Withania somnifera (Ashwagandha) Extract—A Randomized, Double-Blind, Placebo Controlled Trial with an Open Label Extension on Healthy Participants. JCM 2021, 10, 3644. [Google Scholar] [CrossRef]
  123. Challacombe, S.J.; Percival, R.S.; Marsh, P.D. Age-Related Changes in Immunoglobulin Isotypes in Whole and Parotid Saliva and Serum in Healthy Individuals. Oral Microbiol. Immunol. 1995, 10, 202–207. [Google Scholar] [CrossRef]
  124. Yen, C.J.; Lin, S.L.; Huang, K.T.; Lin, R.H. Age-Associated Changes in Interferon-Gamma and Interleukin-4 Secretion by Purified Human CD4+ and CD8+ T Cells. J. Biomed Sci. 2000, 7, 317–321. [Google Scholar] [CrossRef]
  125. Yalcin, A.D.; Gorczynski, R.M.; Kahraman, M.S.; Demirel, M.U.; Terzioglu, E. CD40, CD45 CTLA-4 Levels Are Elevated in Healthy Older Adults. Clin. Lab 2012, 58, 449–456. [Google Scholar] [PubMed]
  126. Priyanka, G.; Anil Kumar, B.; Lakshman, M.; Manvitha, V.; Kala Kumar, B. Adaptogenic and Immunomodulatory Activity of Ashwagandha Root Extract: An Experimental Study in an Equine Model. Front. Vet. Sci. 2020, 7, 541112. [Google Scholar] [CrossRef] [PubMed]
  127. Mikolai, J.; Erlandsen, A.; Murison, A.; Brown, K.A.; Gregory, W.L.; Raman-Caplan, P.; Zwickey, H.L. In Vivo Effects of Ashwagandha (Withania somnifera) Extract on the Activation of Lymphocytes. J. Altern. Complement. Med. 2009, 15, 423–430. [Google Scholar] [CrossRef]
  128. Ziauddin, M.; Phansalkar, N.; Patki, P.; Diwanay, S.; Patwardhan, B. Studies on the Immunomodulatory Effects of Ashwagandha. J. Ethnopharmacol. 1996, 50, 69–76. [Google Scholar] [CrossRef] [PubMed]
  129. Clark, R.I.; Salazar, A.; Yamada, R.; Fitz-Gibbon, S.; Morselli, M.; Alcaraz, J.; Rana, A.; Rera, M.; Pellegrini, M.; Ja, W.W.; et al. Distinct Shifts in Microbiota Composition during Drosophila Aging Impair Intestinal Function and Drive Mortality. Cell Rep. 2015, 12, 1656–1667. [Google Scholar] [CrossRef]
  130. Conley, M.N.; Wong, C.P.; Duyck, K.M.; Hord, N.; Ho, E.; Sharpton, T.J. Aging and Serum MCP-1 Are Associated with Gut Microbiome Composition in a Murine Model. PeerJ 2016, 4, e1854. [Google Scholar] [CrossRef]
  131. Purushotham, P.M.; Kim, J.; Jo, E.; Senthil, K. Withanolides against TLR4-Activated Innate Inflammatory Signalling Pathways: A Comparative Computational and Experimental Study. Phytother. Res. 2017, 31, 152–163. [Google Scholar] [CrossRef]
  132. Wong, M.K. COVID-19 Mortality and Progress Toward Vaccinating Older Adults—World Health Organization, Worldwide, 2020–2022. MMWR Morb. Mortal. Wkly. Rep. 2023, 72, 113–118. [Google Scholar] [CrossRef] [PubMed]
  133. Rea, I.M.; Alexander, H.D. Triple Jeopardy in Ageing: COVID-19, Co-Morbidities and Inflamm-Ageing. Ageing Res. Rev. 2022, 73, 101494. [Google Scholar] [CrossRef]
  134. Akbar, A.N.; Gilroy, D.W. Aging Immunity May Exacerbate COVID-19. Science 2020, 369, 256–257. [Google Scholar] [CrossRef]
  135. Bonafè, M.; Prattichizzo, F.; Giuliani, A.; Storci, G.; Sabbatinelli, J.; Olivieri, F. Inflamm-Aging: Why Older Men Are the Most Susceptible to SARS-CoV-2 Complicated Outcomes. Cytokine Growth Factor Rev. 2020, 53, 33–37. [Google Scholar] [CrossRef]
  136. Yang, X.; Yu, Y.; Xu, J.; Shu, H.; Xia, J.; Liu, H.; Wu, Y.; Zhang, L.; Yu, Z.; Fang, M.; et al. Clinical Course and Outcomes of Critically Ill Patients with SARS-CoV-2 Pneumonia in Wuhan, China: A Single-Centered, Retrospective, Observational Study. Lancet Respir. Med. 2020, 8, 475–481. [Google Scholar] [CrossRef] [PubMed]
  137. Fajgenbaum, D.C.; June, C.H. Cytokine Storm. N. Engl. J. Med. 2020, 383, 2255–2273. [Google Scholar] [CrossRef]
  138. Oh, J.H.; Kwon, T.K. Withaferin A Inhibits Tumor Necrosis Factor-α-Induced Expression of Cell Adhesion Molecules by Inactivation of Akt and NF-κB in Human Pulmonary Epithelial Cells. Int. Immunopharmacol. 2009, 9, 614–619. [Google Scholar] [CrossRef]
  139. Piao, L.; Canguo, Z.; Wenjie, L.; Xiaoli, C.; Wenli, S.; Li, L. Lipopolysaccharides-Stimulated Macrophage Products Enhance Withaferin A-Induced Apoptosis via Activation of Caspases and Inhibition of NF-κB Pathway in Human Cancer Cells. Mol. Immunol. 2017, 81, 92–101. [Google Scholar] [CrossRef]
  140. Davis, L.; Kuttan, G. Effect of Withania somnifera On Cytokine Production in Nol and Cyclophosphamide Treated Mice. Immunopharmacol. Immunotoxicol. 1999, 21, 695–703. [Google Scholar] [CrossRef]
  141. Zinatizadeh, M.R.; Zarandi, P.K.; Ghiasi, M.; Kooshki, H.; Mohammadi, M.; Amani, J.; Rezaei, N. Immunosenescence and Inflamm-Ageing in COVID-19. Ageing Res. Rev. 2023, 84, 101818. [Google Scholar] [CrossRef] [PubMed]
  142. Balkrishna, A.; Pokhrel, S.; Singh, H.; Joshi, M.; Mulay, V.P.; Haldar, S.; Varshney, A. Withanone from Withania somnifera Attenuates SARS-CoV-2 RBD and Host ACE2 Interactions to Rescue Spike Protein Induced Pathologies in Humanized Zebrafish Model. DDDT 2021, 15, 1111–1133. [Google Scholar] [CrossRef]
  143. STRING: Functional Protein Association Networks. Available online: https://string-db.org/ (accessed on 25 December 2023).
