Hostname: page-component-7dd5485656-bvgqh Total loading time: 0 Render date: 2025-10-27T16:48:18.538Z Has data issue: false hasContentIssue false

The effect of Withania somnifera (Ashwagandha) on mental health symptoms in individuals with mental disorders: systematic review and meta-analysis

Published online by Cambridge University Press:  27 October 2025

Mattia Marchi
Affiliation:
Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy Department of Mental Health and Drug Abuse, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
Pietro Grenzi
Affiliation:
Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
Antonio Travascio
Affiliation:
Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy Department of Mental Health and Drug Abuse, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
Daniele Uberti
Affiliation:
Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy Department of Mental Health and Drug Abuse, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
Edoardo De Micheli
Affiliation:
Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
Fabio Quartaroli
Affiliation:
Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
Giuseppe Laquatra
Affiliation:
Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
Luca Pingani
Affiliation:
Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy Department of Mental Health and Drug Abuse, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
Silvia Ferrari
Affiliation:
Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy Department of Mental Health and Drug Abuse, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
Gian M. Galeazzi*
Affiliation:
Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy Department of Mental Health and Drug Abuse, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
*
Correspondence: Gian M. Galeazzi. Email: gianmaria.galeazzi@unimore.it.
Rights & Permissions [Opens in a new window]

Abstract

Background

Withania somnifera (WS) is considered an adaptogen agent with reported antistress, cognition facilitating and anti-inflammatory properties, which may be beneficial in the treatment of mental disorders.

Aims

This systematic review investigated the efficacy and tolerability of Withania somnifera for mental health symptoms in individuals with mental disorders.

Method

The protocol of this review was registered with PROSPERO (CRD42023467959). PubMed, Scopus, PsycINFO, CINAHL, Embase and CENTRAL were searched for randomised controlled trials comparing Withania somnifera to any comparator, in people of any age, with any mental disorder. The meta-analyses were based on standardised mean differences (SMDs) and odds ratios with 95% confidence intervals, estimated through frequentist and Bayesian-hierarchical models with random-effects.

Results

Fourteen studies, corresponding to 360 people treated with Withania somnifera and 353 controls were included. Anxiety disorders were the predominant diagnostic category. Thirteen trials administered Withania somnifera orally (median dose 600 mg/day), one with Shirodhara therapy. The median follow-up time was 8 weeks. Although limited by the small number of studies, substantial between-study heterogeneity, and outlier effects, our investigation showed Withania somnifera effectiveness in improving anxiety (outlier-corrected SMD: −1.13 (95% CI: −1.65; −0.60), pooled SMD: −1.962 (95% CI: −2.66; −0.57)), depression (SMD: −1.28 (95% CI: −2.40; −0.16) and stress (SMD: −0.95 (95% CI: −1.46; −0.43) symptoms and sleep quality (SMD: −1.35 (95% CI: −1.79; −0.91). The effect size was confirmed using the Bayesian for anxiety but not for depression. No significant difference between Withania somnifera and the comparators was found for safety and tolerability.

Conclusions

We found evidence supporting the effectiveness of Withania somnifera in treating anxiety symptoms. Future trials should replicate this finding in larger samples and further clarify a possible Withania somnifera role in depression and insomnia treatment.

Information

Type
Review
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
© The Author(s), 2025. Published by Cambridge University Press on behalf of Royal College of Psychiatrists

Withania somnifera is a perennial herbaceous plant, belonging to the ‘Solanaceae’ family and native to the Indian areas, which then spread also to Eastern Asia, Southern and Northern Equatorial Africa and Southern-Central Europe. Reference Kulkarni and Dhir1

Although the scientific name of the species is Withania somnifera, the plant has been referred to with different names. Probably the most popular name is Ashwagandha which means ’smell of horse’ in Indian dialect (‘aswa’ meaning ‘horse’ and ‘gandha’ meaning ’smell’) in allusion both to the smell of this animal and to the qualities of the herbal extract which can improve strength, vitality and tonicity. Reference Deocaris, Widodo, Wadhwa and Kaul2,Reference Modak, Dixit, Londhe, Ghaskadbi and Devasagayam3 The plant is also popularly known in India by different vernacular names like Punir (Hindi), Ashvaganda (Bengal, Bombay), Aksan (Punjab), Amukkira (Tamil) and Tilli (Marathi). Withania somnifera is also often referred to as the ‘Indian Ginseng’. Reference Kulkarni and Dhir1 The phytotherapeutic principles of Withania somnifera have been discovered and used by Indian and Asian medicine since ancient times; in fact, for thousands of years this plant has been a natural remedy widely used in Ayurvedic and traditional Indian and Asian medicine. Reference Deocaris, Widodo, Wadhwa and Kaul2,Reference Singh, Bhalla, de Jager and Gilca4

Sold as natural supplements, the active constituents can be obtained from the roots, the bark, leaves, seeds and fruits, and used to prepare infusions, tonic and energetics drinks, and in the form of capsules, tablets or drops. The various products based on Withania somnifera can be found in herbalists or directly in natural-remedies specialised shops, at variable prices. Alternatively, it is possible to buy Withania somnifera supplements by taking advantage of online shopping. Commercial extracts are generally not titled. Reference Gaurav, Yadav, Maurya, Yadav, Yadav and Shukla5 The major biochemical constituents of Withania somnifera are steroidal alkaloids and lactones, a class of constituents known as withanolides (steroidal lactones with an ergostane skeleton). The withanolides have a structural resemblance with the active constituents of the plant called ‘Panax ginseng’ and are therefore known as ginsenosides. Reference Kulkarni and Dhir1,Reference Gaurav, Yadav, Maurya, Yadav, Yadav and Shukla5

According to available evidence, the Withania somnifera root extract appears to be practically non-toxic. Previous research reported excellent tolerability of Withania somnifera after weeks of continuous use, with no significant difference in the incidence of adverse effects compared to a placebo. Reference Mandlik Ingawale and Namdeo6,Reference Brunetti, Lo Faro, Tini, Busardò and Carlier7 Withania somnifera is classified as an adaptogen; in fact its phytocomplex could modulate the main dopaminergic and serotonergic neurotransmitters, even if further research is needed to verify the specific mechanism of this activity. Reference Dar, Hamid and Ahmad8 Withania somnifera preparations are also reported to modulate the γ-amino-butyric acid (GABA) transmission and cholinergic neurotransmission, involved in many central nervous system-related disorders. Reference Dar, Hamid and Ahmad8,Reference Sarris, McIntyre and Camfield9 The active constituents of the plant (withaferin A, sitoindosides VII–X) are reported to have an antioxidant activity which may contribute, at least in part, to the reported antistress, immunomodulatory, cognition facilitating, anti-inflammatory and anti-ageing properties. Reference Kulkarni and Dhir1,Reference Mácsai, Datki, Csupor, Horváth and Zomborszki10 Given its longstanding use in traditional medicine, it has been used for a wide range of activities for different health conditions. In psychiatry, its supplementation has been tested to treat anxiety, insomnia and stress-induced feelings of depression. Reference Zhang, Yan, Wu, Yang, Zhu and Yan11Reference Mikulska, Malinowska, Ignacyk, Szustowski, Nowak and Pesta14 A recent review Reference Singh, Bhalla, de Jager and Gilca4 reported that Withania somnifera induced a calming anxiolytic effect that was comparable to lorazepam in three standard anxiety tests, and a previously published, comprehensive, meta-analysis of nutraceutical and phytoceutical compounds in psychiatric disorders suggested Withania somnifera for the treatment of anxiety and stress, although with a low level of evidence. Reference Sarris, Ravindran, Yatham, Marx, Rucklidge and McIntyre15 The investigations also support the use of Withania somnifera as a mood stabiliser in clinical conditions of anxiety and depression. Reference Singh, Bhalla, de Jager and Gilca4 Furthermore, the results obtained in a clinical trial and reported in a recent review suggested that the extract of this plant might be effective in combination with standard antidepressants in the treatment of obsessive–compulsive disorder (OCD). Reference Ayati, Sarris, Chang, Emami and Rahimi16,Reference Jahanbakhsh, Manteghi, Emami, Mahyari, Gholampour and Mohammadpour17 In addition, Withania somnifera showed promise as a safe and effective treatment for improving immediate and general memory, executive function, attention and information-processing speed in patients with mild cognitive impairment, suggesting possible use in Alzheimer’s disease. Reference Kumar, Phaneuf and Julien18Reference Ng, Loke, Foo, Tan, Chan and Lim20

Despite the promising properties of Withania somnifera, its use in clinical practice has received little attention. Crucially, existing systematic reviews on Withania somnifera use in psychiatry have focused on specific disorders and included a limited number of studies. This systematic review aims to bridge this gap by investigating the safety and efficacy of Withania somnifera in the treatment of mental health symptoms of people with mental disorders. By providing a comprehensive summary of the existing evidence, our study aims to provide valuable insights to potentially inform future research and advance mental health treatment options.

Method

This systematic review and meta-analysis was performed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Reference Page, McKenzie, Bossuyt, Boutron, Hoffmann and Mulrow21 The protocol of this study was registered with PROSPERO (CRD42023467959).