  144. Otasek, D.; Morris, J.H.; Bouças, J.; Pico, A.R.; Demchak, B. Cytoscape Automation: Empowering Workflow-Based Network Analysis. Genome Biol. 2019, 20, 185. [Google Scholar] [CrossRef] [PubMed]
  145. Panda, V.; Deshmukh, A.; Hare, A.; Singh, S.; Hingorani, L.; Sudhamani, S. Effect of Withania somnifera Hydroalcoholic Extract and Other Dietary Interventions in Improving Muscle Strength in Aging Rats. J. Ayurveda Integr. Med. 2021, 12, 623–632. [Google Scholar] [CrossRef]
  146. Gupta, S.K.; Dua, A.; Vohra, B.P.S. Withania somnifera (Ashwagandha) Attenuates Antioxidant Defense in Aged Spinal Cord and Inhibits Copper Induced Lipid Peroxidation and Protein Oxidative Modifications. Drug Metab. Drug Interact. 2003, 19, 211–222. [Google Scholar] [CrossRef] [PubMed]
  147. Sanap, A.; Chandravanshi, B.; Shah, T.; Tillu, G.; Dhanushkodi, A.; Bhonde, R.; Joshi, K. Herbal Pre-Conditioning Induces Proliferation and Delays Senescence in Wharton’s Jelly Mesenchymal Stem Cells. Biomed. Pharmacother. 2017, 93, 772–778. [Google Scholar] [CrossRef] [PubMed]
  148. Naß, J.; Abdelfatah, S.; Efferth, T. Induction of Stress Resistance and Extension of Lifespan in Chaenorhabditis Elegans Serotonin-Receptor Knockout Strains by Withanolide A. Phytomedicine 2021, 84, 153482. [Google Scholar] [CrossRef] [PubMed]
  149. Widodo, N.; Shah, N.; Priyandoko, D.; Ishii, T.; Kaul, S.C.; Wadhwa, R. Deceleration of Senescence in Normal Human Fibroblasts by Withanone Extracted From Ashwagandha Leaves. J. Gerontol. Ser. Biol. Sci. Med. Sci. 2009, 64A, 1031–1038. [Google Scholar] [CrossRef] [PubMed]
  150. Mácsai, L.; Datki, Z.L.; Csupor, D.; Horváth, A.; Zomborszki, Z.P. Biological Activities of Four Adaptogenic Plant Extracts and Their Active Substances on a Rotifer Model. Evid. -Based Complement. Altern. Med. 2018, 2018, 3690683. [Google Scholar] [CrossRef]
  151. Panossian, A.; Seo, E.-J.; Efferth, T. Novel Molecular Mechanisms for the Adaptogenic Effects of Herbal Extracts on Isolated Brain Cells Using Systems Biology. Phytomedicine 2018, 50, 257–284. [Google Scholar] [CrossRef]
  152. Salvadori, L.; Belladonna, M.L.; Castiglioni, B.; Paiella, M.; Panfili, E.; Manenti, T.; Ercolani, C.; Cornioli, L.; Chiappalupi, S.; Gentili, G.; et al. KYMASIN UP Natural Product Inhibits Osteoclastogenesis and Improves Osteoblast Activity by Modulating Src and P38 MAPK. Nutrients 2022, 14, 3053. [Google Scholar] [CrossRef]
  153. De Rose, F.; Marotta, R.; Talani, G.; Catelani, T.; Solari, P.; Poddighe, S.; Borghero, G.; Marrosu, F.; Sanna, E.; Kasture, S.; et al. Differential Effects of Phytotherapic Preparations in the hSOD1 Drosophila Melanogaster Model of ALS. Sci. Rep. 2017, 7, 41059. [Google Scholar] [CrossRef]
  154. Akhoon, B.A.; Rathor, L.; Pandey, R. Withanolide A Extends the Lifespan in Human EGFR-Driven Cancerous Caenorhabditis Elegans. Exp. Gerontol. 2018, 104, 113–117. [Google Scholar] [CrossRef]
  155. Konar, A.; Gupta, R.; Shukla, R.K.; Maloney, B.; Khanna, V.K.; Wadhwa, R.; Lahiri, D.K.; Thakur, M.K. M1 Muscarinic Receptor Is a Key Target of Neuroprotection, Neuroregeneration and Memory Recovery by i-Extract from Withania somnifera. Sci. Rep. 2019, 9, 13990. [Google Scholar] [CrossRef]
  156. Bhattacharya, S.K.; Bhattacharya, A.; Sairam, K.; Ghosal, S. Anxiolytic-Antidepressant Activity of Withania somnifera Glycowithanolides: An Experimental Study. Phytomedicine 2000, 7, 463–469. [Google Scholar] [CrossRef] [PubMed]
  157. De Rose, F.; Marotta, R.; Poddighe, S.; Talani, G.; Catelani, T.; Setzu, M.D.; Solla, P.; Marrosu, F.; Sanna, E.; Kasture, S.; et al. Functional and Morphological Correlates in the Drosophila LRRK2 Loss-of-Function Model of Parkinson’s Disease: Drug Effects of Withania somnifera (Dunal) Administration. PLoS ONE 2016, 11, e0146140. [Google Scholar] [CrossRef] [PubMed]
  158. Kumar, R.; Gupta, K.; Saharia, K.; Pradhan, D.; Subramaniam, J.R. Withania somnifera Root Extract Extends Lifespan of Caenorhabditis Elegans. Ann. Neurosci. 2013, 20, 13–16. [Google Scholar] [CrossRef]
  159. Raguraman, V.; Subramaniam, J.R. Withania somnifera Root Extract Enhances Telomerase Activity in the Human HeLa Cell Line. ABB 2016, 07, 199–204. [Google Scholar] [CrossRef]
  160. Kaur, J.; Seshadri, S.; Golla, K.H.; Sampara, P. Efficacy and Safety of Standardized Ashwagandha (Withania somnifera) Root Extract on Reducing Stress and Anxiety in Domestic Dogs: A Randomized Controlled Trial. J. Vet. Behav. 2022, 51, 8–15. [Google Scholar] [CrossRef]
  161. Sharma, A.K.; Basu, I.; Singh, S. Efficacy and Safety of Ashwagandha Root Extract in Subclinical Hypothyroid Patients: A Double-Blind, Randomized Placebo-Controlled Trial. J. Altern. Complement. Med. 2018, 24, 243–248. [Google Scholar] [CrossRef] [PubMed]
  162. Verma, N.; Gupta, S.K.; Tiwari, S.; Mishra, A.K. Safety of Ashwagandha Root Extract: A Randomized, Placebo-Controlled, Study in Healthy Volunteers. Complement. Ther. Med. 2021, 57, 102642. [Google Scholar] [CrossRef]
  163. Langade, D.; Thakare, V.; Kanchi, S.; Kelgane, S. Clinical Evaluation of the Pharmacological Impact of Ashwagandha Root Extract on Sleep in Healthy Volunteers and Insomnia Patients: A Double-Blind, Randomized, Parallel-Group, Placebo-Controlled Study. J. Ethnopharmacol. 2021, 264, 113276. [Google Scholar] [CrossRef]
  164. Peñas-LLedó, E.; Terán, E.; Sosa-Macías, M.; Galaviz-Hernández, C.; Gil, J.-P.; Nair, S.; Diwakar, S.; Hernández, I.; Lara-Riegos, J.; Ramírez-Roa, R.; et al. Challenges and Opportunities for Clinical Pharmacogenetic Research Studies in Resource-Limited Settings: Conclusions From the Council for International Organizations of Medical Sciences–Ibero-American Network of Pharmacogenetics and Pharmacogenomics Meeting. Clin. Ther. 2020, 42, 1595–1610.e5. [Google Scholar] [CrossRef]
  165. ClinicalTrials.Gov. Available online: https://clinicaltrials.gov/ (accessed on 26 December 2023).
  166. ANZCTR. Available online: https://www.anzctr.org.au/ (accessed on 26 December 2023).
  167. International Clinical Trials Registry Platform (ICTRP). Available online: https://www.who.int/clinical-trials-registry-platform (accessed on 26 December 2023).
  168. Harris, M. The Impact of Ashwagandha on Stress, Sleep and Food Cravings in College Students: A Mixed Method Double-Blinded Randomized Control Trial. 2022. Available online: https://clinicaltrials.gov/study/NCT05430685?intr=ashwagandha&rank=2 (accessed on 26 December 2023).
  169. Behl, D. A Randomized Placebo-Controlled Trial of Ashwagandha (Withania somnifera) for Cognitive Dysfunction Associated with Cancer Chemotherapy. 2023. Available online: https://clinicaltrials.gov/study/NCT04092647?intr=ashwagandha&rank=5 (accessed on 26 December 2023).
  170. Kulkarni, D.N.S. Effect of Ashwagandha (Withania somnifera) as an Adjunct to Scaling and Root Planing on Salivary Antioxidant and Serum c Reactive Protein Levels in Chronic Generalized Periodontitis—A Randomized Double Blind Clinico-Biochemical Trial. 2018. Available online: https://classic.clinicaltrials.gov/ct2/show/NCT03533972 (accessed on 26 December 2023).
  171. NCT04092647. The Effects of Ashwagandha in Endurance Exercise Performance. 2018. Available online: https://clinicaltrials.gov/study/NCT03596307?intr=ashwagandha&rank=9 (accessed on 26 December 2023).
  172. Abrahao Agessandro Nuclear Factor Kappa Beta Inhibition in Patients with Amyotrophic Lateral Sclerosis: A Phase II Randomized Placebo Controlled Trial. 2022. Available online: https://clinicaltrials.gov/study/NCT05031351?intr=ashwagandha&limit=100&page=1&rank=11 (accessed on 26 December 2023).
  173. Chengappa, K.N.R. Sensoril® (Ashwagandha), an Immunomodulator and Anti-Inflammatory Agent for Schizophrenia: A Parallel Group, Randomized Double Blind, and Placebo Controlled Study. 2017. Available online: https://clinicaltrials.gov/study/NCT01793935?intr=ashwagandha&limit=100&page=1&rank=14 (accessed on 26 December 2023).
  174. Chengappa, K.N.R. Sensoril® (Ashwagandha)—A Standardized Extract From a Medicinal Plant—(Withania somnifera) for Cognitive Enhancement in Persons with Bipolar Disorder: A Parallel Group, Randomized Double Blind, and Placebo Controlled Study. 2016. Available online: https://clinicaltrials.gov/study/NCT00761761?intr=ashwagandha&limit=100&page=1&rank=16 (accessed on 26 December 2023).
  175. Marder, S. Adjunctive Withania somnifera (Ashwagandha) for Persistent Symptoms in People with Schizophrenia. 2023. Available online: https://clinicaltrials.gov/study/NCT03437668?intr=ashwagandha&limit=100&page=1&rank=15 (accessed on 26 December 2023).
  176. Natreon, Inc. A Phase II Double-Blind, Parallel Group, Randomized, Placebo Controlled Clinical Trial of Sensoril® for Patients With Generalized Anxiety Disorder. 2015. Available online: https://clinicaltrials.gov/study/NCT01311180?intr=ashwagandha&limit=100&page=1&rank=17 (accessed on 26 December 2023).
  177. Goldsberry Whitney Combination Therapy with Liposomal Doxorubicin and Withaferin A (Ashwagandha, ASWD) in Recurrent Ovarian Cancer. 2023. Available online: https://clinicaltrials.gov/study/NCT05610735?intr=ashwagandha&limit=100&page=1&rank=18 (accessed on 26 December 2023).
  178. Peters Warren Functional Assessment of Ashwagandaha Root Extract during Weight Loss. 2023. Available online: https://clinicaltrials.gov/study/NCT03112824?intr=ashwagandha&limit=100&page=1&rank=19 (accessed on 26 December 2023).
  179. El-Khodor Bassem A Randomized, Single-Blind, Placebo-Controlled Trial for the Role of a Dietary Supplement in Lowering S-Adenosylhomocysteine (SAH) in Healthy Adults With Elevated Plasma SAH and Normal Homocysteine Levels and Identification of Participants with Elevated Plasma SAH in the General Population Using the MethylQ Score. 2023. Available online: https://clinicaltrials.gov/study/NCT05994794?intr=ashwagandha&limit=100&page=1&rank=20 (accessed on 26 December 2023).
  180. University of Roma La Sapienza Effect on Weight Loss of an Oral Association of Cinnamon Bark (Cinnamomum Cassia) and Withania somnifera in Adult Patients with Overweight or Obesity: A Randomized, Prospective, Placebo-Controlled, Multicenter, Cross-Over, Pilot Study. 2022. Available online: https://clinicaltrials.gov/study/NCT05210218?intr=ashwagandha&limit=100&page=1&rank=21 (accessed on 26 December 2023).
  181. Vedic Lifesciences Pvt Ltd. Efficacy and Tolerability of Sensoril® in Improving Immunity and Thereby Reducing Incidence of Upper Respiratory Tract Infections; 2021. Available online: https://clinicaltrials.gov/study/NCT04733924?intr=ashwagandha&limit=100&page=1&rank=23 (accessed on 26 December 2023).
  182. Koch Klaus Effects of an Adaptogenic Extract on Electrical Activity of Brain in Elderly Subjects with Cognitive Impairment: A Randomized, Double Blind, Placebo-Controlled, Two Arms Cross-over Study. 2019. Available online: https://clinicaltrials.gov/study/NCT03780621?intr=ashwagandha&limit=100&page=1&rank=25 (accessed on 26 December 2023).
  183. Practitioners Alliance Network Treatment of Fibromyalgia and CFS With Ribose, Ashwagandha, Rhodiola, Licorice, Schisandra and Green Tea Extract. 2020. Available online: https://clinicaltrials.gov/study/NCT04598243?intr=ashwagandha&limit=100&page=1&rank=26 (accessed on 26 December 2023).
  184. Kulkarni Vishwesh a Community-Based Participatory Research to Assess the Feasibility of Ayurveda Intervention in Patients with Mild-to-Moderate COVID-19. 2021. Available online: https://clinicaltrials.gov/study/NCT04716647?intr=ashwagandha&limit=100&page=1&rank=27 (accessed on 26 December 2023).
  185. Srivastava Shalini A Randomized, Double Blind, Placebo Controlled, Parallel Group Study to Assess the Effect of Multi-Herb Formulae VL-G-A57 and an Ashwagandha Root Formula (VL-G-E12) on the Modulation of the Hypothalamic-Pituitary-Adrenal Axis (HPA Axis) and Related Symptoms. 2023. Available online: https://clinicaltrials.gov/study/NCT05602389?intr=ashwagandha&limit=100&page=1&rank=29 (accessed on 26 December 2023).
  186. Eckel Robert Protandim and the Metabolic Syndrome. Available online: https://clinicaltrials.gov/study/NCT01125501?intr=ashwagandha&rank=27 (accessed on 17 December 2023).
  187. Srivastava Shalini To Assess the Lanconone® (E-OA-07) Efficacy in Physical Activity-Related Pain-LEAP Study. 2018. Available online: https://clinicaltrials.gov/study/NCT03262805?intr=ashwagandha&limit=100&page=1&rank=32 (accessed on 26 December 2023).
  188. Patil Sandip Assessment of the Effect of StemAlive® Herbal Supplement on the Levels of Circulating Hematopoietic Stem Cells in Human Volunteers. 2014. Available online: https://clinicaltrials.gov/study/NCT02027467?intr=ashwagandha&limit=100&page=1&rank=33 (accessed on 26 December 2023).
  189. Zavorsky, G.S. The Effect of Protandim Supplementation on Oxidative Damage and Athletic Performance. 2019. Available online: https://clinicaltrials.gov/study/NCT02172625?intr=ashwagandha&limit=100&page=1&rank=35 (accessed on 26 December 2023).
  190. Vaisman Nachum Study Details|Study to Assess the Efficacy of Cognitex|ClinicalTrials.Gov. Available online: https://clinicaltrials.gov/study/NCT00719953?intr=ashwagandha&rank=31 (accessed on 17 December 2023).
  191. Nutraceutical Wellness Inc. A Randomized, Double-Blind, Placebo-Controlled Study to Evaluate the Safety and Efficacy of a Nutraceutical Supplement with Standardized Botanicals in Males with Self-Perceived Thinning Hair. 2023. Available online: https://clinicaltrials.gov/study/NCT05339958?intr=ashwagandha&limit=100&page=1&rank=38 (accessed on 26 December 2023).
  192. Ramon Adi Randomized Double Blind Placebo-Controlled Trial to Investigate the Effect of a Botanical Formulation, LLP-01, on Proteomic Inflammatory Biomarkers and Epigenetic Changes. 2023. Available online: https://clinicaltrials.gov/study/NCT06065241?intr=ashwagandha&limit=100&page=1&rank=41 (accessed on 26 December 2023).
  193. Bhat Dr Shreepad Study the Result of Ayurvedic SUVED & Reimmugen (Colostrum) Treatment on Vascular Disease, CAD, CVA, DVT. (SHARP). 2017. Available online: https://clinicaltrials.gov/study/NCT02920125?intr=ashwagandha&limit=100&page=1&rank=43 (accessed on 26 December 2023).
  194. SF Research Institute, Inc. Effects of Ashwagandha Extract (Capsule KSM-66 300 Mg) on Sexual Health in Healthy Women: A Prospective, Randomized, Double-Blind, Placebo-Controlled Study. 2023. Available online: https://clinicaltrials.gov/study/NCT05831241?intr=ashwagandha&limit=100&page=1&rank=4 (accessed on 26 December 2023).
  195. SF Research Institute, Inc. Role of Ashwagandha Extract (Capsule KSM-66 300 Mg) in Improving Sexual Health in Healthy Men: A Prospective, Randomized, Double-Blind, Placebo-Controlled Study. 2023. Available online: https://clinicaltrials.gov/study/NCT05840731?intr=ashwagandha&limit=100&page=1&rank=3 (accessed on 26 December 2023).
  196. Steels, E. A Randomised Placebo-Controlled Trial Investigating Effectiveness of Two Herbal Combinations for Short-Term Stress Management in Healthy Adults; Australian New Zealand Clinical Trials Registry: Camperdown, NSW, Australia, 2023. [Google Scholar]
  197. Lauche, R. Feasibility of Herbal and Nutritional Medicines for Managing Post-Flood Stress and Anxiety: A Randomised Controlled Trial; Australian New Zealand Clinical Trials Registry: Camperdown, NSW, Australia, 2022. [Google Scholar]
  198. Ee, C. Effectiveness of Adjunct Naturopathy for Pregnancy Rates in Women with Diminished Ovarian Reserve Compared to Usual Care Alone: Feasibility of a Randomised Controlled Trial; Australian New Zealand Clinical Trials Registry: Camperdown, NSW, Australia, 2021. [Google Scholar]
  199. Lopresti, A. Examining the Efficacy and Safety of a Novel Standardised Ashwagandha (Withania somnifera) Root Extract in Overweight Middle-to-Older Age Adults Experiencing High Stress and Fatigue: A Randomised, Double-Blind, Placebo-Controlled Trial; Australian New Zealand Clinical Trials Registry: Camperdown, NSW, Australia, 2021. [Google Scholar]
  200. Drummond, P. Effect of Ashwagandha Supplementation on Testosterone Levels and Vitality in Healthy, Overweight Males with Mild to Moderate Symptoms of Fatigue or Reduced Vitality—A Randomised, Double-Blind, Placebo-Controlled Study; Australian New Zealand Clinical Trials Registry: Camperdown, NSW, Australia, 2017. [Google Scholar]
  201. Downey, L. Examining the Effects of Ionix Supreme on Stress, Mood, Energy and Anxiety in Healthy Younger Adults; Australian New Zealand Clinical Trials Registry: Camperdown, NSW, Australia, 2017. [Google Scholar]
  202. Rao, A. A Prospective Double Blinded, Randomised, Placebo-Controlled Study to Evaluate Safety and Efficacy of Two (2) Herbal Formulations in Reducing Menopausal Symptoms in Otherwise Healthy Women; Australian New Zealand Clinical Trials Registry: Camperdown, NSW, Australia, 2015. [Google Scholar]
  203. Multi-disciplinary Research Unit—GSMC A Research Study to Investigate Ashwagandha as a Supplementary Therapy for Treatment of Alcohol Use Disorder. 2022. Available online: https://trialsearch.who.int/Trial2.aspx?TrialID=CTRI/2022/11/047340 (accessed on 26 December 2023).