Search strategy and selection criteria

We searched the PubMed (Medline), Scopus, PsycINFO, CINAHL, Embase and CENTRAL (covering also the International Clinical Trials Registry Platform (ICTRP) and clinicaltrials.gov) databases until 15 May 2025, using the strategy outlined in Supplemental Table 1 available at https://doi.org/10.1192/bjo.2025.10885. No restrictions regarding language of publication or publication date were set. All randomised controlled trials (RCTs) comparing Withania somnifera used as a monotherapy or as an add-on treatment to placebo or other active comparators, in people of any age, with any mental disorder were eligible for the review. Diagnosis was defined according to standard operational diagnostic criteria according to the DSM 22 or ICD 23 or validated psychometric tools. Studies were excluded if the Population, Intervention, Comparison, Outcomes and Study framework did not fit with that defined in the review protocol in PROSPERO (CRD42023467959). Specifically, studies that considered a sample of healthy volunteers were excluded. Similarly, participants with exclusively physical conditions, dementia or substance use disorder were excluded, unless they had a co-occurring mental disorder. Studies where all participants received at least one dose of Withania somnifera, or that did not provide post-treatment assessment of psychopathology were also excluded. No other limits on participants’ characteristics, concurrent treatment or comorbidity were set. We excluded qualitative studies, case reports, case series and reviews, although the reference lists of the reviews have been screened to identify any potentially eligible studies that could have been missed during the literature search. We only included studies published in peer-reviewed journals, excluding conference abstracts and dissertations. If data from the same trial were published in multiple papers, we considered only the publication reporting more complete information or, in case of parity in this criterion, the largest sample size, to maximise the power of the analyses. Sample overlap was ruled out through a careful check of the trial registration codes as well as the place and year(s) of sampling.

Data collection and extraction

Two review authors (M.M. and P.G.), working independently, screened all retrieved articles in the original search for inclusion, first on the title, followed by the abstract and then analysed full texts to check compliance with inclusion/exclusion criteria. All disagreements were explored until consensus was reached, and if consensus was not possible, another member of the team was consulted (G.M.G.).

Data extraction started on 12 December 2023. For each eligible trial, the two review authors independently extracted the following information: (a) study characteristics (first author last name, year of publication, country, study setting, number of participants randomised in each arm, number of participants with outcome assessment and duration of the trial); (b) participant characteristics (age, gender, psychiatric diagnoses and stage of illness, symptoms severity at baseline and on-going psychiatric treatment); (c) intervention details (comparator used, prescribed Withania somnifera dosage and bioactives’ concentration in the Withania somnifera extracts, frequency of administration, route of administration and co-interventions) and (d) outcome measures of interest and time of data collection. Extraction sheets for each study were cross-checked for consistency and any disagreement was resolved by discussion within the research group.

Outcome measures

For our primary analyses we considered efficacy, which we measured as the difference in the post-treatment score of mental health symptoms among treatment and control arms. Finally, we considered tolerability as: the rate of dropouts due to any cause, the rate of dropouts due to serious adverse effects, the overall rate of side effects and the rate of death, in each trial arm. We assessed those outcomes available at the times closest to the median follow-up time across the studies.

Statistical analyses

Where possible, we summarised quantitative data among studies using meta-analyses. We used inverse-variance models with random effects to summarise both continuous and dichotomous outcome data. Reference DerSimonian and Laird24 For continuous outcome data, we calculated the Hedges’ g standardised mean differences (SMDs) and the corresponding 95% confidence intervals; for dichotomous outcome data, we calculated the pooled odds ratios and the corresponding 95% CIs. Reference Higgins, Li, Deeks, Higgins, Thomas, Chandler, Cumpston, Li and Page25 We used data from the intention-to-treat analyses for both continuous and dichotomous outcomes. The results were summarised using forest plots. Standard Q tests and the I 2 statistic (i.e. the percentage of variability in prevalence estimates attributable to heterogeneity rather than sampling error or chance, with values of I 2 ≥75% indicating high heterogeneity) were used to assess between-study heterogeneity. Reference Higgins and Thompson26

When the meta-analysis included at least ten studies, Reference Sterne, Sutton, Ioannidis, Terrin, Jones and Lau27 we performed funnel plot analysis and the Egger test to test for publication bias. If analyses showed a significant risk of publication bias, we would use the trim-and-fill method to estimate the number of missing studies and the adjusted effect size. Reference Duval and Tweedie28Reference Sterne, Egger, Moher, Higgins and Green31 For meta-analysis including at least ten studies, univariable meta-regression analysis was performed to examine sources of between-study heterogeneity on a range of study pre-specified characteristics (i.e. participants’ gender, age in the entire sample; country where the study was conducted; trial duration; Withania somnifera dose; concentration of withanolides (i.e. the main active component of Withania somnifera extracts) and concurrent medication). Sensitivity analyses included leave-one-out and subgroup meta-analyses using diagnosis as the grouping variable.

When high between-study heterogeneity persisted despite sensitivity analyses, we conducted Bayesian hierarchical meta-analyses to estimate the SMD with 95% credible intervals (95% CrI) using weakly informative priors. This analytic extension was not pre-specified in the PROSPERO-registered protocol and was introduced post hoc in response to peer-review suggestions, to further assess the robustness of the findings given the extent of between-study heterogeneity observed in some comparisons. Specifically, we applied a normal(0,1) prior for the intercept and a cauchy(0,0.5) prior for the s.d. of the random effects. Reference Gelman32 As a sensitivity analysis, we also tested an alternative model using an exponential(1) prior for the s.d. to evaluate the robustness of posterior estimates to prior specification. Reference Gelman, Simpson and Betancourt33 Overall, with this approach we aimed to assess the robustness of the heterogeneity estimates (τ) and the overall pooled effect under different plausible assumptions about the distribution of the true effect sizes across studies.

The analyses were performed using meta, metafor and brms packages in R (version 4.4.0, for MacOS). Reference Balduzzi, Rücker and Schwarzer34Reference Bürkner37 Statistical tests were two-sided and used a significance threshold of p-value <0.05.

Risk of bias assessment and the GRADE

Bias risk in the included studies was independently assessed by two reviewers (M.M. and F.Q.), using the Cochrane risk of bias tool. Reference Higgins, Altman, Gøtzsche, Jüni, Moher and Oxman38 All disagreements were discussed until consensus, and if necessary, another member of the team was consulted (G.M.G.). Each item on the risk of bias assessment was scored as high, low or unclear, and the GRADE tool was used to assess the overall certainty of evidence. Reference Schünemann, Brożek, Guyatt and Oxman39 Further information is available in the Supplementary Material.

Deviations to the PROSPERO protocol

The main deviation from the PROSPERO-registered protocol was the inclusion of Bayesian hierarchical meta-analyses, which were introduced post hoc in response to peer-review feedback as detailed in the statistical analyses section.

Results

Study characteristics

As shown in Fig. 1, from 387 records screened on title and abstract, 20 full texts were analysed. One additional RCT was found on clinicaltrials.gov but this was an incomplete trial due to inability to reach the target sample size due to COVID-19, 40 therefore this was not included in our final selection. The review process led to the selection of 14 studies Reference Jahanbakhsh, Manteghi, Emami, Mahyari, Gholampour and Mohammadpour17,Reference Andrade, Aswath, Chaturvedi, Srinivasa and Raguram41Reference Pandit, Srivastav, Sur, Chaudhuri, Wang and Biswas53 referring to 14 independent samples, corresponding to a total of 713 people (of which 360 treated with Withania somnifera and 353 controls), which were included in the quantitative synthesis (see also Supplementary Table 2 reporting the list of studies excluded after full-text review, along with the reason for exclusion).

Fig. 1 Preferred Reporting Items for Systematic Reviews and Meta-analyses flow diagram.

The trials were conducted in 4 countries: India (n = 8; 57.2%), Iran (n = 3; 21.4%), USA (n = 2; 14.3%) and Canada (n = 1; 7.1%). The overall percentage of females across the studies was 45.7%, mean age was 38.3 (s.d.: 10.9) years, with only one study (7.1%) involving underage participants. The most common diagnostic category was anxiety disorders (n = 5; 35.7%), followed by insomnia and chronic stress (each n = 2; 14.3%), major depressive disorder (MDD), bipolar disorder, schizophrenia and schizo-affective disorder, attention-deficit hyperactivity disorder (ADHD) and OCD, each n = 1 (7.1%). The clinical severity at the baseline was symptomatic or mild/moderate for most of the studies, except for one study performed on participants with moderate/high severity anxiety and one on participants with bipolar disorder in clinical remission. In 13 trials (92.9%) Withania somnifera was administered orally with daily dosing ranging from 10 to 12 000 mg (median 600 mg). One trial (7.1%) administered Withania somnifera with Shirodhara (oil dripping) therapy. Withanolides minimum concentration in the Withania somnifera extracts ranged from 1.5 to 8%, although this information was not reported by six studies. Withania somnifera was administered as add-on treatment in seven trials (50.0%), as the sole therapeutic agent in four trials (28.6%) and three trials (21.4%) did not report information on concurrent medication. For the studies administering Withania somnifera as add-on treatment, the concurrent treatment consisted of selective serotonin reuptake inhibitor (SSRI) antidepressants in two studies, mood stabilisers, antipsychotics, anti-ADHD medications without other specification and phytotherapeutic regimen with Nardostachys jatamansi and Lavandula stoechas, each n = 1. Most of the trials used placebo as the comparator (n = 12; 85.7%), one study (7.1%) used a combination of placebo and cognitive behavioural therapy (CBT) and one study (7.1%) considered the ongoing phytotherapeutic regimen alone as the control group. The median trial’s duration was 8 weeks, ranging from 6 to 12 weeks. The outcomes reported by the trials were: anxiety (n = 10), depression (n = 5), sleep (n = 4), stress (n = 3) and mania, psychotic symptoms and OCD symptoms (each n = 1). The main characteristics of the studies included in the review are summarised in Table 1 (see also Supplementary Table 3 for information on the composition of Withania somnifera extracts used in each study and Supplementary Table 4 for details on the outcome measures used in each study).