  204. Dr. Willmar Schwabe India Pvt. Ltd. A Clinical Trial to Study the Effects of Ashwagandha in Patients with Stress and Anxiety Having Cardiovascular Comorbidities. 2023. Available online: https://trialsearch.who.int/Trial2.aspx?TrialID=CTRI/2023/07/054711 (accessed on 26 December 2023).
  205. Sami Sabinsa Group Limited Ashwagandha and Selenium Combination as a Supplement in Patients with Subclinical Hypothyroidism. 2023. Available online: https://trialsearch.who.int/Trial2.aspx?TrialID=CTRI/2023/07/054940 (accessed on 26 December 2023).
  206. Yerram, C.; Jillella, A.; Reddy, V. Effects of Withania somnifera Root Extract Serum Application on Hair Health in Healthy Adults: A Prospective, Double-Blind, Randomized, Parallel, Placebo-Controlled Study. J. Ayurveda Integr. Med. 2023, 14, 100817. [Google Scholar] [CrossRef]
  207. Choudhary, B.; Shetty, A.; Langade, D. Efficacy of Ashwagandha (Withania somnifera [L.] Dunal) in Improving Cardiorespiratory Endurance in Healthy Athletic Adults. AYU 2015, 36, 63–68. [Google Scholar] [CrossRef] [PubMed]
  208. Chandrasekhar, K.; Kapoor, J.; Anishetty, S. A Prospective, Randomized Double-Blind, Placebo-Controlled Study of Safety and Efficacy of a High-Concentration Full-Spectrum Extract of Ashwagandha Root in Reducing Stress and Anxiety in Adults. Indian J. Psychol. Med. 2012, 34, 255–262. [Google Scholar] [CrossRef] [PubMed]
  209. Ambiye, V.R.; Langade, D.; Dongre, S.; Aptikar, P.; Kulkarni, M.; Dongre, A. Clinical Evaluation of the Spermatogenic Activity of the Root Extract of Ashwagandha (Withania somnifera) in Oligospermic Males: A Pilot Study. Evid. Based Complement. Altern. Med. 2013, 2013, 571420. [Google Scholar] [CrossRef] [PubMed]
  210. Wankhede, S.; Langade, D.; Joshi, K.; Sinha, S.R.; Bhattacharyya, S. Examining the Effect of Withania somnifera Supplementation on Muscle Strength and Recovery: A Randomized Controlled Trial. J. Int. Soc. Sports Nutr. 2015, 12, 43. [Google Scholar] [CrossRef] [PubMed]
  211. Dongre, S.; Langade, D.; Bhattacharyya, S. Efficacy and Safety of Ashwagandha (Withania somnifera) Root Extract in Improving Sexual Function in Women: A Pilot Study. BioMed Res. Int. 2015, 2015, 284154. [Google Scholar] [CrossRef] [PubMed]
  212. Choudhary, D.; Bhattacharyya, S.; Joshi, K. Body Weight Management in Adults under Chronic Stress through Treatment with Ashwagandha Root Extract: A Double-Blind, Randomized, Placebo-Controlled Trial. J. Evid. Based Complement. Altern. Med. 2017, 22, 96–106. [Google Scholar] [CrossRef] [PubMed]
  213. Choudhary, D.; Bhattacharyya, S.; Bose, S. Efficacy and Safety of Ashwagandha (Withania somnifera (L.) Dunal) Root Extract in Improving Memory and Cognitive Functions. J. Diet. Suppl. 2017, 14, 599–612. [Google Scholar] [CrossRef] [PubMed]
  214. Langade, D.; Kanchi, S.; Salve, J.; Debnath, K.; Ambegaokar, D. Efficacy and Safety of Ashwagandha (Withania somnifera) Root Extract in Insomnia and Anxiety: A Double-Blind, Randomized, Placebo-Controlled Study. Cureus 2019, 11, e5797. [Google Scholar] [CrossRef] [PubMed]
  215. Kelgane, S.B.; Salve, J.; Sampara, P.; Debnath, K. Efficacy and Tolerability of Ashwagandha Root Extract in the Elderly for Improvement of General Well-Being and Sleep: A Prospective, Randomized, Double-Blind, Placebo-Controlled Study. Cureus 2020, 12, e7083. [Google Scholar] [CrossRef]
  216. Gopal, S.; Ajgaonkar, A.; Kanchi, P.; Kaundinya, A.; Thakare, V.; Chauhan, S.; Langade, D. Effect of an Ashwagandha (Withania somnifera) Root Extract on Climacteric Symptoms in Women during Perimenopause: A Randomized, Double-Blind, Placebo-Controlled Study. J. Obstet. Gynaecol. 2021, 47, 4414–4425. [Google Scholar] [CrossRef]
  217. Chauhan, S.; Srivastava, M.K.; Pathak, A.K. Effect of Standardized Root Extract of Ashwagandha (Withania somnifera) on Well-being and Sexual Performance in Adult Males: A Randomized Controlled Trial. Health Sci. Rep. 2022, 5, e741. [Google Scholar] [CrossRef] [PubMed]
  218. Ajgaonkar, A.; Jain, M.; Debnath, K. Efficacy and Safety of Ashwagandha (Withania somnifera) Root Extract for Improvement of Sexual Health in Healthy Women: A Prospective, Randomized, Placebo-Controlled Study. Cureus 2022, 14, e30787. [Google Scholar] [CrossRef] [PubMed]
  219. Salve, J.; Pate, S.; Debnath, K.; Langade, D. Adaptogenic and Anxiolytic Effects of Ashwagandha Root Extract in Healthy Adults: A Double-Blind, Randomized, Placebo-Controlled Clinical Study. Cureus 2019, 11, e6466. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Active compounds of Withania somnifera (A) Withanolide A; (B) Withaferin; (C) 12-Deoxy-withastramonolide; (D) Withanoside V; (E) Withanone; (F) Withanoside IV; (G) Withanolide B; (H) 27-Hydroxywithanone; (I) Withanoside VI; (J) Physagulin-d. All the chemical structures were drawn using ChemDraw version 20.1.1.125.
Figure 1. Active compounds of Withania somnifera (A) Withanolide A; (B) Withaferin; (C) 12-Deoxy-withastramonolide; (D) Withanoside V; (E) Withanone; (F) Withanoside IV; (G) Withanolide B; (H) 27-Hydroxywithanone; (I) Withanoside VI; (J) Physagulin-d. All the chemical structures were drawn using ChemDraw version 20.1.1.125.
Pharmaceuticals 17 00597 g001aPharmaceuticals 17 00597 g001b
Figure 2. Molecular pathways for Withania somnifera in inflammaging. Withaferin A, an active compound of Withania somnifera, activates NRF2 by dissociating it from Keap1 which translocates to the nucleus and further binds to sMaf leading to transcription of antioxidant HO-1, which reduces oxidative stress. EGF binds to EGFR activating RAS/RAF pathway and phosphorylates ERK /p38/JNK; this further activates NF-κB, which transcribes cytokines that increase inflammation and aging. WAD inhibits p38, JNK, and ERK, while also suppressing ROS. SIRT1 can be activated by Withania somnifera, which inhibits NF-κB. IGF-1 binds to TLR and activates the PI3K/AKT pathway, which activates FOXO and transcribes MnSOD. Ubiquitination that leads to aging can be prevented by Withania somnifera. Also, Withania somnifera increases FOXO activity. Oxidative stress leads to DNA damage and inflammation, which can be reduced by Withania somnifera. Withania somnifera acts on FOXO3A and SIRT3, which helps in increasing cell longevity. Abbreviations: Keap1: Kelch like ECH associated protein 1; NRF2: Nuclear factor erythroid 2-related factor 2; GSK3β: Glycogen synthase kinase 3 beta; HO-1: Heme oxygenase-1; SOD: Superoxide dismutase; sMaf: Small musculoaponeurotic fibrosarcoma; ROS: Reactive oxygen species; ONOO: Peroxynitrite; NO: Nitric oxide; O2.−: Superoxide; L-Arg: Arginine; iNOS: Inducible nitric oxide synthase; COX-2: Cyclooxygenase-2; EGF: Epidermal growth factor; EGFR: Epidermal growth factor receptor; WAD: Withagenin A diglucoside; PTK/PTP: Protein tyrosine kinase/Protein tyrosine phosphatase; RAS: Rat sarcoma; RAF: Rapidly accelerated fibrosarcoma; JNK: c-Jun N-terminal kinase; ERK: Extracellular signal-regulated kinase; NF-κB: Nuclear factor-kappa B; IκB: Nuclear factor-kappa-B inhibitor; IGF-1: Insulin-like growth factor 1; TLR: Toll-like receptor; SIRT: Sirtuin; PI3K: Phosphatidylinositol 3-kinase; PIP2: Phosphatidylinositol 4,5-bisphosphate; PIP3: Phosphatidylinositol-3,4,5-trisphosphate; PDK: Pyruvate Dehydrogenase Kinase; AKT: Protein kinase; BSGK1: Serum/glucocorticoid regulated kinase 1; FOXO: Forkhead box O; NAD+: Oxidized nicotinamide adenine dinucleotide; NADH: Reduced nicotinamide adenine dinucleotide; MnSOD2: Manganese superoxide dismutase.
Figure 2. Molecular pathways for Withania somnifera in inflammaging. Withaferin A, an active compound of Withania somnifera, activates NRF2 by dissociating it from Keap1 which translocates to the nucleus and further binds to sMaf leading to transcription of antioxidant HO-1, which reduces oxidative stress. EGF binds to EGFR activating RAS/RAF pathway and phosphorylates ERK /p38/JNK; this further activates NF-κB, which transcribes cytokines that increase inflammation and aging. WAD inhibits p38, JNK, and ERK, while also suppressing ROS. SIRT1 can be activated by Withania somnifera, which inhibits NF-κB. IGF-1 binds to TLR and activates the PI3K/AKT pathway, which activates FOXO and transcribes MnSOD. Ubiquitination that leads to aging can be prevented by Withania somnifera. Also, Withania somnifera increases FOXO activity. Oxidative stress leads to DNA damage and inflammation, which can be reduced by Withania somnifera. Withania somnifera acts on FOXO3A and SIRT3, which helps in increasing cell longevity. Abbreviations: Keap1: Kelch like ECH associated protein 1; NRF2: Nuclear factor erythroid 2-related factor 2; GSK3β: Glycogen synthase kinase 3 beta; HO-1: Heme oxygenase-1; SOD: Superoxide dismutase; sMaf: Small musculoaponeurotic fibrosarcoma; ROS: Reactive oxygen species; ONOO: Peroxynitrite; NO: Nitric oxide; O2.−: Superoxide; L-Arg: Arginine; iNOS: Inducible nitric oxide synthase; COX-2: Cyclooxygenase-2; EGF: Epidermal growth factor; EGFR: Epidermal growth factor receptor; WAD: Withagenin A diglucoside; PTK/PTP: Protein tyrosine kinase/Protein tyrosine phosphatase; RAS: Rat sarcoma; RAF: Rapidly accelerated fibrosarcoma; JNK: c-Jun N-terminal kinase; ERK: Extracellular signal-regulated kinase; NF-κB: Nuclear factor-kappa B; IκB: Nuclear factor-kappa-B inhibitor; IGF-1: Insulin-like growth factor 1; TLR: Toll-like receptor; SIRT: Sirtuin; PI3K: Phosphatidylinositol 3-kinase; PIP2: Phosphatidylinositol 4,5-bisphosphate; PIP3: Phosphatidylinositol-3,4,5-trisphosphate; PDK: Pyruvate Dehydrogenase Kinase; AKT: Protein kinase; BSGK1: Serum/glucocorticoid regulated kinase 1; FOXO: Forkhead box O; NAD+: Oxidized nicotinamide adenine dinucleotide; NADH: Reduced nicotinamide adenine dinucleotide; MnSOD2: Manganese superoxide dismutase.
Pharmaceuticals 17 00597 g002
Figure 3. Molecular pathways for Withania somnifera in inflammaging modulated by COVID-19 and the microbiome. Microbiome releases LPS that activate TLR4 that initiates the activation of NF-κB pathway via MyD88 and IKBα. Withania somnifera inhibits TLR4 receptor as well as NF-κB. Angiotensinogen converts Ang I to Ang II, while Ang II activates the AT1R receptor; this also leads to activation of NF-κB via pro-oxidants and NOX2. Ang II activates Ang I–VII via the ACE receptor. S-protein of SARS-CoV-2 binds to ACE2 receptor to enter the cell; Withania somnifera inhibits the ACE2 receptor. TNF-α binds to TNFR receptor and activates the TRADD/TRAF pathway, which upregulates AP1 via p38 and transcribes inflammatory cytokines. IL-6 binds to IL-6R and activates the JAK1/STAT3 pathway, which leads to tissue injury by release of inflammatory cytokines. Withania somnifera inhibits TNF-α and IL-6, which help in the suppression of tissue injury. SARS-CoV-2: Severe Acute Respiratory Syndrome Coronavirus 2; DAMP: Damage-Associated Molecular Pattern; PAMP: Pathogen-Associated Molecular Pattern; Ang: Angiotensin; LPS: Lipopolysaccharide; S-Protein: Spike Protein; TLR4: Toll-like Receptor 4; Ang I: Angiotensin I; Ang II: Angiotensin II; Ang I-VII: Angiotensin I-VII; AT1R: Angiotensin II Receptor Type 1; ACE2 receptor: Angiotensin-Converting Enzyme 2 Receptor; IL-6: Interleukin-6; TNF-α: Tumor Necrosis Factor Alpha; IL-6R gp130: Interleukin-6 Receptor Glycoprotein 130; TNF/TNFR complex: Tumor Necrosis Factor/Tumor Necrosis Factor Receptor Complex; MyD88: Myeloid Differentiation Primary Response 88; Pro-oxidant: Substances promoting oxidative stress; TRAF2: Tumor Necrosis Factor Receptor Associated Factor 2; TRADD: Tumor Necrosis Factor Receptor Type 1-Associated DEATH Domain Protein; TRAF6: Tumor Necrosis Factor Receptor Associated Factor 6; p38: mitogen-activated protein kinases; NOX2: NADPH Oxidase 2; JAK1: Janus Kinase 1; TAB1: TGF-beta-Activated Kinase 1-Binding Protein 1; TAB2: TGF-beta-Activated Kinase 1-Binding Protein 2; IKBα: Inhibitor of Nuclear Factor Kappa-B Kinase Subunit Alpha; IKBβ: Inhibitor of Nuclear Factor Kappa-B Kinase Subunit Beta; NF-κB: Nuclear Factor Kappa-Light-Chain-Enhancer of Activated B Cells; STAT3: Signal Transducer and Activator of Transcription 3; AP1: Activator Protein 1; ROS: Reactive Oxygen Species; IL-6: Inflammatory cytokine-6; IL-8: Inflammatory cytokine-8; IL-12: Inflammatory cytokine-12.
Figure 3. Molecular pathways for Withania somnifera in inflammaging modulated by COVID-19 and the microbiome. Microbiome releases LPS that activate TLR4 that initiates the activation of NF-κB pathway via MyD88 and IKBα. Withania somnifera inhibits TLR4 receptor as well as NF-κB. Angiotensinogen converts Ang I to Ang II, while Ang II activates the AT1R receptor; this also leads to activation of NF-κB via pro-oxidants and NOX2. Ang II activates Ang I–VII via the ACE receptor. S-protein of SARS-CoV-2 binds to ACE2 receptor to enter the cell; Withania somnifera inhibits the ACE2 receptor. TNF-α binds to TNFR receptor and activates the TRADD/TRAF pathway, which upregulates AP1 via p38 and transcribes inflammatory cytokines. IL-6 binds to IL-6R and activates the JAK1/STAT3 pathway, which leads to tissue injury by release of inflammatory cytokines. Withania somnifera inhibits TNF-α and IL-6, which help in the suppression of tissue injury. SARS-CoV-2: Severe Acute Respiratory Syndrome Coronavirus 2; DAMP: Damage-Associated Molecular Pattern; PAMP: Pathogen-Associated Molecular Pattern; Ang: Angiotensin; LPS: Lipopolysaccharide; S-Protein: Spike Protein; TLR4: Toll-like Receptor 4; Ang I: Angiotensin I; Ang II: Angiotensin II; Ang I-VII: Angiotensin I-VII; AT1R: Angiotensin II Receptor Type 1; ACE2 receptor: Angiotensin-Converting Enzyme 2 Receptor; IL-6: Interleukin-6; TNF-α: Tumor Necrosis Factor Alpha; IL-6R gp130: Interleukin-6 Receptor Glycoprotein 130; TNF/TNFR complex: Tumor Necrosis Factor/Tumor Necrosis Factor Receptor Complex; MyD88: Myeloid Differentiation Primary Response 88; Pro-oxidant: Substances promoting oxidative stress; TRAF2: Tumor Necrosis Factor Receptor Associated Factor 2; TRADD: Tumor Necrosis Factor Receptor Type 1-Associated DEATH Domain Protein; TRAF6: Tumor Necrosis Factor Receptor Associated Factor 6; p38: mitogen-activated protein kinases; NOX2: NADPH Oxidase 2; JAK1: Janus Kinase 1; TAB1: TGF-beta-Activated Kinase 1-Binding Protein 1; TAB2: TGF-beta-Activated Kinase 1-Binding Protein 2; IKBα: Inhibitor of Nuclear Factor Kappa-B Kinase Subunit Alpha; IKBβ: Inhibitor of Nuclear Factor Kappa-B Kinase Subunit Beta; NF-κB: Nuclear Factor Kappa-Light-Chain-Enhancer of Activated B Cells; STAT3: Signal Transducer and Activator of Transcription 3; AP1: Activator Protein 1; ROS: Reactive Oxygen Species; IL-6: Inflammatory cytokine-6; IL-8: Inflammatory cytokine-8; IL-12: Inflammatory cytokine-12.