Table 1 Characteristics of the included studies

ADHD, attention-deficit hyperactivity disorder; AP, antipsychotics; C, control; CBT, cognitive–behavioural therapy; GAD, generalised anxiety disorder; MDD, major depressive disorder; mg, milligram; N, number; NA, not applicable; NOS, not otherwise specified; NR, information not reported; OCD, obsessive–compulsive disorder; PBO, placebo; po, per os (by mouth/orally); RCT, randomised controlled trial; SSRI, selective serotonin reuptake inhibitor; SZ, schizophrenia; T, treatment; WS, Withania somnifera.

Effect of Withania somnifera on anxiety symptoms

The meta-analysis of the effect of Withania somnifera on anxiety symptoms included ten studies and yielded significant evidence supporting the efficacy of the treatment (SMD: −1.62 (95% CI: −2.66; −0.57)), although with evidence of high between-study heterogeneity (I 2: 94%; p-value <0.001). Subgroup meta-analysis by diagnostic category revealed a significant between-group difference (p-value: 0.008); however, the pooled estimates indicated treatment effectiveness across most conditions, except for the single study conducted in individuals with bipolar disorder, which did not show a significant effect. The results are summarised in Fig. 2.

Fig. 2 Forest plot of anxiety among Withania somnifera and control groups: overall and by diagnostic subgroup. ADHD, attention-deficit and hyperactivity disorder; BD, bipolar disorder; IV, inverse variance; WS, Withania somnifera. The measures of anxiety across the studies were scaled as lower scores indicate lower anxiety symptoms. Reference to the included studies (in alphabetical order): Andrade et al, 2000 Reference Andrade, Aswath, Chaturvedi, Srinivasa and Raguram41 ; Chengappa et al, 2013 Reference Chengappa, Bowie, Schlicht, Fleet, Brar and Jindal42 ; Cooley et al, 2009 Reference Cooley, Szczurko, Perri, Mills, Bernhardt and Zhou45 ; Fuladi et al, 2021 Reference Fuladi, Emami, Mohammadpour, Karimani, Manteghi and Sahebkar46 ; Hosseini et al, 2019 Reference Hosseini, Moharari, Soltanifar, Mohammad Poor, Hosseini and Zahedian48 ; Khyati et al, 2013 Reference Khyati and Anup49 ; Langade et al, 2019 Reference Langade, Kanchi, Salve, Debnath and Ambegaokar50 ; Langade et al 2021 Reference Langade, Thakare, Kanchi and Kelgane51 ; Majeed et al, 2023 Reference Majeed, Nagabhushanam, Murali, Vishwanathan, Mamidala and Mundkur52 ; Pandit et al, 2024. Reference Pandit, Srivastav, Sur, Chaudhuri, Wang and Biswas53

Our investigation found evidence of publication bias. Notably, most studies reported outcomes favouring Withania somnifera, one showed a null effect and none reported negative results. This can be seen in the funnel plot displayed in Supplementary Figure 1 and was confirmed by Egger’s test p-value <0.001. However, the estimate of the effect size obtained by inputting missing studies into the funnel plot until symmetry is reached using Duval and Tweedie’s trim-and-fill procedure was highly similar (SMD: −1.62 (95% CI: −2.67; −0.57)).

To investigate the source of heterogeneity, leave-one-out analysis and meta-regression were performed.

Leave-one-out analysis, in which the meta-analysis was serially repeated after the exclusion of each study, found that by excluding the study from Khyati et al Reference Khyati and Anup49 the effect size decreased more than 25%, remaining statistically significant (SMD: −1.13 (95% CI: −1.65; −0.60); I 2: 85%). Interestingly, this study used the highest dose of Withania somnifera, which was more than ten times higher than the other trials. Instead, excluding other studies produced overall inconsequential changes (≤15%) in both the pooled estimate and heterogeneity. The results of leave-one-out analysis are available in Supplementary Table 5.

Meta-regression analyses were performed on the following variables, potentially associated with heterogeneity: (a) the mean age of participants; (b) the percentage of females in the total sample; (c) the country where the study was conducted; (d) Withania somnifera dose administered; (e) an interaction term of Withania somnifera dose × withanolides % in the extract used; (f) trial duration and (g) Withania somnifera use as monotherapy or add-on treatment. In the univariable meta-regression model, the variables resulting significantly associated with the estimate of the treatment effect was the Withania somnifera dose, with a larger anxiolytic effect observed in trials administering Withania somnifera at a higher dose (unstandardised regression coefficient (B: −0.004 (95% CI: −0.007; −0.001)) and the use as add-on treatment (B: −0.753 (95% CI: −1.47; −0.038)). It should be noted that this result might be strongly influenced by the outlier effect played by the study from Khyati et al Reference Khyati and Anup49 which used a dose of Withania somnifera more than ten times higher than the median dose. Univariable meta-regression results are displayed in Supplementary Table 6.

The effect of Withania somnifera on anxiety symptoms was further examined using a Bayesian hierarchical meta-analysis, which confirmed a statistically significant effect of the treatment (SMD: −1.23 (95% CrI: −2.19; −0.12)). The estimated between-study heterogeneity (τ) was 1.71 (95% CrI: 1.04; 2.82). This model applied weakly informative priors: a normal(0,1) prior for the intercept and a cauchy(0,0.5) prior for the standard deviation of the random effects and converged at 4000 iterations. Results are shown in Fig. 3. For sensitivity analysis, the model was re-estimated using an alternative prior for τ (i.e. exponential(1)). Results were consistent across priors, with stable posterior distributions for both τ and the overall treatment effect (see Supplementary Figure 2). Model comparison using leave-one-out cross-validation indicated similar predictive performance (difference in Expected Log Pointwise Predictive Density (∆ELPD): −0.1). Given the conservative nature of the cauchy(0,0.5) prior, we retained it in the final model.

Fig. 3 Bayesian forest plot of study-level and pooled posterior estimates of Withania somnifera efficacy on anxiety. The measures of anxiety across the studies were scaled as lower scores indicate lower anxiety symptoms. Reference to the included studies (in alphabetical order): Andrade et al, 2000 Reference Andrade, Aswath, Chaturvedi, Srinivasa and Raguram41 ; Chengappa et al, 2013 Reference Chengappa, Bowie, Schlicht, Fleet, Brar and Jindal42 ; Cooley et al, 2009 Reference Cooley, Szczurko, Perri, Mills, Bernhardt and Zhou45 ; Fuladi et al, 2021 Reference Fuladi, Emami, Mohammadpour, Karimani, Manteghi and Sahebkar46 ; Hosseini et al, 2019 Reference Hosseini, Moharari, Soltanifar, Mohammad Poor, Hosseini and Zahedian48 ; Khyati et al, 2013 Reference Khyati and Anup49 ; Langade et al, 2019 Reference Langade, Kanchi, Salve, Debnath and Ambegaokar50 ; Langade et al 2021 Reference Langade, Thakare, Kanchi and Kelgane51 ; Majeed et al, 2023 Reference Majeed, Nagabhushanam, Murali, Vishwanathan, Mamidala and Mundkur52 ; Pandit et al, 2024. Reference Pandit, Srivastav, Sur, Chaudhuri, Wang and Biswas53

Finally, we included a Withania somnifera dose as a moderator in the Bayesian model. To enhance interpretability, we log-transformed the dose variable and centred the intercept at the median dose of 600 mg/day. At this reference dose, the model estimated a significant treatment effect on anxiety symptoms (SMD: −1.27 (95% CrI: −2.17; −0.21)). However, we found no evidence of a statistically significant increase in treatment efficacy for doses above the median (B: −0.50 (95% CrI: −1.19; 0.16)).

Effect of Withania somnifera on depression symptoms

The meta-analysis of the effect of Withania somnifera on depression symptoms included five studies, and specifically for one of these the outcome data were extracted from a secondary publication on the same sample. Reference Gannon, Brar, Rai and Chengappa54 The pooled estimate of the treatment effect favoured Withania somnifera (SMD: −1.28 (95% CI: −2.40; −0.16])), with high between-study heterogeneity (I 2: 93%; p-value <0.001). While generally recommended with a minimum of ten studies, we however conducted an exploratory subgroup analysis based on participants’ diagnoses to investigate potential sources of heterogeneity. The test for subgroup differences was statistically significant (p-value <0.001), with the largest effect of Withania somnifera on depression symptoms observed in studies involving participants with anxiety, followed by those with MDD. No significant effect was found in the single studies involving participants with bipolar disorder and schizophrenia. These results should be interpreted cautiously due to the limited number of studies within each subgroup. The results are summarised in Fig. 4.

Fig. 4 Forest plot of depression among Withania somnifera and control groups: overall and by diagnostic subgroup. BD, bipolar disorder; IV, inverse variance; MDD, major depressive disorder; SZ, schizophrenia; WS, Withania somnifera. The measures of depression across the studies were scaled as lower scores indicate lower depression symptoms. Reference to the included studies (in alphabetical order): Chengappa et al, 2013 Reference Chengappa, Bowie, Schlicht, Fleet, Brar and Jindal42 ; Chengappa et al, 2018 Reference Chengappa, Brar, Gannon and Schlicht43 ; Fulzele et al, 2014 Reference Fulzele, Ingle, Huda and Mishra47 ; Majeed et al, 2023 Reference Majeed, Nagabhushanam, Murali, Vishwanathan, Mamidala and Mundkur52 ; Pandit et al, 2024. Reference Pandit, Srivastav, Sur, Chaudhuri, Wang and Biswas53

Due to the limited number of studies, we did not conduct a test for publication bias and meta-regression.

Leave-one-out analysis showed that by excluding the study from Chengappa et al, Reference Chengappa, Bowie, Schlicht, Fleet, Brar and Jindal42 the pooled estimate favouring Withania somnifera reached statistical significance (SMD: −1.28 (95% CI: −2.45; -0.10)), with high heterogeneity (I 2: 91%). In addition, by excluding the study from Majeed et al, Reference Majeed, Nagabhushanam, Murali, Vishwanathan, Mamidala and Mundkur52 there was a marked decrease in the heterogeneity (I 2: 71%) and in the effect size (SMD: −0.39 (95% CI: −1.01; 0.24)). The results of the leave-one-out analysis are available in Supplementary Table 7.

To further examine the robustness of the findings in a context with high heterogeneity, we performed Bayesian hierarchical meta-analysis using weakly informative priors (i.e. normal(0,1) for the intercept, cauchy(0,0.5) and exponential(1) for τ). Both models converged at 4000 iterations and produced similar findings in terms of posterior distribution of τ and the overall treatment effect (see Supplementary Figure 3; ∆ELPD: −0.3). Given its conservative features, the model using a cauchy(0,0.5) prior was retained as the primary analysis. This model yielded a non-statistically significant treatment effect (SMD: −0.95 (95% CrI: −1.96; 0.24)) and high between-study heterogeneity (τ: 1.29 (95% CrI: 0.62; 2.66)). The results of the Bayesian meta-analysis are displayed in Fig. 5.

Fig. 5 Bayesian forest plot of study-level and pooled posterior estimates of Withania somnifera efficacy on depression. The measures of depression across the studies were scaled as lower scores indicate lower depression symptoms. Reference to the included studies (in alphabetical order): Chengappa et al, 2013 Reference Chengappa, Bowie, Schlicht, Fleet, Brar and Jindal42 ; Chengappa et al, 2018 Reference Chengappa, Brar, Gannon and Schlicht43 ; Fulzele et al, 2014 Reference Fulzele, Ingle, Huda and Mishra47 ; Majeed et al, 2023 Reference Majeed, Nagabhushanam, Murali, Vishwanathan, Mamidala and Mundkur52 ; Pandit et al, 2024. Reference Pandit, Srivastav, Sur, Chaudhuri, Wang and Biswas53

Effect of Withania somnifera on sleep quality

The meta-analysis of the effect of Withania somnifera on sleep included four studies. The pooled estimate favoured Withania somnifera (SMD: −1.35 (95% CI: −1.79; −0.91)) with moderate between-study heterogeneity (I 2: 53%, p-value: 0.097). The results are presented in Fig. 6.

Fig. 6 Forest plot of sleep quality among Withania somnifera and control groups. IV: inverse variance; WS, Withania somnifera. The measures of sleep quality across the studies were scaled as lower scores indicate better sleep quality. Reference to the included studies (in alphabetical order): Langade et al, 2019 Reference Langade, Kanchi, Salve, Debnath and Ambegaokar50 ; Langade et al 2021 Reference Langade, Thakare, Kanchi and Kelgane51 ; Majeed et al, 2023 Reference Majeed, Nagabhushanam, Murali, Vishwanathan, Mamidala and Mundkur52 ; Pandit et al, 2024. Reference Pandit, Srivastav, Sur, Chaudhuri, Wang and Biswas53

Due to the limited number of studies included in the meta-analysis, sensitivity analyses could not be performed.

Effect of Withania somnifera on other mental health symptoms

Four other outcome measures were reported in the included studies. These consisted of schizophrenia symptoms, mania symptoms, OCD symptoms and perceived stress. Except for perceived stress, each outcome was reported by only one study, precluding meta-analysis. The meta-analysis for perceived stress was based on three studies, indicating a significant effect in favour of Withania somnifera (SMD: −0.95 (95% CI: −1.46; −0.43)), with moderate between-study heterogeneity (I 2: 58%, p-value: 0.090). The results are shown in Fig. 7.

Fig. 7 Forest plot of perceived stress among Withania somnifera and control groups. IV: inverse variance; WS: Withania somnifera. The measures of stress across the studies were scaled as lower scores indicate lower perceived stress. Reference to the included studies (in alphabetical order): Chengappa et al, 2018 Reference Chengappa, Brar, Gannon and Schlicht43 ; Choudhary et al, 2017 Reference Choudhary, Bhattacharyya and Joshi44 ; Pandit et al, 2024. Reference Pandit, Srivastav, Sur, Chaudhuri, Wang and Biswas53

Analysis of safety and tolerability

The analysis of the safety and tolerability of Withania somnifera treatment was made by assessing the rates of adverse events, drop-out due to any cause and drop out due to death or serious adverse events across the experimental and control groups. There were four studies Reference Fuladi, Emami, Mohammadpour, Karimani, Manteghi and Sahebkar46Reference Khyati and Anup49 not reporting information on the number of treatment’s adverse effects across the trial’s arms, although these stated in the respective papers that there were no significant differences between the experimental and control groups. Additionally, two studies Reference Fulzele, Ingle, Huda and Mishra47,Reference Khyati and Anup49 did not report information on the number of drop-outs during the trial. Notably, no death or drop-out due to severe treatment adverse effects occurred both in the experimental and in the control groups of all the included trials. The meta-analysis of the drop-out due to any cause included ten studies, and the meta-analysis of the rate of any adverse event was based on five studies. Our investigation found similar rates of drop-out due to any cause and of any adverse effect across the experimental and control groups, supporting no significant differences between Withania somnifera and the placebo in terms of safety and tolerability (odds ratio 0.81 (95% CI: 0.42; 1.58) and odds ratio 0.99 (95% CI: 0.54; 1.82) for the rate of drop-out due to any cause and any adverse effect, respectively), with no evidence of between-study heterogeneity. The most reported adverse events were gastro-intestinal disturbances (e.g. diarrhoea or dyspepsia), which were transitory and not severe. The results are displayed in Fig. 8.

Fig. 8 Forest plots of drop-out due to any cause (Panel A) and any adverse effect (Panel B) among Withania somnifera and control groups. MH: Mantel-Haenszel; WS: Withania somnifera. Lower rates indicate better safety/tolerability. Reference to the included studies (in alphabetical order): Andrade et al, 2000 Reference Andrade, Aswath, Chaturvedi, Srinivasa and Raguram41 ; Chengappa et al, 2013 Reference Chengappa, Bowie, Schlicht, Fleet, Brar and Jindal42 ; Chengappa et al, 2018 Reference Chengappa, Brar, Gannon and Schlicht43 ; Choudhary et al, 2017 Reference Choudhary, Bhattacharyya and Joshi44 ; Cooley et al, 2009 Reference Cooley, Szczurko, Perri, Mills, Bernhardt and Zhou45 ; Fuladi et al, 2021 Reference Fuladi, Emami, Mohammadpour, Karimani, Manteghi and Sahebkar46 ; Hosseini et al, 2019 Reference Hosseini, Moharari, Soltanifar, Mohammad Poor, Hosseini and Zahedian48 ; Jahanbakhsh et al, 2016 Reference Jahanbakhsh, Manteghi, Emami, Mahyari, Gholampour and Mohammadpour17 ; Langade et al, 2019 Reference Langade, Kanchi, Salve, Debnath and Ambegaokar50 ; Langade et al 2021 Reference Langade, Thakare, Kanchi and Kelgane51 ; Majeed et al, 2023. Reference Majeed, Nagabhushanam, Murali, Vishwanathan, Mamidala and Mundkur52

GRADE of the evidence

A detailed summary on the risk of bias in all 14 trials is reported in Supplementary Figs 4 and 5, along with an assessment of the quality of the evidence (see Supplementary Table 8). In the GRADE system, the evidence from RCTs is initially set to high; there are then criteria that can be used either to downgrade or upgrade (see further information in the Supplementary Material). The quality of the evidence ranged from very low for depression symptoms to low for anxiety symptoms, sleep quality and stress. The main reasons for downgrading the certainty of evidence were the limited number of studies contributing to each pooled estimate, the high level of heterogeneity between the studies and bias threats related to the blinding procedures.