Pharmaceuticals 17 00597 g003
Figure 4. Withania somnifera-modulated putative protein–protein interaction network in inflammaging. Protein–protein interaction (PPI) network comprising upregulated and downregulated proteins modulated by Withania somnifera in inflammaging. The PPI with 21 nodes and 262 edges was constructed using STRING database [143] and Cytoscape version 3.10.1 [144]. FOXO3: Forkhead box O3; MAPK8: Mitogen-activated protein kinase 8; AKT1: RAC-alpha serine/threonine-protein kinase; PTGS2: Prostaglandin-endoperoxide synthase 2; MMP-1: Matrix metalloproteinase-1; IL-4: Interleukin-4; TNF: Tumor necrosis factor; IL-8: Interleukin-8; JCHAIN: Joining chain of multimeric IgA and IgM; HSPA4: Heat shock 70 kDa protein 4; TGFβ1: Transforming growth factor beta-1; SOD1: Superoxide dismutase; FCGRT: Fc fragment of IgG receptor and transporter; AIF1: Allograft inflammatory factor 1; IL-6: Interleukin-6; IL-10: Interleukin-10; SIRT3: Sirtuin-3; IFNγ: Interferon-gamma; NOX4: NADPH oxidase 4; IL-12β: Interleukin-12 subunit beta; TLR4: Toll-like receptor 4.
Figure 4. Withania somnifera-modulated putative protein–protein interaction network in inflammaging. Protein–protein interaction (PPI) network comprising upregulated and downregulated proteins modulated by Withania somnifera in inflammaging. The PPI with 21 nodes and 262 edges was constructed using STRING database [143] and Cytoscape version 3.10.1 [144]. FOXO3: Forkhead box O3; MAPK8: Mitogen-activated protein kinase 8; AKT1: RAC-alpha serine/threonine-protein kinase; PTGS2: Prostaglandin-endoperoxide synthase 2; MMP-1: Matrix metalloproteinase-1; IL-4: Interleukin-4; TNF: Tumor necrosis factor; IL-8: Interleukin-8; JCHAIN: Joining chain of multimeric IgA and IgM; HSPA4: Heat shock 70 kDa protein 4; TGFβ1: Transforming growth factor beta-1; SOD1: Superoxide dismutase; FCGRT: Fc fragment of IgG receptor and transporter; AIF1: Allograft inflammatory factor 1; IL-6: Interleukin-6; IL-10: Interleukin-10; SIRT3: Sirtuin-3; IFNγ: Interferon-gamma; NOX4: NADPH oxidase 4; IL-12β: Interleukin-12 subunit beta; TLR4: Toll-like receptor 4.
Pharmaceuticals 17 00597 g004
Table 1. Preclinical studies for Withania somnifera in inflammation and aging.
Table 1. Preclinical studies for Withania somnifera in inflammation and aging.
Sr No.Withania somnifera SourcePreclinical ModelDoseCommentsReferences
1Withania somnifera water-soluble extractNormal Rat kidney Epithelial cells 1:1000 dilution of Withania somnifera capsule containing 2.5% of withanolideWithania somnifera water-soluble extract prevented TNF-α-induced CCL5 increase, reduced CCL2 expression, and inhibited NF-κB activation; also effective against lipopolysaccharide-induced inflammation and aging-related disorders[53]
2Withania somnifera hydro-alcoholic extract Male Sprague Dawley rats500 mg/kg orally (for 60 days)Withania somnifera hydro-alcoholic extract decreased glucose, inflammatory markers (IL-6, TNF-α, CRP), AMPK, malondialdehyde, and Bax (downregulation); upregulated Bcl-2, enhancing muscle health in aging.[145]
3Withania somnifera extractMale albino rats50, 100, and 150 mg/kgWithania somnifera (150 mg/kg) enhances glutathione peroxidase, an antioxidant enzyme, showing potential in protecting against oxidative stress, especially in aging individuals, due to its antioxidant properties[146]
4Withania somnifera root extractWharton’s Jelly Mesenchymal Stem Cells 5, 10, 20, 40, and 50 mg/mLWithania somnifera root extracts (5 mg/mL) upregulated proliferation marker Ki-67, while downregulating senescence marker p21, enhancing Wharton’s Jelly Mesenchymal Stem Cells growth[147]
5Withanolide A dimethyl sulfoxide ExtractCaenorhabditis elegans100 µMWithanolide A effectively reduced ROS levels, increased stress resistance, and extended lifespan in various C. elegans strains, while upregulating serotonin receptor and transporter genes, including the DA1814 strain[148]
6Withanone dimethyl sulfoxide ExtractNormal human fibroblasts (TIG-1, MRC5, and WI-38) cell line5 µg/mLWithanone protects normal cells, downregulates aging marker p21 WAF1, extends lifespan, reduces molecular damage, and upregulates proteasomal activity, suggesting antiaging potential[149]
7Withania somnifera Ethanol extractRotifers (Phylum Rotifera)100 µMWithania somnifera extract contained withaferin A, withanolide A, and withanolide B. Its effects on rotifers included changes in MCF, BSI, and toxicity[150]
8Withania somnifera Root Milk extractT98G neuroglia cells5.0 µg/mLAdaptogens influenced 88 genes linked to stress responses, including neuronal and melatonin pathways, consistently upregulating key genes such as UCN, GNRH1, and RORA, suggesting stress protection and aging-related disorders[151]
9Withania somnifera Dried root extractHuman dermal fibroblasts 25, 50, and 100 µMWithagenin A diglucoside reduces ROS, preserving collagen by inhibiting MMP-1 via MAPK, Akt, c-Jun, COX-2, and NF-κB pathways (downregulation). Withagenin A diglucoside also decreases IL-6 and IL-8 (downregulation), alleviating skin inflammation. These findings suggest Withagenin A diglucoside’s potential in cosmetics and pharmaceuticals for combating skin aging[55]
10Withania somnifera Dried leaf powder extractMiddle-aged female albino rats200 mg/kgReduced anxiety and inflammation (downregulating Iba1, TNF-α, IL-1β, IL-6) in middle-aged rats, while upregulating astrocyte marker Glial fibrillary acidic protein [72]
11Withania somnifera, Silybum marianum and Trigonella foenum-graecum extractRAW 264.7, C2C12 myoblasts and Primary human Osteoblast cells10 µg/mL to 40 µg/mLDietary product of Withania somnifera, Silybum marianum, and Trigonella foenum-graecum extract downregulates TRAP and OC-related genes and inhibits osteoclast formation by downregulating RANK receptor pathways, including Src and p38 MAPK pathways, aiding osteoporosis treatment and bone mineralization[152]
12Withania somnifera Ethanol extractC2C12 cells100 µg/mLWithania somnifera formulation upregulates MyHC-II, promotes protein synthesis via Akt pathway, and enhances myoblast differentiation through p38 MAPK/myogenin pathway, countering muscle atrophy markers[61]
13Withania somnifera leaf hydro-alcoholic extractMale Wistar rats10 mg/kgWithania somnifera restores SIRT1 and NRF2 daily rhythms and phases, indicating potential circadian and antioxidant benefits in aging[81]
14Withania somnifera extractDrosophila melanogaster0.1% w/wWithania somnifera extract improved survival, motor function, and neuronal health, and reduced damaged mitochondria, showing potential therapeutic benefits for Amyotrophic Lateral Sclerosis-like conditions[153]
15Withanolides A dimethyl sulfoxide ExtractCaenorhabditis elegans2, 5, 25, and 50 µMWithanolides A extended C. elegans lifespan (29%), lowered lipofuscin (aging marker), and upregulated neuroprotective factors (SGK-1, DAF-16, SKN-1, HSF-1), signifying antiaging and neuroprotective potential[20]
16Withania somnifera dried root ethanol extractTail tendons of male Wistar rats100 mgWithania somnifera, especially its ethanolic extract, reduced collagen glycation, AGE formation, and cross-linking, potentially through its antioxidant properties, offering therapeutic potential against diabetes and aging[60]
17Withanolides A dimethyl sulfoxide (DMSO) ExtractCaenorhabditis elegans5, 50, 100, 250, and 500 μMWithanolide A at 5 μM upregulated Serum and glucocorticoid regulated kinase-1 in the insulin/insulin-like growth factor-1 pathway, downregulating fat accumulation, and extended lifespan in C. elegans models, showing antiaging potential[154]
18Withania dimethyl sulfoxide (DMSO) ExtractMale Swiss albino strain mice200 mg/kgWithania somnifera upregulated genes such as KLK8 and factors such as MAP2c, enhancing dendritic growth, memory, and neuroprotection while reducing age-related neurodegeneration[155]
19Withania somnifera root extractCharles Foster strain male rats20 mg/kg and 50 mg/kgThe study supported Withania somnifera for mood stabilization, attributing anxiolytic and antidepressant effects to GABA-mimetic activity, aligning with clinical use in Ayurveda[156]
20Withania somnifera rootMethanol extractDrosophila melanogaster males0.1, 1 and 10% w/wThe study employed LRRK2WD40 mutant fruit flies as a marker for Parkinson’s disease, showing motor impairments and mitochondrial dysfunction. Withania somnifera root upregulated protective factors but also downregulated toxic ones[157]
21Withaferin A methanol extractSH-SY5Y cells50 nM to 1 µMThe study identified markers (NF-κB, inflammasome), genes (HDAC2), and factors involved in Alzheimer’s disease. NF-κB and inflammasome genes were upregulated, while HDAC2 was downregulated[90]
22Withania somnifera root extract Caenorhabditis elegans100 ng/mlWithania somnifera root extract extended C. elegans lifespan, particularly in acr-16 mutants[158]
23Withania somnifera root extract powder (KSM-66)HeLa cell line10–50 µg/mLWithania somnifera root extract enhances telomerase in HeLa cells, indicating potential antiaging effects[159]
24Withania somnifera root extract (KSM-66)Dogs15 mg/kg Withania somnifera root extract might alleviate stress, anxiety, pain and enhance well-being in dogs[160]
Table 2. Clinical trials of Withania somnifera in inflammation and aging (adapted from www.clinicaltrials.gov (accessed on 26 December 2023) [165], ANZCTR www.anzctr.org.au/ (accessed on 26 December 2023) [166], and ICTRP www.who.int/clinical-trials-registry-platform (accessed on 26 December 2023) [167].