Discussion

This systematic review set out to investigate the efficacy and safety of the treatment with Withania somnifera on people with mental disorders. Specifically, we synthesised evidence on the effect of Withania somnifera on anxiety and depression symptoms, while also exploring its effects on sleep quality, schizophrenia, mania, OCD and perceived stress. Our investigation points out a statistically significant effect of Withania somnifera in improving anxiety, sleep quality and stress. The effect on anxiety symptoms should be interpreted considering high between-study heterogeneity, and was limited on the other mental health outcomes by the small number of studies contributing to each pooled estimate. In spite of this, the observed anxiolytic effect aligns with previous findings on both clinical and healthy populations. Reference Singh, Bhalla, de Jager and Gilca4,Reference Akhgarjand, Asoudeh, Bagheri, Kalantar, Vahabi and Shab-Bidar12,Reference Pratte, Nanavati, Young and Morley55 However, our study is the largest meta-analysis on Withania somnifera in clinical settings. Interestingly, the observed anxiolytic effect of Withania somnifera extends beyond anxiety disorder populations, having also been observed in individuals with ADHD, depression and insomnia. Reference Hosseini, Moharari, Soltanifar, Mohammad Poor, Hosseini and Zahedian48,Reference Langade, Kanchi, Salve, Debnath and Ambegaokar50Reference Majeed, Nagabhushanam, Murali, Vishwanathan, Mamidala and Mundkur52 Although only one trial in ADHD and one trial in depression investigated the effect of Withania somnifera on anxiety symptoms, its broad pharmacological profile, modulating GABAergic, cholinergic and anti-inflammatory pathways, Reference Kulkarni and Dhir1,Reference Sarris, McIntyre and Camfield9,Reference Mácsai, Datki, Csupor, Horváth and Zomborszki10 could underlie its trans-diagnostic anxiolytic effects. However, it is interesting to note that the study from Chengappa et al, Reference Chengappa, Bowie, Schlicht, Fleet, Brar and Jindal42 which was conducted on patients with bipolar disorder in clinical remission, provided estimates crossing the line of no effect for both anxiety and depression symptoms. Similarly, the other study from Chengappa et al, Reference Chengappa, Brar, Gannon and Schlicht43 performed on people with acute exacerbation of schizophrenia, did not find evidence supporting the efficacy of the treatment on depressive symptoms of participants. Finally, the study from Fulzele et al Reference Fulzele, Ingle, Huda and Mishra47 that was performed on participants with mild to moderate depression also reported a small, yet significant, effect supporting the anxiolytic and antidepressant effect of Withania somnifera. It could be argued that in both clinically severe or clinically stable patients, anxiety and depressive symptoms are very unlikely to improve any further with Withania somnifera.

We found a dose–response relationship for the anxiolytic effect, although caution is warranted as doses ranged widely across studies (from 10 to 12 000 mg/day) and this evidence was markedly influenced by one study which administered an exceptionally high dose of Withania somnifera (12 000 mg daily). Reference Khyati and Anup49 However, our Bayesian meta-analysis suggested no statistically significant advantage of doses above 600 mg/day, indicating that this median dose may represent an optimal balance of efficacy and tolerability. It is important to acknowledge that, beyond the dose, Withania somnifera extracts exhibit differences in the bioactives composition influencing specific neural circuits, neurotransmitters, inflammatory responses, redox mechanisms and immune systems, all of which are purportedly relevant to mental disorders. Despite challenges in obtaining detailed information on the composition of Withania somnifera extracts used in the included trials, our investigation found no significant interaction of dose and minimum withanolides concentration on the anxiolytic effect. Previous studies on the pharmacokinetic features of Withania somnifera reported a short half-life, requiring two or three daily oral administrations to reach adequate bioavailability. Reference Mandlik Ingawale and Namdeo6,Reference Vaidya, Naik, Ganu, Parmar, Jagtap and Saste56 Speculatively, these characteristics may contribute to dose-related effects and could limit the sustained efficacy of Withania somnifera if dosing is suboptimal or inconsistent. However, the multitude of bioactives in Withania somnifera extracts and the inconsistent reporting of these across the trials, limit considerations of Withania somnifera pharmacokinetics and pharmacodynamics. Additionally, most of the studies on the pharmacological and safety profile of Withania somnifera were performed on doses around 500 mg/day, therefore the use of higher doses of Withania somnifera should be further investigated, especially in terms of safety when administered with other treatments.

The frequentist meta-analysis for depressive symptoms yielded a statistically significant pooled effect favouring Withania somnifera, although with high between-study heterogeneity. Importantly, the effect was not confirmed in the Bayesian hierarchical approach, emphasising uncertainty under different assumptions. This discrepancy indicates that the evidence for the antidepressant effect of Withania somnifera is less robust and more likely influenced by small-studies effects, between-study heterogeneity or confounding factors and therefore should be interpreted with caution. Notably, by looking at the contribution of the individual studies, leave-one-out analysis showed a potentially enhanced antidepressant effect in individuals with concurrent anxiety and stress, providing hints for future trials. Additionally, the study from Majeed et al Reference Majeed, Nagabhushanam, Murali, Vishwanathan, Mamidala and Mundkur52 which administered a Withania somnifera extract supplemented with piperine – a compound with antidepressant-like properties Reference Huang, Chen, Wang, Lin, Wu and Yan57,Reference Wattanathorn, Chonpathompikunlert, Muchimapura, Priprem and Tankamnerdthai58 – showed a particularly large antidepressant effect.

Although the meta-analyses of sleep quality and stress were based on a small number of studies limiting the certainty of the evidence, the pooled effects were statistically significant in favour of Withania somnifera, warranting further investigation of Withania somnifera in stress and insomnia treatment in large and high-quality RCTs. The beneficial effect of Withania somnifera on sleep quality and stress confirmed previous research findings both among clinical samples and healthy volunteers. Reference Akhgarjand, Asoudeh, Bagheri, Kalantar, Vahabi and Shab-Bidar12,Reference Cheah, Norhayati, Husniati Yaacob and Abdul Rahman13,Reference Baker, Kirby, O’Connor, Lindsay, Hutchins and Harris59 These effects, substantiated by the found anxiolytic properties, reinforce Withania somnifera adaptogen potential.

The meta-analysis was precluded for mania, OCD and schizophrenia symptoms as each was assessed in only one trial. However, these individual studies reported a reduction in schizophrenia symptoms for Withania somnifera used as an add-on to antipsychotics, whereas no significant effects were observed for mania or OCD symptoms. Similar results were obtained for other add-on treatments for schizophrenia, such as sarcosine and antioxidants, Reference Marchi, Galli, Magarini, Mattei and Galeazzi60,Reference Magalhães, Dean, Andreazza, Berk and Kapczinski61 which share some similarities with Withania somnifera, warranting further investigations on Withania somnifera as a potential treatment option with a good tolerability.

Finally, in terms of safety and tolerability, Withania somnifera treatment appears to be safe and well-accepted by patients, as supported by the similar rate of drop-out due to any cause and of adverse effects across the treatment and control arms. The reported adverse events were generally mild, transitory and did not require treatment interruption.

Limitations

The results of this study should be interpreted considering its limitations.

First, the relatively small number of studies included in the final selection limited the possibility to perform robust meta-analytic investigations. This limitation resulted in imprecision, evident in large confidence intervals or confidence intervals crossing the line of no-effect and susceptibility to effect of outlier studies. In addition, three out of the four meta-analyses performed the number of studies included was less than ten, falling short of the threshold required for robust testing of publication bias. Reference Sterne, Sutton, Ioannidis, Terrin, Jones and Lau27 Further, for mania, OCD and schizophrenia there was only one trial available, significantly limiting any possibility to make suggestions on the use of Withania somnifera in these specific disorders. Second, Withania somnifera is a treatment from the Ayurvedic medicine tradition. However, the identification of studies was conducted across databases in the biomedical sciences, which may have limited coverage of traditional and alternative medicine reports. This limitation may challenge the comprehensiveness of the search strategy. Third, there was substantial between-study heterogeneity, compounded by studies providing exceptionally outlier estimates, posing challenges for a nuanced investigation of the source of heterogeneity. Fourth, the risk of bias assessment found threats related to allocation and blinding procedures, reflected in the low to very low grading of the evidence. Therefore, these findings should be interpreted with caution and confirmed in future high-quality trials. Fifth, some studies did not report information on some participants’ characteristics, Withania somnifera extract composition, concurrent treatments or the adverse events of treatment. Stricter adherence to the consolidated standards of reporting trials is needed. Finally, the selected studies were all performed in India, Iran or North America. This geographical limitation potentially influences the generalisability of the findings.

Implications for research and practice

The results of this research highlight the therapeutic potential of Withania somnifera, whose main benefits were observed in the treatment of anxiety, which is one of the most common mental disorders. Reference Alonso, Liu, Evans-Lacko, Sadikova, Sampson and Chatterji62 Despite a large effect size observed on anxiety symptoms, the certainty of the evidence was rated low mainly due to high between-study heterogeneity.

Sleep disorders and depressive symptoms are also rather common in the general population even as transient symptoms and display a close correlation with anxiety disorders. Reference Arias de la Torre, Vilagut, Ronaldson, Bakolis, Dregan and Martín63,Reference Morin and Jarrin64 This correlation extends beyond epidemiological observations, delving into shared neurobiological underpinnings which could be effectively targeted by the Withania somnifera treatment. Reference Chellappa and Aeschbach65,Reference Johnson, Roth and Breslau66 The improvement in anxiety and stress-related symptoms could account for a better ability to regulate emotions in response to adverse stimuli, consistent with the suggested adaptogen effect of Withania somnifera. Reference Etkin and Wager67 In future, it could be rational to investigate prospective studies of whether Withania somnifera can be used in individuals with high levels of stress to prevent them from transitioning to anxiety disorders.

The broad pharmacological profile could be useful in other disorders, like depression, or conditions with a strong link to environmental risk factors, such as stress and burnout. Furthermore, the reported enhancements in cognition and the anti-inflammatory properties of Withania somnifera add layers to its potential applicability in other psychiatric conditions. Reference Mikulska, Malinowska, Ignacyk, Szustowski, Nowak and Pesta14,Reference Ng, Loke, Foo, Tan, Chan and Lim20,Reference Chen, Liu, Cheng, Li, Huang and Ding68 Interestingly, Withania somnifera also showed elsewhere the positive impact on diabetes management, anti-ischaemic properties and effects on sexual function, comorbidities often associated with psychiatric disorders. Reference Mikulska, Malinowska, Ignacyk, Szustowski, Nowak and Pesta14,Reference Lopresti and Smith69,Reference Zalpour, Zare and Nasiri70 Future trials could investigate the benefit of Withania somnifera supplementation for individuals with mental disorders, especially those vulnerable to developing these comorbidities.

While the results of this study suggest a favourable safety profile of Withania somnifera, in agreement with previous research, Reference Brunetti, Lo Faro, Tini, Busardò and Carlier7,Reference Vaidya, Naik, Ganu, Parmar, Jagtap and Saste56,Reference Tandon and Yadav71 the need for cautious observation persists. Standardisation ofWithania somnifera titles in commercial preparation should be pursued to ensure that users receive the expected benefits and to support accurate assessments of its pharmacological effects in research. In addition, large-scale investigations, with a specific focus on concomitant use with other medications and potential interactions, are needed. The safety and tolerability aspects, derived from a synthesis of clinical and preclinical evidence, should guide future randomised trials. These studies should adhere to robust therapeutic and safety standards, conducted on a larger scale, to improve understanding of Withania somnifera’s potential in psychiatric care.

In conclusion, our study provided evidence for the clinical effectiveness of at least 600 mg/day of Withania somnifera in reducing anxiety symptoms. Despite the large anxiolytic effect size found, the high between-study heterogeneity warrants cautious interpretation. Additionally, a positive effect of Withania somnifera was found also on sleep quality and perceived stress, although caution is advised in these findings due to the limited data available. Future research should prioritise exploring Withania somnifera’s role in anxiety, depression and insomnia treatment, considering its promising trends. Overall, although Withania somnifera showed potential in psychiatric care, more, thorough, studies are crucial to define its therapeutic boundaries and optimise its clinical utility.

Supplementary material

The supplementary material is available online at https://doi.org/10.1192/bjo.2025.10885

Data availability

The codes for reproducing the dataset and the analyses can be accessed here: https://github.com/MattiaMarchi/Withania-somnifera_Ashwagandha_-mental-illness.

Author contributions

Conceptualisation and planning: M.M., P.G. and G.M.G. Acquisition: M.M., P.G. and F.Q. Analysis of the data: M.M. Interpretation of the data: M.M., P.G. and G.M.G. Drafting: M.M., P.G., A.T., D.U., E.D.M. and G.L. Critical revision of the manuscript: M.M., L.P., S.F. and G.M.G. All authors approved the final submitted version of the manuscript.

Funding

This research received no specific grant from any funding agency, commercial or not-for-profit sectors.

Declaration of interest

None.

Ethical standards

This systematic review is exempt from ethics approval because we are collecting and synthesising data from previous studies in which ethical approval has already been obtained by the trial investigators in their respective local sites.

References

Kulkarni, SK, Dhir, A. Withania somnifera: an Indian ginseng. Prog Neuropsychopharmacol Biol Psychiatry 2008; 32: 1093–105.10.1016/j.pnpbp.2007.09.011CrossRefGoogle ScholarPubMed
Deocaris, CC, Widodo, N, Wadhwa, R, Kaul, SC. Merger of ayurveda and tissue culture-based functional genomics: inspirations from systems biology. J Transl Med 2008; 6: 14.10.1186/1479-5876-6-14CrossRefGoogle ScholarPubMed
Modak, M, Dixit, P, Londhe, J, Ghaskadbi, S, Devasagayam, T. Indian herbs and herbal drugs used for the treatment of diabetes. J Clin Biochem Nutr 2007; 40: 163–73.10.3164/jcbn.40.163CrossRefGoogle ScholarPubMed
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: 208–13.10.4314/ajtcam.v8i5S.9CrossRefGoogle ScholarPubMed
Gaurav, H, Yadav, D, Maurya, A, Yadav, H, Yadav, R, Shukla, AC, et al. Biodiversity, biochemical profiling, and pharmaco-commercial applications of Withania somnifera: a review. Molecules 2023; 28: 1208.10.3390/molecules28031208CrossRefGoogle ScholarPubMed
Mandlik Ingawale, DS, Namdeo, AG. Pharmacological evaluation of Ashwagandha highlighting its healthcare claims, safety, and toxicity aspects. J Diet Suppl 2021; 18: 183226.10.1080/19390211.2020.1741484CrossRefGoogle ScholarPubMed
Brunetti, P, Lo Faro, AF, Tini, A, Busardò, FP, Carlier, J. Pharmacology of herbal sexual enhancers: a review of psychiatric and neurological adverse effects. Pharmaceuticals (Basel) 2020; 13: 309.10.3390/ph13100309CrossRefGoogle ScholarPubMed
Dar, NJ, Hamid, A, Ahmad, M. Pharmacologic overview of Withania somnifera, the Indian ginseng. Cell Mol Life Sci 2015; 72: 4445–60.10.1007/s00018-015-2012-1CrossRefGoogle ScholarPubMed
Sarris, J, McIntyre, E, Camfield, DA. Plant-based medicines for anxiety disorders, part 2: a review of clinical studies with supporting preclinical evidence. CNS Drugs 2013; 27: 301–19.10.1007/s40263-013-0059-9CrossRefGoogle ScholarPubMed
Mácsai, L, Datki, ZL, Csupor, D, Horváth, A, Zomborszki, ZP. Biological activities of four adaptogenic plant extracts and their active substances on a rotifer model. Evid Based Complement Alternat Med 2018; 2018: 3690683.10.1155/2018/3690683CrossRefGoogle ScholarPubMed
Zhang, W, Yan, Y, Wu, Y, Yang, H, Zhu, P, Yan, F, et al. Medicinal herbs for the treatment of anxiety: a systematic review and network meta-analysis. Pharmacol Res 2022; 179: 106204.10.1016/j.phrs.2022.106204CrossRefGoogle ScholarPubMed
Akhgarjand, C, Asoudeh, F, Bagheri, A, Kalantar, Z, Vahabi, Z, Shab-Bidar, S, et al. Does Ashwagandha supplementation have a beneficial effect on the management of anxiety and stress? A systematic review and meta-analysis of randomized controlled trials. Phytother Res 2022; 36: 4115–24.10.1002/ptr.7598CrossRefGoogle Scholar
Cheah, KL, Norhayati, MN, Husniati Yaacob, L, Abdul Rahman, R. Effect of Ashwagandha (Withania somnifera) extract on sleep: a systematic review and meta-analysis. PLOS One 2021; 16: e0257843.10.1371/journal.pone.0257843CrossRefGoogle ScholarPubMed
Mikulska, P, Malinowska, M, Ignacyk, M, Szustowski, P, Nowak, J, Pesta, K, et al. Ashwagandha (Withania somnifera) – current research on the health-promoting activities: a narrative review. Pharmaceutics 2023; 15: 1057.10.3390/pharmaceutics15041057CrossRefGoogle ScholarPubMed
Sarris, J, Ravindran, A, Yatham, LN, Marx, W, Rucklidge, JJ, McIntyre, RS, et al. Clinician guidelines for the treatment of psychiatric disorders with nutraceuticals and phytoceuticals: the World Federation of Societies of Biological Psychiatry (WFSBP) and Canadian Network for Mood and Anxiety Treatments (CANMAT) Taskforce. World J Biol Psychiatry 2022; 23: 424–55.10.1080/15622975.2021.2013041CrossRefGoogle Scholar
Ayati, Z, Sarris, J, Chang, D, Emami, SA, Rahimi, R. Herbal medicines and phytochemicals for obsessive–compulsive disorder. Phytother Res 2020; 34: 1889–901.10.1002/ptr.6656CrossRefGoogle ScholarPubMed
Jahanbakhsh, SP, Manteghi, AA, Emami, SA, Mahyari, S, Gholampour, B, Mohammadpour, AH, et al. Evaluation of the efficacy of Withania somnifera (Ashwagandha) root extract in patients with obsessive–compulsive disorder: a randomized double-blind placebo-controlled trial. Complement Ther Med 2016; 27: 25–9.10.1016/j.ctim.2016.03.018CrossRefGoogle ScholarPubMed
Kumar, S, Phaneuf, D, Julien, J-P. Withaferin-A treatment alleviates TAR DNA-binding protein-43 pathology and improves cognitive function in a mouse model of FTLD. Neurotherapeutics 2021; 18: 286–96.10.1007/s13311-020-00952-0CrossRefGoogle Scholar
Lewis, JE, Poles, J, Shaw, DP, Karhu, E, Khan, SA, Lyons, AE, et al. The effects of twenty-one nutrients and phytonutrients on cognitive function: a narrative review. J Clin Transl Res 2021; 7: 575–620.Google ScholarPubMed
Ng, QX, Loke, W, Foo, NX, Tan, WJ, Chan, HW, Lim, DY, et al. A systematic review of the clinical use of Withania somnifera (Ashwagandha) to ameliorate cognitive dysfunction. Phytother Res 2020; 34: 583–90.10.1002/ptr.6552CrossRefGoogle ScholarPubMed
Page, MJ, McKenzie, JE, Bossuyt, PM, Boutron, I, Hoffmann, TC, Mulrow, CD, et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. BMJ 2021; 372: n71.10.1136/bmj.n71CrossRefGoogle ScholarPubMed
American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders 5th ed., Text Rev. APA, 2022.Google Scholar
World Health Organization. ICD-11: International Classification of Diseases 11th Rev. WHO, 2022.Google Scholar
DerSimonian, R, Laird, N. Meta-analysis in clinical trials. Control Clin Trials 1986; 7: 177–88.10.1016/0197-2456(86)90046-2CrossRefGoogle ScholarPubMed
Higgins, JPT, Li, T, Deeks, JJ. Choosing effect measures and computing estimates of effect. In Cochrane Handbook for Systematic Reviews of Interventions, version 6.1 (eds Higgins, JPT, Thomas, J, Chandler, J, Cumpston, M, Li, T, Page, MJ, et al): Ch. 6. Cochrane, 2020.Google Scholar
Higgins, JPT, Thompson, SG. Quantifying heterogeneity in a meta-analysis. Stat Med 2002; 21: 1539–58.10.1002/sim.1186CrossRefGoogle ScholarPubMed
Sterne, JAC, Sutton, AJ, Ioannidis, JPA, Terrin, N, Jones, DR, Lau, J, et al. Recommendations for examining and interpreting funnel plot asymmetry in meta-analyses of randomised controlled trials. BMJ 2011; 343: d4002.10.1136/bmj.d4002CrossRefGoogle ScholarPubMed
Duval, S, Tweedie, R. Trim and fill: a simple funnel-plot–based method of testing and adjusting for publication bias in meta-analysis. Biometrics 2000; 56: 455–63.10.1111/j.0006-341X.2000.00455.xCrossRefGoogle ScholarPubMed
Sutton, AJ, Duval, SJ, Tweedie, RL, Abrams, KR, Jones, DR. Empirical assessment of effect of publication bias on meta-analyses. BMJ 2000; 320: 1574–7.10.1136/bmj.320.7249.1574CrossRefGoogle ScholarPubMed
Terrin, N, Schmid, CH, Lau, J, Olkin, I. Adjusting for publication bias in the presence of heterogeneity. Stat Med 2003; 22: 2113–26.10.1002/sim.1461CrossRefGoogle ScholarPubMed
Sterne, JA, Egger, M, Moher, D. Addressing reporting biases. In Cochrane Handbook for Systematic Reviews of Interventions, version 5.0.1 (eds Higgins, JPT, Green, S): 297333. John Wiley & Sons, Ltd, 2008.10.1002/9780470712184.ch10CrossRefGoogle Scholar
Gelman, A. Prior distributions for variance parameters in hierarchical models. Bayesian Anal 2006; 1: 515–34.10.1214/06-BA117ACrossRefGoogle Scholar
Gelman, A, Simpson, D, Betancourt, M. The prior can often only be understood in the context of the likelihood. Entropy 2017; 19: 555.10.3390/e19100555CrossRefGoogle Scholar
Balduzzi, S, Rücker, G, Schwarzer, G. How to perform a meta-analysis with R: a practical tutorial. Evid Based Ment Health 2019; 22: 153–60.10.1136/ebmental-2019-300117CrossRefGoogle Scholar
Schwarzer, G. meta: General Package for Meta-Analysis. R Foundation, 2021 (https://CRAN.R-project.org/package=meta).Google Scholar
R Foundation for Statistical Computing. R: A Language and Environment for Statistical Computing. R Foundation, 2024 (https://www.R-project.org).Google Scholar
Bürkner, P-C. Bayesian item response modeling in R with brms and Stan. J Stat Softw 2021; 100: 154.10.18637/jss.v100.i05CrossRefGoogle Scholar
Higgins, JPT, Altman, DG, Gøtzsche, PC, Jüni, P, Moher, D, Oxman, AD, et al. The Cochrane Collaboration’s tool for assessing risk of bias in randomised trials. BMJ 2011; 343: d5928.10.1136/bmj.d5928CrossRefGoogle ScholarPubMed
Schünemann, H, Brożek, J, Guyatt, G, Oxman, A. GRADE Handbook for Grading Quality of Evidence and Strength of Recommendations. GRADE Working Group, 2013 (https://gdt.gradepro.org/app/handbook/handbook.html).Google Scholar
Cochane Central Register of Controlled Trials. NCT03437668. Adjunctive Withania somnifera (Ashwagandha) for Persistent Symptoms in People with Schizophrenia. Conchrane Library, 2018 (https://www.cochranelibrary.com/central/doi/10.1002/central/CN-01523105/full).Google Scholar
Andrade, C, Aswath, A, Chaturvedi, SK, Srinivasa, M, Raguram, R. A double-blind, placebo-controlled evaluation of the anxiolytic efficacy of an ethanolic extract of Withania somnifera. Indian J Psychiatry 2000; 42: 295301.Google ScholarPubMed
Chengappa, KNR, Bowie, CR, Schlicht, PJ, Fleet, D, Brar, JS, Jindal, R. Randomized placebo-controlled adjunctive study of an extract of Withania somnifera for cognitive dysfunction in bipolar disorder. J Clin Psychiatry 2013; 74: 1076–83.10.4088/JCP.13m08413CrossRefGoogle ScholarPubMed
Chengappa, KNR, Brar, JS, Gannon, JM, Schlicht, PJ. 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: 17m11826.10.4088/JCP.17m11826CrossRefGoogle ScholarPubMed
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: 96106.10.1177/2156587216641830CrossRefGoogle ScholarPubMed
Cooley, K, Szczurko, O, Perri, D, Mills, EJ, Bernhardt, B, Zhou, Q, et al. Naturopathic care for anxiety: a randomized controlled trial ISRCTN78958974. PLOS One 2009; 4: e6628.10.1371/journal.pone.0006628CrossRefGoogle ScholarPubMed
Fuladi, S, Emami, SA, Mohammadpour, AH, Karimani, A, Manteghi, AA, Sahebkar, A. Assessment of the efficacy of Withania somnifera root extract in patients with generalized anxiety disorder: a randomized double-blind placebo-controlled trial. Curr Rev Clin Exp Pharmacol 2021; 16: 191–6.10.2174/1574884715666200413120413CrossRefGoogle ScholarPubMed
Fulzele, AV, Ingle, N, Huda, MN, Mishra, DS. Comparative study to evaluate the effect of a herbal preparation & shirodhara in the management of major depressive disorder. Int J Pharm Res 2014; 6: 506–11.Google Scholar
Hosseini, N, Moharari, F, Soltanifar, A, Mohammad Poor, A, Hosseini, A, Zahedian, F. Evaluation of the effectiveness of Withania somnifera root extract on the anxiety symptoms among children with ADHD in mashhad. Pak J Med Health Sci 2019; 13: 210–6.Google Scholar
Khyati, S, Anup, B. A randomized double blind placebo controlled study of ashwagandha on generalized anxiety disorder. Int Ayurvedic Med J 2013; 1: 17.Google Scholar
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 ScholarPubMed
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: 113276.10.1016/j.jep.2020.113276CrossRefGoogle ScholarPubMed
Majeed, M, Nagabhushanam, K, Murali, A, Vishwanathan, DT, Mamidala, RV, Mundkur, L. A standardized Withania somnifera (Linn.) root extract with piperine alleviates the symptoms of anxiety and depression by increasing serotonin levels: a double-blind, randomized, placebo-controlled study. J Integr Complement Med 2023; 30: 459–68.10.1089/jicm.2023.0279CrossRefGoogle Scholar
Pandit, S, Srivastav, AK, Sur, TK, Chaudhuri, S, Wang, Y, Biswas, TK. Effects of Withania somnifera extract in chronically stressed adults: a randomized controlled trial. Nutrients 2024; 16: 1293.10.3390/nu16091293CrossRefGoogle ScholarPubMed
Gannon, JM, Brar, J, Rai, A, Chengappa, KNR. Effects of a standardized extract of Withania somnifera (Ashwagandha) on depression and anxiety symptoms in persons with schizophrenia participating in a randomized, placebo-controlled clinical trial. Ann Clin Psychiatry 2019; 31: 123–9.10.1177/104012371903100208CrossRefGoogle Scholar
Pratte, MA, Nanavati, KB, Young, V, Morley, CP. An alternative treatment for anxiety: a systematic review of human trial results reported for the ayurvedic herb ashwagandha (Withania somnifera). J Altern Complement Med 2014; 20: 901–8.10.1089/acm.2014.0177CrossRefGoogle ScholarPubMed
Vaidya, VG, Naik, NN, Ganu, G, Parmar, V, Jagtap, S, Saste, G, et al. Clinical pharmacokinetic evaluation of Withania somnifera (L.) Dunal root extract in healthy human volunteers: a non-randomized, single dose study utilizing UHPLC-MS/MS analysis. J Ethnopharmacol 2023; 322: 117603.10.1016/j.jep.2023.117603CrossRefGoogle ScholarPubMed
Huang, W, Chen, Z, Wang, Q, Lin, M, Wu, S, Yan, Q, et al. Piperine potentiates the antidepressant-like effect of trans-resveratrol: involvement of monoaminergic system. Metab Brain Dis 2013; 28: 585–95.10.1007/s11011-013-9426-yCrossRefGoogle ScholarPubMed
Wattanathorn, J, Chonpathompikunlert, P, Muchimapura, S, Priprem, A, Tankamnerdthai, O. Piperine, the potential functional food for mood and cognitive disorders. Food Chem Toxicol 2008; 46: 3106–10.10.1016/j.fct.2008.06.014CrossRefGoogle ScholarPubMed
Baker, C, Kirby, JB, O’Connor, J, Lindsay, KG, Hutchins, A, Harris, M. The perceived impact of Ashwagandha on stress, sleep quality, energy, and mental clarity for college students: qualitative analysis of a double-blind randomized control trial. J Med Food 2022; 25: 1095–101.10.1089/jmf.2022.0042CrossRefGoogle ScholarPubMed
Marchi, M, Galli, G, Magarini, FM, Mattei, G, Galeazzi, GM. Sarcosine as an add-on treatment to antipsychotic medication for people with schizophrenia: a systematic review and meta-analysis of randomized controlled trials. Expert Opin Drug Metabol Toxicol 2021; 17: 483–93.10.1080/17425255.2021.1885648CrossRefGoogle Scholar
Magalhães, PVS, Dean, O, Andreazza, AC, Berk, M, Kapczinski, F. Antioxidant treatments for schizophrenia. Cochrane Database Syst Rev 2016; 2: CD008919.Google ScholarPubMed
Alonso, J, Liu, Z, Evans-Lacko, S, Sadikova, E, Sampson, N, Chatterji, S, et al. Treatment gap for anxiety disorders is global: results of the World Mental Health Surveys in 21 countries. Depress Anxiety 2018; 35: 195208.10.1002/da.22711CrossRefGoogle ScholarPubMed
Arias de la Torre, J, Vilagut, G, Ronaldson, A, Bakolis, I, Dregan, A, Martín, V, et al. Prevalence and variability of depressive symptoms in Europe: update using representative data from the second and third waves of the European Health Interview Survey (EHIS-2 and EHIS-3). The Lancet Public Health 2023; 8: e889–98.10.1016/S2468-2667(23)00220-7CrossRefGoogle ScholarPubMed
Morin, CM, Jarrin, DC. Epidemiology of insomnia: prevalence, course, risk factors, and public health burden. Sleep Med Clin 2022; 17: 173–91.10.1016/j.jsmc.2022.03.003CrossRefGoogle ScholarPubMed
Chellappa, SL, Aeschbach, D. Sleep and anxiety: from mechanisms to interventions. Sleep Med Rev 2022; 61: 101583.10.1016/j.smrv.2021.101583CrossRefGoogle ScholarPubMed
Johnson, EO, Roth, T, Breslau, N. The association of insomnia with anxiety disorders and depression: exploration of the direction of risk. J Psychiatr Res 2006; 40: 700–8.10.1016/j.jpsychires.2006.07.008CrossRefGoogle ScholarPubMed
Etkin, A, Wager, TD. Functional neuroimaging of anxiety: a meta-analysis of emotional processing in PTSD, social anxiety disorder, and specific phobia. Am J Psychiatry 2007; 164: 1476–88.10.1176/appi.ajp.2007.07030504CrossRefGoogle ScholarPubMed
Chen, W-Y, Liu, H-C, Cheng, Y-C, Li, H, Huang, C-C, Ding, Y-W, et al. Effect of pharmacological and neurostimulation interventions for cognitive domains in patients with bipolar disorder: a systematic review and network meta-analysis of randomized controlled trials. Clin Epidemiol 2021; 13: 1039–49.10.2147/CLEP.S335584CrossRefGoogle ScholarPubMed
Lopresti, AL, Smith, SJ. Ashwagandha (Withania somnifera) for the treatment and enhancement of mental and physical conditions: a systematic review of human trials. J Herbal Med 2021; 28: 100434.10.1016/j.hermed.2021.100434CrossRefGoogle Scholar
Zalpour, A, Zare, E, Nasiri, M. The effects of herbal medicines on sexual function and sexual satisfaction among women: a systematic review and meta-analysis. Jundishapur J Nat Pharm Prod 2023; 18.Google Scholar
Tandon, N, Yadav, SS. Safety and clinical effectiveness of Withania somnifera (Linn.) dunal root in human ailments. J Ethnopharmacol 2020; 255: 112768.10.1016/j.jep.2020.112768CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1 Preferred Reporting Items for Systematic Reviews and Meta-analyses flow diagram.

Figure 1

Table 1 Characteristics of the included studies

Figure 2

Fig. 2 Forest plot of anxiety among Withania somnifera and control groups: overall and by diagnostic subgroup. ADHD, attention-deficit and hyperactivity disorder; BD, bipolar disorder; IV, inverse variance; WS, Withania somnifera. The measures of anxiety across the studies were scaled as lower scores indicate lower anxiety symptoms. Reference to the included studies (in alphabetical order): Andrade et al, 200041; Chengappa et al, 201342; Cooley et al, 200945; Fuladi et al, 202146; Hosseini et al, 201948; Khyati et al, 201349; Langade et al, 201950; Langade et al 202151; Majeed et al, 202352; Pandit et al, 2024.53

Figure 3

Fig. 3 Bayesian forest plot of study-level and pooled posterior estimates of Withania somnifera efficacy on anxiety. The measures of anxiety across the studies were scaled as lower scores indicate lower anxiety symptoms. Reference to the included studies (in alphabetical order): Andrade et al, 200041; Chengappa et al, 201342; Cooley et al, 200945; Fuladi et al, 202146; Hosseini et al, 201948; Khyati et al, 201349; Langade et al, 201950; Langade et al 202151; Majeed et al, 202352; Pandit et al, 2024.53

Figure 4

Fig. 4 Forest plot of depression among Withania somnifera and control groups: overall and by diagnostic subgroup. BD, bipolar disorder; IV, inverse variance; MDD, major depressive disorder; SZ, schizophrenia; WS, Withania somnifera. The measures of depression across the studies were scaled as lower scores indicate lower depression symptoms. Reference to the included studies (in alphabetical order): Chengappa et al, 201342; Chengappa et al, 201843; Fulzele et al, 201447; Majeed et al, 202352; Pandit et al, 2024.53

Figure 5

Fig. 5 Bayesian forest plot of study-level and pooled posterior estimates of Withania somnifera efficacy on depression. The measures of depression across the studies were scaled as lower scores indicate lower depression symptoms. Reference to the included studies (in alphabetical order): Chengappa et al, 201342; Chengappa et al, 201843; Fulzele et al, 201447; Majeed et al, 202352; Pandit et al, 2024.53

Figure 6

Fig. 6 Forest plot of sleep quality among Withania somnifera and control groups. IV: inverse variance; WS, Withania somnifera. The measures of sleep quality across the studies were scaled as lower scores indicate better sleep quality. Reference to the included studies (in alphabetical order): Langade et al, 201950; Langade et al 202151; Majeed et al, 202352; Pandit et al, 2024.53

Figure 7

Fig. 7 Forest plot of perceived stress among Withania somnifera and control groups. IV: inverse variance; WS: Withania somnifera. The measures of stress across the studies were scaled as lower scores indicate lower perceived stress. Reference to the included studies (in alphabetical order): Chengappa et al, 201843; Choudhary et al, 201744; Pandit et al, 2024.53

Figure 8

Fig. 8 Forest plots of drop-out due to any cause (Panel A) and any adverse effect (Panel B) among Withania somnifera and control groups. MH: Mantel-Haenszel; WS: Withania somnifera. Lower rates indicate better safety/tolerability. Reference to the included studies (in alphabetical order): Andrade et al, 200041; Chengappa et al, 201342; Chengappa et al, 201843; Choudhary et al, 201744; Cooley et al, 200945; Fuladi et al, 202146; Hosseini et al, 201948; Jahanbakhsh et al, 201617; Langade et al, 201950; Langade et al 202151; Majeed et al, 2023.52

Supplementary material: File

Marchi et al. supplementary material

Marchi et al. supplementary material
Download Marchi et al. supplementary material(File)
File 493.6 KB
Submit a response

eLetters

No eLetters have been published for this article.