Table 2. Clinical trials of Withania somnifera in inflammation and aging (adapted from www.clinicaltrials.gov (accessed on 26 December 2023) [165], ANZCTR www.anzctr.org.au/ (accessed on 26 December 2023) [166], and ICTRP www.who.int/clinical-trials-registry-platform (accessed on 26 December 2023) [167].
Registry ID NumberStatusPhaseNumber of ParticipantsConditions or DiseaseObjectiveDoseReference
NCT05430685CompletedPhase 260Craving, stress, sleep, well being Effect of Withania somnifera in sleep, stress, and food cravings of healthy college students 350 mg twice daily for 30 days [168]
NCT04092647RecruitingPhase 280Chemo FogRole of Withania somnifera in cognitive dysfunction350 mg for 9 weeks[169]
(Kulkarni, 2018)CompletedPhase 1 & 250Chronic PeriodontitisRole of Withania somnifera in serum C reactive protein and salivary antioxidant in chronic generalized periodontitis500 mg twice daily for 1 month[170]
NCT03596307UnknownNA12HealthyRole of Withania somnifera in endurance exercise performance180 mg for 2 days and 16 days[171]
NCT05031351RecruitingPhase 275Amyotrophic lateral SclerosisSafety of Withania somnifera in participants with amyotrophic lateral sclerosis1088 mg and 544 mg for 8 weeks [172]
NCT01793935CompletedNA68Schizophrenia, schizoaffective disorderTo determine whether Withania somnifera can reduce psychopathology scores and stress scores in schizophrenia 500 mg for first week followed by 1000 mg for 12 weeks[173]
NCT00761761CompletedPhase 360Bipolar disorderEffect of Withania somnifera in bipolar disorder250 mg for first week followed by 500 mg for 8 weeks[174]
NCT03437668Active, not recruiting Phase 2 & 366SchizophreniaRole of Withania somnifera in persistent symptoms of schizophrenia500 mg for 12 weeks[175]
NCT01311180CompletedPhase 2120Generalized anxiety disorder Role of Withania somnifera in generalized anxiety disorder 250 mg in morning for 7 days and 250 mg twice a day for 7 weeks[176]
NCT05610735Not yet recruiting Phase 1 & 272Recurrent ovarian cancer To determine the tolerance of Withania somnifera with liposomal doxorubicinAdministration of liposomal doxorubicin 40 mg/m2 on day 1 of a 28-day cycle for 4 cycles; 2 g, 4 g, and 8 g of Withania somnifera for 2 years[177]
NCT03112824CompletedNA35Craving, obesity, sleep disturbance, stress reactionRole of Withania somnifera in weight loss300 mg of Withania somnifera twice a day for 12 weeks[178]
NCT05994794Active, not recruiting NA40Elevated S-adenosylhomocysteineRole of dietary supplement of normal homocysteine levels in healthy adultsDietary supplement of Withania somnifera, creatine, and apha-GPC[179]
NCT05210218Completed NA 40Obesity Dietary supplement of cinnamon and Withania somnifera in weight loss Cinnamon and Withania somnifera (300 mg and 150 mg) for 4 weeks[180]
NCT04733924CompletedNA77Immune health Investigate the efficacy and tolerability in improve immunity and reducing respiratory tract infection 125 mg and 250 mg for 84 days[181]
NCT03780621CompletedPhase 116Cognitive impairmentEffect of adaptogenic extract on cognitive impairment550 mg Andrographis and 10 mg of withanolides[182]
NCT04598243Unknown statusEarly Phase 170Fibromyalgia, chronic fatigue syndromeRibose, Withania somnifera, Rhodiola, Licorice, Schisandra, and Green Tea Extract for treating fibromyalgia and chronic fatigue syndromeHerbal combination of Ribose, Withania somnifera, Rhodiola, liquorice, Schisandra, and Green Tea Extract [183]
NCT04716647CompletedNA28COVID-19Investigate the feasibility of ayurveda in treating COVID-19Withania somnifera: 250 mg to 5 g; Giloy: 500 mg to 1 g; Tulsi: 500 mg−1g[184]
NCT05602389CompletedNA186StressInvestigate the role of multi-herb formulae and Withania somnifera root formula in stress 700 mg daily for 60 days[185]
NCT01125501WithdrawnNA 0Metabolic SyndromeEvaluate the effects of Protandim on protein profile changes and markers of inflammation and oxidation in subjects (40–60 years of age) with metabolic syndromeOne capsule containing protandim (derived from the botanical extracts Bacopa monniera, Silybum marianum, Withania somnifera, Camellia sinensis, and Curcuma longa) daily for 30 days[186]
NCT03262805CompletedNA73Knee OsteoarthritisEvaluate the efficacy of Lanconone® (E-OA-07) in physical activity-related pain—LEAP study1000 mg twice daily of Lanconone (Shyonak, Withania somnifera, Shunthi, Guggul, Chopchini, Rasna, and Shallaki) for 4 weeks [187]
NCT02027467CompletedNA23HealthyInvestigate the effect of StemAlive® supplement on the levels of stem cells in human volunteers (hematoalive)Three capsules of StemAlive® (green tea, Withania somnifera) twice daily for 14 days [188]
NCT02172625CompletedNA40Oxidative StressTo examine the impact of Protandim supplementation on oxidative damage and athletic performanceOne pill daily for 90 days of pritandim (675 mg) containing Bacopa extract 150 mg; milk thistle 225 mg; Withania somnifera 150 mg; green tea 75 mg; turmeric 75 mg[189]
NCT00719953CompletedPhase 430Elderly memory impairmentTo assess the efficacy of CognitexOne capsule thrice a day containing 600 mg GPC, 100 mg PS-omega 3, 20 mg vinpocetine, 50 mg uridine-5′-monophosphate (disodium), 550 mg plant extracts (150 mg wild blueberry, 125 mg Withania somnifera, 150 mg grape seed, 125 mg hops, ginger and rosemary)[190]
NCT05339958CompletedNA112Hair Thinning To assess the effectiveness and safety of a novel dietary supplement containing botanical ingredients for hair thinning in men over the course of six months of continuous daily usageFour capsules of Nutrafol® Men (Sensoril Withania somnifera, BCM-95 BioCurcumin, USPlus Saw Palmetto, EVNolMax 20% Tocotrienol/Tocopherol complex, Bioperine (piperine), Cynatine HNS (solubilized keratin), and Capsimax (capsaicin)) once daily for 6 months [191]
NCT06065241Active, not recruitingNA40Aging InflammationTo investigate whether the botanical formulation, LLP-01, has a significant clinical effect on proteomic inflammatory biomarkers and epigenetic changes in healthy, older individualsTwo LLP-01 capsules containing 1000 mg (extracts from Withania somnifera, Rosmarinus officinalis, Curcuma longa, Cotinus coggygria, Panax ginseng, Cordyceps militaris, Camellia sinensis, Cotinus coggygria, and Piper nigrum) for 60 days[192]
NCT02920125CompletedPhase 396Coronary artery disease, cerebrovascular disease, ischemic heart disease, deep vein thrombosis, peripheral arterial diseases, and vascular diseaseTo evaluate result of Ayurvedic SUVED & Reimmugen (Colostrum) treatment on vascular disease, CAD, CVA, and DVT500 mg of SUVED capsule (Terminalia Arjuna, Withania somnifera, Terminalia chebula, Cyperus rotundus, Apium graveolens, Vitis vinifera, Piper longum, Fagonia Arabica, Emblica officinalis, Terminalia belerica, Nymphaea stellate, Punica granatum, Bacopa Monnieri) for 3 months [193]
NCT05831241Recruiting Phase 445Sexual healthTo determine the effect of Withania somnifera extract (Capsule KSM-66) on sexual health in healthy women300 mg twice daily of KSM-66 capsule for 8 weeks[194]
NCT05840731RecruitingPhase 445Sexual healthTo investigate the role of Withania somnifera extract (Capsule KSM-66) in improving sexual health in healthy men300 mg twice daily of KSM-66 capsule for 8 weeks[195]
ACTRN12623000287639pNot yet recruitingPhase 2100StressTo investigate effectiveness of herbal formulations for stressCompare stress reduction: three tablets (Ziziphus, Passionflower, Lemon balm, Chamomile) vs. twp tablets (Withania somnifera, stigma) daily for 21 days[196]
ACTRN12622001226796Not yet recruitingNA100Stress, anxietyTo assess the feasibility of herbal and nutritional medicines for managing post-flood stress and anxiety: a randomized controlled trialEvaluate post-flood stress: Noble Kava (200 mg daily, two capsules twice), Executive B stress Formula (two tablets daily), Withania somnifera complex day (four tablets daily).[197]
ACTRN12621001769875Active, not recruitingNA40Diminished ovarian reserve, infertilityTo assess the effectiveness of adjunct naturopathy for pregnancy rates in women with diminished ovarian reserve compared to usual care alone: feasibility of a randomized controlled trialAssessing naturopathic practice for women with diminished ovarian reserve: four consultations, five supplements (including Withania somnifera), a d personalized herbal medicine for 16 weeks.[198]
ACTRN12621001551886CompletedNA120High stress, fatigue, inflammationTo examine the efficacy and safety of a novel standardized Withania somnifera root extract in overweight middle-to-older age adults experiencing high stress and fatigue: a randomized, double-blind, placebo-controlled trialWithania somnifera extract (one capsule taken orally, twice daily with or without food, delivering 400 mg a day for 12 weeks)[199]
ACTRN12617000971336CompletedNA50Energy and vitality, hormonal changes, wellbeingTo examine the impact of Withania somnifera supplementation on testosterone levels and vitality in healthy, overweight menTwo tablets daily (containing 10.5 mg of withanolide glycosides) for 8 weeks[200]
ACTRN12617000698370CompletedNA78Stress, cognitive performance, anxietyTo examine the effects of Ionix Supreme on stress, mood, energy, and anxietyIonix Supreme liquid Oral liquid, 60 mL daily for 4 weeks, ingredients include Chinese Wolfberry, Withania somnifera, vitamins and minerals[201]
ACTRN12615000324516Active, not recruitingPhase 3/Phase 4180MenopauseTo evaluate the safety and efficacy of two herbal formulations in reducing menopausal symptoms in otherwise healthy women.Capsule Twice daily: Tinospora, Asparagus, Withania somnifera, Commiphora. Fenugreek capsule, 300mg extract, twice daily for 3 months[202]
CTRI/2022/11/047340Not Yet RecruitingNA60Alcohol related disordersTo investigate Withania somnifera as a supplementary therapy for treatment of alcohol use disorderWithania somnifera 250 mg two tablets before bedtime for 90 days[203]
CTRI/2023/07/054711CompletedPhase 360Generalized anxiety disorderTo study the effects of Withania somnifera in patients with stress and anxiety having cardiovascular comorbiditiesWithania somnifera: 300 mg twice a day after meals[204]
CTRI/2023/07/054940Open to RecruitmentPhase 3/ Phase 460Iodine-deficiency hypothyroidismTo evaluate the safety and efficacy of Withania somnifera and selenium combination as a supplement in patients with subclinical hypothyroidismWithania somnifera 500 mg and selenium 40 mcg: one capsule once a day after dinner for 90 days[205]
CTRI/2022/11/047539CompletedNA68Hair health Safety and efficacy of Withania somnifera on hair health One to two drops of Withania somnifera topical formulation daily for 75 days[206]
-CompletedNA50Quality of life and cardiorespiratory endurance Efficacy of Withania somnifera in improving quality of life and cardiorespiratory endurance in human athletic adults300 mg twice daily of KSM-66 capsule for 12 weeks [207]
-Completed -64Stress and anxiety To assess the safety and efficacy of a high concentration full-spectrum extract of Withania somnifera root in reducing stress and anxiety in adults300 mg Withania somnifera or placebo capsule twice daily for 60 days [208]
-Completed-68Male infertility To evaluate Withania somnifera impact on sperm production in oligospermic males through a clinical pilot study 225 mg Withania somnifera capsule thrice daily for 12 weeks [209]
-Completed -50Muscle strength To examine the effect of Withania somnifera supplementation on muscle strength and recovery300 mg twice daily for 8 weeks [210]
CTRI/2015/07/006045Completed-50Female sexual dysfunction To determine the efficacy and safety of a high concentration Withania somnifera root extract (HCARE) supplementation for improving sexual function in healthy females300 mg twice daily for 8 weeks [211]
Completed-52Chronic stressTo evaluate the safety and efficacy of a standardized root extract of Withania somnifera through a double-blind, randomized, placebo-controlled trial300 mg twice daily for 8 weeks [212]
CTRI/2019/11/021990Completed-50Mild cognitive impairmentTo evaluate the efficacy and safety of Withania somnifera in improving memory and cognitive functioning in adults with mild cognitive impairment 300 mg twice daily for 8 weeks [213]
CTRI/2016/05/006903Completed-50Subclinical hypothyroidism To evaluate the efficacy and safety of Withania somnifera root extract in subclinical hypothyroid patients300 mg twice daily for 8 weeks [161]
-Completed-60Insomnia and anxietyTo determine the efficacy and safety of Withania somnifera root extract in patients with insomnia and anxiety 300 mg twice daily for 10 weeks [214]
-Completed-50Mental alertnessTo assess the safety, efficacy, and tolerability of Withania somnifera root extract on the improvement of general health and sleep in elderly people300 mg twice daily for 12 weeks [215]
CTRI/2016/04/006791Completed-50Cardiorespiratory enduranceTo evaluate the efficacy and safety of Withania somnifera root extract in enhancing cardiorespiratory endurance in healthy athletic adults300 mg twice daily for 8 weeks [26]
CTRI/2019/10/021547Completed-100PerimenopauseTo assess the efficacy and tolerability of an Withania somnifera root extract on the climacteric symptoms, quality of life, and hormonal parameters in perimenopausal women300 mg twice daily for 8 weeks [216]
CTRI/2016/05/006906Completed-50Overall well-being To evaluate the effect of Withania somnifera root extract on improving sexual health in adult males300 mg twice daily for 8 weeks [195,217]
-Completed-80Poor sexual functionTo evaluate the efficacy and safety of standardized Withania somnifera root extract in improving sexual function in healthy females300 mg twice daily for 8 weeks [218]
-Completed-75Stress, anxiety, and impeded sleepTo investigate the adaptogenic and anxiolytic effects of Withania somnifera root extract in healthy adults 250 mg/day and 600 mg/day twice daily for 8 weeks[219]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Basudkar, V.; Gujrati, G.; Ajgaonkar, S.; Gandhi, M.; Mehta, D.; Nair, S. Emerging Vistas for the Nutraceutical Withania somnifera in Inflammaging. Pharmaceuticals 2024, 17, 597. https://doi.org/10.3390/ph17050597

AMA Style

Basudkar V, Gujrati G, Ajgaonkar S, Gandhi M, Mehta D, Nair S. Emerging Vistas for the Nutraceutical Withania somnifera in Inflammaging. Pharmaceuticals. 2024; 17(5):597. https://doi.org/10.3390/ph17050597

Chicago/Turabian Style

Basudkar, Vivek, Gunjan Gujrati, Saiprasad Ajgaonkar, Manav Gandhi, Dilip Mehta, and Sujit Nair. 2024. "Emerging Vistas for the Nutraceutical Withania somnifera in Inflammaging" Pharmaceuticals 17, no. 5: 597. https://doi.org/10.3390/ph17050597

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop