ArticlePDF AvailableLiterature Review

Effectiveness of Probiotics, Prebiotics, and Synbiotics in Managing Insulin Resistance and Hormonal Imbalance in Women with Polycystic Ovary Syndrome (PCOS): A Systematic Review of Randomized Clinical Trials

MDPI
Nutrients
Authors:

Abstract and Figures

Background/Objectives: Polycystic ovary syndrome is a common endocrine disorder in women of reproductive age characterized by insulin resistance and hormonal imbalances. Recent research suggests that probiotics and synbiotics may improve these parameters by modulating the gut microbiota. This study systematically reviewed randomized clinical trials evaluating the impact of probiotic, prebiotic, and synbiotic supplementation on insulin resistance and hormonal parameters in women with PCOS. Methods: Exhaustive searches were conducted in PubMed, Cochrane CENTRAL, Scopus, Web of Science, and Embase, following PRISMA guidelines. Randomized trials assessing supplementation with probiotics, prebiotics, or synbiotics for at least 8 weeks in women diagnosed with PCOS according to the Rotterdam criteria were included. Data on participants, interventions, and outcomes related to insulin resistance and hormones were extracted. Results: Eleven studies from Iran involving overweight or obese women aged 15 to 48 were included. Probiotic and synbiotic supplementation showed significant improvements in insulin resistance (reductions in HOMA-IR, fasting glucose, and insulin), lipid profiles (decreased LDL and triglycerides; increased HDL), and hormonal balance (increased SHBG, decreased total testosterone). Synbiotics had more pronounced effects than probiotics or prebiotics alone. Adherence was high, and side effects were minimal. Conclusions: Despite promising results, limitations such as small sample sizes, homogeneous populations, and short intervention durations limit the generalization of the findings. Larger, longer, multicenter trials with diverse populations and standardized methodologies are needed to confirm the efficacy and safety of synbiotics in managing PCOS. Integrating these interventions could improve clinical management and quality of life for affected women, but additional evidence is required to support widespread use.
Content may be subject to copyright.
Citation: Martinez Guevara, D.; Vidal
Cañas, S.; Palacios, I.; Gómez, A.;
Estrada, M.; Gallego, J.; Liscano, Y.
Effectiveness of Probiotics, Prebiotics,
and Synbiotics in Managing Insulin
Resistance and Hormonal Imbalance
in Women with Polycystic Ovary
Syndrome (PCOS): A Systematic
Review of Randomized Clinical Trials.
Nutrients 2024,16, 3916. https://
doi.org/10.3390/nu16223916
Academic Editor: Stefano Guandalini
Received: 16 October 2024
Revised: 12 November 2024
Accepted: 14 November 2024
Published: 16 November 2024
Copyright: © 2024 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
Systematic Review
Effectiveness of Probiotics, Prebiotics, and Synbiotics in
Managing Insulin Resistance and Hormonal Imbalance in
Women with Polycystic Ovary Syndrome (PCOS): A Systematic
Review of Randomized Clinical Trials
Darly Martinez Guevara, Sinthia Vidal Cañas, Isabela Palacios, Alejandra Gómez, María Estrada, Jonathan Gallego
and Yamil Liscano *
Grupo de Investigación en Salud Integral (GISI), Departamento Facultad de Salud, Universidad Santiago de Cali,
Cali 5183000, Colombia; darly.martinez00@usc.edu.co (D.M.G.); sinthia.vidal00@usc.edu.co (S.V.C.);
isabela.palacios00@usc.edu.co (I.P.); alejandra.gomez01@usc.edu.co (A.G.); maria.estrada03@usc.edu.co (M.E.);
jhonatan.gallego00@usc.edu.co (J.G.)
*Correspondence: yamil.liscano00@usc.edu.co
Abstract: Background/Objectives: Polycystic ovary syndrome is a common endocrine disorder in
women of reproductive age characterized by insulin resistance and hormonal imbalances. Recent
research suggests that probiotics and synbiotics may improve these parameters by modulating the gut
microbiota. This study systematically reviewed randomized clinical trials evaluating the impact of
probiotic, prebiotic, and synbiotic supplementation on insulin resistance and hormonal parameters in
women with PCOS. Methods: Exhaustive searches were conducted in PubMed, Cochrane CENTRAL,
Scopus, Web of Science, and Embase, following PRISMA guidelines. Randomized trials assessing
supplementation with probiotics, prebiotics, or synbiotics for at least 8 weeks in women diagnosed
with PCOS according to the Rotterdam criteria were included. Data on participants, interventions,
and outcomes related to insulin resistance and hormones were extracted. Results: Eleven studies
from Iran involving overweight or obese women aged 15 to 48 were included. Probiotic and synbiotic
supplementation showed significant improvements in insulin resistance (reductions in HOMA-IR,
fasting glucose, and insulin), lipid profiles (decreased LDL and triglycerides; increased HDL), and
hormonal balance (increased SHBG, decreased total testosterone). Synbiotics had more pronounced
effects than probiotics or prebiotics alone. Adherence was high, and side effects were minimal.
Conclusions: Despite promising results, limitations such as small sample sizes, homogeneous
populations, and short intervention durations limit the generalization of the findings. Larger, longer,
multicenter trials with diverse populations and standardized methodologies are needed to confirm
the efficacy and safety of synbiotics in managing PCOS. Integrating these interventions could improve
clinical management and quality of life for affected women, but additional evidence is required to
support widespread use.
Keywords: probiotics; insulin resistance; hormonal imbalance; polycystic ovary syndrome (PCOS);
gut microbiota; synbiotics; metabolic health; women’s health; systematic review; randomized
controlled trials
1. Introduction
Polycystic ovary syndrome (PCOS) is a prevalent endocrine disorder affecting 5% to
10% of women of reproductive age, characterized by hyperandrogenism, ovulatory dys-
function, and a polycystic ovarian morphology [
1
,
2
]. Diagnosis is commonly based on the
Rotterdam criteria, which require at least two of the following: oligo- or anovulation, clini-
cal and/or biochemical signs of hyperandrogenism, and a polycystic ovarian morphology
detected by ultrasound [
3
5
]. PCOS phenotypes (A–D) vary in metabolic profiles and risks.
Hyperandrogenic phenotypes (OD-HA and HA-PCOM) exhibit adverse metabolic profiles,
Nutrients 2024,16, 3916. https://doi.org/10.3390/nu16223916 https://www.mdpi.com/journal/nutrients
Nutrients 2024,16, 3916 2 of 28
including elevated body mass index (BMI), higher waist-to-hip ratio, insulin resistance,
and dyslipidemia, increasing the risk of metabolic syndrome, type 2 diabetes mellitus, and
cardiovascular diseases [
6
8
]. Phenotypes lacking hyperandrogenism generally present
more favorable metabolic profiles, though some normoandrogenic women may still face
metabolic challenges.
PCOS is associated with various metabolic complications, including a higher preva-
lence of metabolic syndrome, reaching up to 56% in some studies [
9
11
]. Insulin resistance
is particularly common, affecting approximately 60% to 75% of women with PCOS when as-
sessed using the Homeostasis Model Assessment for Insulin Resistance (HOMA-IR)
[1214]
.
Hyperinsulinemia exacerbates hyperandrogenism by decreasing sex hormone-binding glob-
ulin (SHBG), increasing free testosterone levels, and worsening clinical manifestations such
as hirsutism, acne, irregular menstrual cycles, and infertility [
15
,
16
]. Insulin directly stim-
ulates ovarian theca cells to increase testosterone production and affects key enzymes in
androgen synthesis [1619].
Current treatments involve combined hormonal contraceptives to regulate menstrual
cycles and reduce androgen effects [
20
,
21
]. However, these may worsen insulin resistance,
alter glucose metabolism, and negatively affect cardiovascular profiles by increasing triglyc-
eride levels and systemic inflammation markers [
22
,
23
]. Thus, while they alleviate some
PCOS symptoms, they may contribute to metabolic complications, highlighting the need
for more personalized treatments [24,25].
Recent research suggests the gut microbiota significantly influences metabolic and
hormonal pathways associated with PCOS. Dysbiosis may contribute to chronic low-grade
inflammation, exacerbating insulin resistance and hormonal imbalances [
26
]. Women with
PCOS show decreased diversity and altered abundance of specific bacterial taxa compared
to healthy controls [2729].
Similarly, alterations in gut microbiota have been linked to type 2 diabetes, with certain
bacterial genera positively or negatively correlated with the disease [
30
,
31
]. Probiotics, par-
ticularly Lactobacillus and Bifidobacterium species, have emerged as potential biotherapeutics
for managing insulin resistance and metabolic disorder [32,33].
Consequently, prebiotics, probiotics, and synbiotics have gained attention as potential
treatments for PCOS by modulating gut microbiota. Probiotics are live microorganisms
that confer health benefits when consumed in adequate amounts, while prebiotics are non-
digestible substances that promote the growth of beneficial microbes [
34
,
35
]. Synbiotics
combine both to enhance the survival and implantation of beneficial microorganisms. These
interventions may address microbiota imbalances, inflammation, insulin resistance, lipid
profiles, and hormonal dysregulation more effectively than traditional treatments [36].
Multiple randomized controlled trials (RCTs) have assessed the role of probiotics and
synbiotics in women with PCOS, reporting improvements in insulin resistance markers,
reductions in androgen levels, and favorable shifts in lipid profiles [
37
40
]. However,
study designs, bacterial strains, dosages, and intervention durations vary significantly,
making it difficult to compare results and establish guidelines. Many studies lack rigorous
methodological design and are often small-scale. Additionally, diagnostic criteria like the
Rotterdam criteria include patients with diverse phenotypes, contributing to heterogeneity.
Uncertainties remain about how probiotics and synbiotics exert beneficial effects in
PCOS patients. Proposed mechanisms include regulation of gut barrier integrity, suppres-
sion of systemic inflammation, improvement of insulin signaling pathways, and effects
on the hypothalamic–pituitary–ovarian axis [
41
,
42
]. More research is needed to establish
precise mechanisms, optimal bacterial strains, dosages, and treatment durations.
Considering these factors, the present systematic review aims to critically evaluate
and synthesize existing RCTs focused on the effects of probiotics and synbiotics on insulin
resistance and hormonal markers in women with PCOS. We hypothesize that probiotic
and synbiotic interventions significantly improve insulin sensitivity and reduce androgen
levels compared to control groups. This review seeks to provide insights into improving
the pathophysiological understanding and personalized treatment management of PCOS.
Nutrients 2024,16, 3916 3 of 28
2. Materials and Methods
This systematic review was conducted in accordance with the Preferred Reporting
Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines [
43
]. The methodol-
ogy was designed to ensure a comprehensive and unbiased selection of relevant studies
evaluating the effectiveness of probiotics and synbiotics in managing insulin resistance and
hormonal imbalance in women with PCOS.
In women with PCOS (P), how does probiotic or prebiotic supplementation, or the
supplementation of both (I), compared to placebo or standard treatments (C), affect insulin
resistance and hormonal balance, thereby influencing metabolic health and quality of
life (O)?
This study’s Prospero registration number is CRD42024587531.
2.1. Eligibility Criteria
The eligibility criteria for this review are detailed in Table 1below.
Table 1. Eligibility criteria.
Criteria Inclusion Exclusion
Study Design
- Randomized controlled trials (RCTs) investigating
supplementation with probiotics, prebiotics, or
synbiotics in women with PCOS.
- Observational studies, reviews, cohort
studies, case series, and studies with
non-randomized designs.
- Studies without an appropriate control
group.
Participants
- Women diagnosed with PCOS according to the
Rotterdam criteria, NIH criteria, or AES criteria. - Studies including men or postmenopausal
women.
-
Aged approximately 15–45 years (reproductive age).
Interventions
- Supplementation with probiotics, prebiotics, or
synbiotics administered in any form (e.g., capsules,
fermented foods).
- Supplementation with compounds other
than probiotics, prebiotics, or synbiotics (e.g.,
metformin alone without combination with
probiotics).
Duration
- Interventions with a minimum duration of 8 weeks
to adequately assess changes in metabolic and
hormonal parameters.
- Interventions with a duration of less than 8
weeks.
Outcomes
- Studies evaluating effects on insulin resistance (e.g.,
HOMA-IR, fasting insulin, fasting glucose),
hormonal parameters (e.g., testosterone, SHBG,
DHEA-S), and changes in other relevant metabolic
markers (e.g., lipid profile, apelin, hs-CRP).
-
Studies that only evaluated clinical outcomes
such as reproductive function without
assessing insulin resistance or hormonal
parameters.
Language - Studies published in English. - Studies published in languages other than
English.
2.2. Information Sources and Search Strategy
A comprehensive search of electronic databases was conducted to identify relevant
studies. The databases searched included the following:
PubMed;
Cochrane Central Register of Controlled Trials (CENTRAL);
Scopus;
Web of Science;
Embase.
Nutrients 2024,16, 3916 4 of 28
The data were organized using Zotero version 6.0 (accessed on 24 August 2024).
2.3. Search Algorithm
The search strategy combined Medical Subject Headings (MeSH) and free-text terms
related to PCOS, probiotics, prebiotics, synbiotics, insulin resistance, and hormonal param-
eters. Boolean operators (AND, OR) were used to refine the search. An example of the
search strategy used in PubMed is as follows:
(“polycystic ovary syndrome” OR “PCOS”) AND (probiotic OR prebiotic OR synbiotic)
AND (insulin OR “insulin resistance” OR “HOMA-IR” OR “pancreatic
β
cell function” OR
“C reactive protein” OR “hormonal status” OR “testosterone” OR “androgens”)
This search algorithm was adapted for each database according to its specific search
functionalities. Additional searches were conducted by reviewing the reference lists of
relevant articles to identify any studies that might have been missed.
2.4. Study Selection
Two reviewers (D.M.G. and S.V.C.) independently screened the titles and abstracts of
all identified articles for eligibility. Full-text articles were retrieved for studies that appeared
to meet the inclusion criteria or if eligibility was unclear from the abstract. Discrepancies
between the reviewers were resolved through discussion, and if necessary, a third reviewer
(Y.L.) was consulted to reach a consensus.
2.5. Data Extraction
Two reviewers (D.M.G. and S.V.C.) independently extracted information from the
primary studies using a standardized data extraction form. The extracted data included
details of the clinical trial:
Study characteristics: first author, publication year, country, and study design.
Participant characteristics: number of participants, age, BMI, sex (all female partici-
pants), and diagnostic criteria for PCOS.
Intervention details: type of probiotic/prebiotic/synbiotic used, bacterial strains,
dosage, form of administration, and duration of intervention.
Comparator: details of the control group (placebo or standard care).
Outcomes measured: primary outcomes (e.g., insulin resistance markers, hormonal
parameters), secondary outcomes (e.g., lipid profile, inflammatory markers), and
methods of measurement.
Results: main findings, statistical significance, and conclusions drawn by the authors.
Limitations: as reported by the authors.
Subsequently, a third reviewer (Y.L.) verified the integrity and accuracy of the recorded
information.
Figure 1was created with the online R package PRISMA2020 [
44
] (https://estech.
shinyapps.io/prisma_flowdiagram/, accessed on 24 August 2024). Figure 2was created
using R software package ggplot2, version 4.3.0 (https://cran.r-project.org/bin/windows/
base/old/4.3.0/, accessed on 24 August 2024).
Nutrients 2024,16, 3916 5 of 28
Nutrients 2024, 16, x FOR PEER REVIEW 5 of 31
Figure 1. PRISMA owchart. A Cohen’s kappa of 0.95 and 0.82 indicates a high level of agreement
between reviewers, ensuring the validity of the results.
Figure 1. PRISMA flowchart. A Cohen’s kappa of 0.95 and 0.82 indicates a high level of agreement
between reviewers, ensuring the validity of the results.
Nutrients 2024, 16, x FOR PEER REVIEW 6 of 31
Figure 2. Clinical outcomes of prebiotic, probiotic, and synbiotic interventions in patients with
PCOS. (A) Distribution of prebiotics, probiotics, and synbiotics used in interventions. (B) Changes
in HOMA-IR, FBS, and insulin by intervention group. HOMA-IR (Homeostatic Model Assessment
for Insulin Resistance) is marked in red. FBS (fasting blood sugar) is in orange. Insulin is in yellow.
The bars reect how each parameter changed based on the type of supplement used (probiotic, syn-
biotic, prebiotic, and control). (C) Relationship between duration of intervention and change in clin-
ical parameters (Δ). Red dots represent changes in HOMA-IR. Green dots represent changes in FBS.
Blue dots represent changes in insulin [38–40,45,46].
2.6. Risk-of-Bias Assessment
The risk-of-bias assessment for the included studies was conducted independently
by two reviewers (D.M.G. and S.V.C.) using the Cochrane Risk of Bias Tool for Random-
ized Trials [47], with data entered into Review Manager version 5.4
®
(RevMan, The
Cochrane Collaboration, accessed on 24 August 2024). The following domains were eval-
uated:
Random sequence generation (selection bias);
Allocation concealment (selection bias);
Blinding of participants and personnel (performance bias);
Blinding of outcome assessment (detection bias);
Incomplete outcome data (arition bias);
Selective reporting (reporting bias).
For each domain, studies were assessed as having a low, high, or unclear risk of bias
based on predetermined guidelines. Discrepancies in the risk-of-bias assessment were re-
solved through discussion between the reviewers, and if necessary, a third reviewer (Y.L.)
was consulted.
To conrm the consistency of the evaluation process, a subset of the studies was re-
assessed, and Cohen’s kappa coecient was computed using IBM SPSS Statistics version
27.0 (IBM Corp., Armonk, NY, USA; accessed on 24 August 2024) to quantify the agree-
ment between reviewers.
Figure 2. Clinical outcomes of prebiotic, probiotic, and synbiotic interventions in patients with
PCOS. (A) Distribution of prebiotics, probiotics, and synbiotics used in interventions. (B) Changes in
HOMA-IR, FBS, and insulin by intervention group. HOMA-IR (Homeostatic Model Assessment for
Nutrients 2024,16, 3916 6 of 28
Insulin Resistance) is marked in red. FBS (fasting blood sugar) is in orange. Insulin is in yellow. The
bars reflect how each parameter changed based on the type of supplement used (probiotic, synbiotic,
prebiotic, and control). (C) Relationship between duration of intervention and change in clinical
parameters (
). Red dots represent changes in HOMA-IR. Green dots represent changes in FBS. Blue
dots represent changes in insulin [3840,45,46].
2.6. Risk-of-Bias Assessment
The risk-of-bias assessment for the included studies was conducted independently by
two reviewers (D.M.G. and S.V.C.) using the Cochrane Risk of Bias Tool for Randomized
Trials [
47
], with data entered into Review Manager version 5.4
®
(RevMan, The Cochrane
Collaboration, accessed on 24 August 2024). The following domains were evaluated:
Random sequence generation (selection bias);
Allocation concealment (selection bias);
Blinding of participants and personnel (performance bias);
Blinding of outcome assessment (detection bias);
Incomplete outcome data (attrition bias);
Selective reporting (reporting bias).
For each domain, studies were assessed as having a low, high, or unclear risk of bias
based on predetermined guidelines. Discrepancies in the risk-of-bias assessment were
resolved through discussion between the reviewers, and if necessary, a third reviewer (Y.L.)
was consulted.
To confirm the consistency of the evaluation process, a subset of the studies was
reassessed, and Cohen’s kappa coefficient was computed using IBM SPSS Statistics version
27.0 (IBM Corp., Armonk, NY, USA; accessed on 24 August 2024) to quantify the agreement
between reviewers.
2.7. Data Synthesis
Due to the heterogeneity among the included studies in terms of interventions, bac-
terial strains, dosages, and outcomes measured, a qualitative synthesis was conducted.
The results are presented in narrative form, accompanied by tables summarizing the key
characteristics and findings of the studies.
2.8. Ethical Considerations
As this study is a systematic review of the published literature, it did not involve direct
interaction with human subjects or animals and thus did not require ethical approval.
3. Results
3.1. Characteristics of the Included Studies
A total of 514 records were identified acrossfive databases, with 96 duplicates removed,
leaving 418 records. Of these, 20 were excluded based on title and abstract screening, using
a peer review process with a Cohen’s kappa of 0.95, indicating excellent agreement between
the reviewers. Subsequently, 398 records were assessed for eligibility through full-text
reviews. Among these, 150 records were excluded for not meeting relevance criteria, 12
due to insufficient data, and 225 because they did not match the required study type. The
Cohen’s kappa for this phase was 0.90, also reflecting excellent agreement. As a result,
11 articles, all conducted in Iran, were ultimately included in this systematic review, as
shown in Figure 1.
3.2. Overview of Study Results
Table 2below outlines the general characteristics of the 11 studies included in this
review. All studies were randomized clinical trials, with seven being double-blind and
four triple-blind, and all were conducted in Iran among women of reproductive age. The
consistent application of the Rotterdam criteria ensured the uniform selection of patients,
thereby facilitating more robust comparisons across the different studies. This uniform
Nutrients 2024,16, 3916 7 of 28
selection was crucial for minimizing variability in patient populations, ensuring that all
participants met standardized diagnostic criteria for PCOS. Consequently, the studies could
be more reliably compared, allowing for a clearer understanding of the interventions’
effects on metabolic and hormonal outcomes in PCOS.
Table 2. General characteristics of studies.
Study Reference Country Diagnostic Criteria Study Design
Esmaeilinezhad et al., 2018 [38] Iran Rotterdam criteria for PCOS Triple-blind RCT
Shoaei et al., 2021 [39] Iran Rotterdam criteria for PCOS Double-blind RCT
Darvishi et al., 2020 [40] Iran Rotterdam criteria for PCOS Double-blind RCT
Karimi et al., 2018 [45] Iran Rotterdam criteria for PCOS Double-blind RCT
Samimi et al., 2018 [46] Iran Rotterdam criteria for PCOS Double-blind RCT
Esmaeilinezhad et al., 2019 [37] Iran Rotterdam criteria for PCOS Triple-blind RCT
Gholizadeh Shamasbi et al., 2018 [48] Iran Rotterdam criteria for PCOS Triple-blind RCT
Arab et al., 2022 [49] Iran Rotterdam criteria for PCOS Double-blind RCT
Karamali et al., 2018 [50] Iran Rotterdam criteria for PCOS Double-blind RCT
Karimi et al., 2020 [51] Iran Rotterdam criteria for PCOS Double-blind RCT
Nasri et al., 2018 [52] Iran Rotterdam criteria for PCOS Double-blind RCT
3.3. Participant Characteristics
Table 3details the characteristics of the participants in the reviewed studies. All
studies exclusively included women, with sample sizes ranging from 23 to 45 participants.
The ages of the participants varied between 15 and 48 years, and the BMI generally ranged
from 25 to 40 kg/m2, classifying most participants as overweight or obese.
Table 3. Participant characteristics.
Study Reference Participants Size Age (Years) BMI (kg/m2)Baseline Parameters
Esmaeilinezhad
et al., 2018 [38]Women
I
(pomegranate+synbiotic):
23,
I (pomegranate): 23, I
(synbiotic beverage): 23,
C: 23
15–48 25–28
Synbiotic pomegranate juice:
HOMA-IR: 6.32 ±1.32
FBS: 112.04 ±9.41 mg/dL
Insulin: 22.80 ±3.97 µIU/mL
QUICKI: 0.294 ±0.008; pomegranate
juice: HOMA-IR: 6.16 ±1.17
FBS: 112.82 ±12.61 mg/dL
Insulin: 22.15 ±3.48 µIU/mL
QUICKI: 0.295 ±0.007; synbiotic
beverage: HOMA-IR: 6.11 ±1.22
FBS: 112.65 ±8.46 mg/dL
Insulin: 22.02 ±4.32 µIU/mL
QUICKI: 0.295 ±0.008; control:
HOMA-IR: 6.95 ±0.91
FBS: 114.56 ±8.16 mg/dL
Insulin: 24.66 ±3.33 µIU/mL
QUICKI: 0.290 ±0.004
Shoaei et al.,
2021 [39]Women I (probiotic): 36
C (placebo): 36 15–40 25–30
Probiotic: HOMA-IR: 2.11 ±0.21
FBS: 85.7 ±2.6 mg/dL
Insulin: 9.8 ±0.9 µIU/mL
QUICKI: N/A; placebo: HOMA-IR:
2.05 ±0.23
FBS: 86.2 ±2.5 mg/dL
Insulin: 9.7 ±0.8 µIU/mL
QUICKI: N/A
Nutrients 2024,16, 3916 8 of 28
Table 3. Cont.
Study Reference Participants Size Age (Years) BMI (kg/m2)Baseline Parameters
Darvishi et al.,
2020 [40]Women I (synbiotic): 34
C (placebo): 34 20–44 25
Synbiotic: HOMA-IR: 3.06 ±1.35
FBS: 91.32 ±8.07 mg/dL
Insulin: 13.36 ±4.89 µIU/mL
HDL: 45.79 ±12.05 mg/dL; placebo:
HOMA-IR: 2.10 ±1.12
FBS: 89.02 ±9.05 mg/dL
Insulin: 9.46 ±4.64 µIU/mL
HDL: 48.14 ±10.22 mg/dL
Karimi et al.,
2018 [45]Women I (synbiotic): 44
C (placebo): 44 19–37 25
Synbiotic: HOMA-IR: 3.77 ±2.35
FBS: 92 ±9 mg/dL
Apelin 36: 27 ±21 nmol/L
CRP: 6.9 ±5.99 mg/L; placebo:
HOMA-IR: 3.6 ±1.92
FBS: 90 ±9 mg/dL
Apelin 36: 26 ±15 nmol/L
CRP: 4.74 ±4.68 mg/L
Samimi et al.,
2018 [46]Women I (synbiotic): 30
C (placebo): 30 18–40 27–35
Synbiotic: FPG: 92.2 ±6.2 mg/dL
Insulin: 12.9 ±4.2 µIU/mL
HOMA-IR: 3.0 ±1.1
Triglycerides: 146.4 ±56.3 mg/dL
VLDL: 29.3 ±11.2 mg/dL
AIP: 0.49 ±0.20
Placebo: FPG: 94.0 ±5.7 mg/dL
Insulin: 12.1 ±6.3 µIU/mL
HOMA-IR: 2.8 ±1.4
Triglycerides: 138.2 ±37.9 mg/dL
VLDL: 27.6 ±7.6 mg/dL
AIP: 0.44 ±0.16
Esmaeilinezhad
et al., 2019 [37]Women
SPJ (synbiotic
pomegranate juice): 23
PJ (pomegranate juice):
23
SB (synbiotic beverage):
23
PB (placebo beverage): 23
15–48 ~25–28
SPJ: TGs: 171 ±57 mg/dL
TC: 180 ±32 mg/dL
LDL-C: 96 ±35 mg/dL
HDL-C: 50 ±12 mg/dL
SBP: 128 ±7 mmHg
Placebo: TGs: 194 ±67 mg/dL
TC: 194 ±23 mg/dL
LDL-C: 113 ±27 mg/dL
HDL-C: 42 ±10 mg/dL
SBP: 134 ±7 mmHg
Gholizadeh
Shamasbi et al.,
2018 [48]
Women I: 31
C: 31 18–45 25–40
Prebiotic: LDL-C:
106.87 ±34.7 mg/dL
HDL-C: 40.55 ±8.8 mg/dL
Total cholesterol:
166.90 ±38.6 mg/dL
TGs: 96.77 ±35.7 mg/dL
FBS: 80.68 ±12.3 mg/dL
hs-CRP: 4.70 ±2.6 mg/dL
Free testosterone: 1.25 ±0.9 pg/mL
DHEA-S: 3.18 ±2.2 µg/mL
Arab et al.,
2022 [49]Women I: 45
C: 43 15–40 25
Probiotic: SHBG:
36.11 ±10.87 nmol/mL
Total testosterone: 0.42 ±0.14 ng/mL
FAI: 3.24 ±1.1
DHEA-S: 6.9 ±2.8 nmol/L
Karamali et al.,
2018 [50]Women I: 30
C: 30 18–40 25
Probiotic: SHBG: 46.3 ±10.3 nmol/L
Total testosterone: 1.3 ±0.7 ng/mL
mF-G scores: 14.1 ±4.9
hs-CRP: 3546.7 ±1003.1 ng/mL
TAC: 935.5 ±344.8 mmol/L
MDA: 2.1 ±0.4 µmol/L
Nutrients 2024,16, 3916 9 of 28
Table 3. Cont.
Study Reference Participants Size Age (Years) BMI (kg/m2)Baseline Parameters
Karimi et al.,
2020 [51]Women I: 44
C: 44 19–37 25
Synbiotic: LDL: 97 ±19 mg/dL
HDL: 46.44 ±7.69 mg/dL
Total cholesterol (TC):
175.2 ±27.5 mg/dL
Triglycerides (TGs): 139 ±78 mg/dL
Nasri et al.,
2018 [52]Women I: 30
C: 30 18–40 25
Synbiotic: SHBG: 37.3 ±13.1 nmol/L
Total testosterone: 2.8 ±1.3 ng/mL
mF-G scores: 15.3 ±5.6
hs-CRP: 2920 ±2251.2 ng/mL
NO: 39.0 ±3.1 µmol/L
MDA: 2.3 ±0.4 µmol/L
Abbreviations: I = intervention group; C = control group; BMI = body mass index; HOMA-IR = Homeostatic
Model Assessment of Insulin Resistance; FBS = fasting blood glucose; QUICKI = Quantitative Insulin Sensitiv-
ity Check Index; HDL-C = high-density lipoprotein cholesterol; LDL-C = low-density lipoprotein cholesterol;
TGs = triglycerides; VLDL = very low-density lipoprotein; CRP = C-reactive protein; hs-CRP = high-sensitivity C-
reactive protein; SHBG = sex hormone-binding globulin; FAI = free androgen index; DHEA-S = dehydroepiandros-
terone sulfate; mF-G = modified Ferriman–Gallwey scores; TAC = total antioxidant capacity; MDA = malondi-
aldehyde; NO = nitric oxide; SPJ = synbiotic pomegranate juice; PJ = pomegranate juice; SB = synbiotic beverage;
PB = placebo beverage; FPG = fasting plasma glucose; N/A = not applicable; AIP = atherogenic index of plasma;
SBP = systolic blood pressure.
For example, Esmaeilinezhad et al., 2018 [
38
] included 92 women, evenly divided
into four groups of 23 participants each: synbiotic pomegranate juice, pomegranate juice,
synbiotic beverage, and control. The baseline parameters measured included HOMA-IR,
fasting blood glucose (FBS), insulin levels, and Quantitative Insulin Sensitivity Check Index
(QUICKI). In another study, by Shoaei et al., 2021 [
39
], 72 women participated, with 36 in
the probiotic group and 36 in the placebo group. The ages ranged from 15 to 40 years, and
the BMI was between 25 and 30 kg/m
2
. The study evaluated HOMA-IR, FBS, and insulin
as baseline parameters.
Similarly, Darvishi et al., 2020 [
40
] included 68 women, divided into two groups of 34
each (synbiotic and placebo), with ages between 20 and 44 years and a BMI of 25 kg/m
2
or higher. The recorded baseline parameters were HOMA-IR, FBS, insulin, and HDL
cholesterol. Other studies, such as Karimi et al., 2018 [
50
] and 2020 [
51
], Samimi et al.,
2018 [
46
], and Arab et al., 2022 [
49
], followed similar structures, focusing on different
metabolic and hormonal markers according to each study’s specific objectives.
In particular, Karimi et al., 2018 [
50
] and 2020 [
51
] included 30 and 44 women, re-
spectively, evaluating parameters such as SHBG, total testosterone, mF-G scores, hs-CRP,
TAC, and malondialdehyde (MDA). Samimi et al., 2018 [
46
] and Gholizadeh Shamasbi
et al., 2018 [
48
] also incorporated measurements of fasting glucose, insulin, HOMA-IR,
triglycerides, VLDL, AIP, and various inflammatory and hormonal markers. Lastly, Arab
et al., 2022 [
49
] assessed 45 women in the intervention group and 43 in the control group,
measuring SHBG, total testosterone, FAI, and DHEA-S.
3.4. Intervention Details and Comparison Groups
The interventions across the reviewed studies encompassed a variety of probiotic,
prebiotic, and synbiotic formulations designed to address metabolic and hormonal distur-
bances in women with PCOS. These interventions varied in strains, dosages, forms, and
colony-forming units (CFUs), all of which could influence their effectiveness (See Table 4).
Nutrients 2024,16, 3916 10 of 28
Table 4. Intervention details and comparison groups.
Study Reference Prebiotic, Probiotic, or Synbiotic Type Pharmaceutical
Form Dosage Duration Comparison Group
Esmaeilinezhad
et al., 2018 [38]Synbiotic in pomegranate juice (SPJ) Juice 2 L per week 12 weeks
Placebo pomegranate
juice
Shoaei et al.,
2021 [39]
Multistrain probiotic with L. casei,L.
acidophilus,L. rhamnosus,L. bulgaricus,B.
breve,B. longum, and S. thermophilus
Capsule One 500 mg
capsule daily 8 weeks Placebo (starch and
maltodextrin)
Darvishi et al.,
2020 [40]
Synbiotic (Lactobacillus casei,L. rhamnosus,
L. bulgaricus,L. acidophilus,Bifidobacterium
longum, and Streptococcus thermophilus)
and prebiotic (inulin and FOS)
Capsule 1 capsule daily,
500 mg 8 weeks Placebo
Karimi et al.,
2018 [45]
Synbiotic with 7 strains of probiotics (L.
acidophilus,L. casei,L. bulgaricus,L.
rhamnosus,B. longum,B. breve, and S.
thermophilus) and prebiotic inulin
(fructo-oligosaccharide)
Capsule 1 capsule daily,
1000 mg 12 weeks Placebo
Samimi et al.,
2018 [46]
Synbiotic with L. acidophilus,L. casei,B.
bifidum, and 800 mg inulin Capsule
2×109CFU/g
of each strain +
800 mg inulin
daily
12 weeks Placebo
Esmaeilinezhad
et al., 2019 [37]
Synbiotic in pomegranate juice
(Lactobacillus rhamnosus GG, Bacillus
coagulans, and Bacillus indicus)
Juice 300 mL daily 8 weeks Placebo (flavored
water)
Gholizadeh
Shamasbi et al.,
2018 [48]
Prebiotic (dextrin) Powder (diluted
in water) 20 g daily 12 weeks Placebo
(maltodextrin)
Arab et al., 2022
[49]
Multistrain probiotic with multiple strains:
Lactobacillus acidophilus (3 ×1010 CFU/g),
Lactobacillus casei (3 ×109CFU/g),
Lactobacillus rhamnosus (1.5 ×109CFU/g),
Lactobacillus bulgaricus (5 ×108CFU/g),
Bifidobacterium breve (2 ×1010 CFU/g),
Bifidobacterium longum (7 ×109CFU/g),
Streptococcus thermophilus (
3×108CFU/g
)
+ 800 mg inulin
Capsule
One 500 mg
capsule daily (7
strains + 800 mg
inulin)
12 weeks Placebo (starch and
maltodextrin)
Karamali et al.,
2018 [50]
Multistrain probiotic with multiple strains:
Lactobacillus acidophilus (3 ×1010 CFU/g),
Lactobacillus casei (3 ×109CFU/g),
Lactobacillus rhamnosus (1.5 ×109CFU/g),
Lactobacillus bulgaricus (5 ×108CFU/g),
Bifidobacterium breve (2 ×1010 CFU/g),
Bifidobacterium longum (7 ×109CFU/g),
Streptococcus thermophilus (3
×
10
8
CFU/g)
+ 800 mg inulin
Capsule
Two capsules
daily (500 mg
each: 7 strains +
800 mg inulin)
12 weeks Placebo (starch and
maltodextrin)
Karimi et al.,
2020 [51]
Synbiotics with multiple strains:
Lactobacillus acidophilus (3 ×1010 CFU/g),
Lactobacillus casei (3 ×109CFU/g),
Lactobacillus bulgaricus (5 ×108CFU/g),
Lactobacillus rhamnosus (7 ×109CFU/g),
Bifidobacterium longum (1 ×109CFU/g),
Bifidobacterium breve (2 ×1010 CFU/g),
Streptococcus thermophilus (
3×108CFU/g
)
+ inulin (fructooligosaccharide)
Capsules
Two capsules
daily (500 mg
each: 7 strains +
inulin)
12 weeks Placebo (starch and
maltodextrin)
Nasri et al.,
2018 [52]
Synbiotic with multiple strains:
Lactobacillus acidophilus (2 ×109CFU/g),
Lactobacillus casei (2 ×109CFU/g),
Bifidobacterium bifidum (2 ×109CFU/g) +
0.8 g inulin
Capsules
Two 500 mg
capsules daily (3
strains + inulin)
12 weeks Placebo (starch and
maltodextrin)
Abbreviations: SPJ = synbiotic pomegranate juice; CFU = colony-forming units; FOS = fructo-oligosaccharides.
Nutrients 2024,16, 3916 11 of 28
For instance, studies like Esmaeilinezhad et al., 2018 [
38
] and Esmaeilinezhad et al.,
2019 [
37
] utilized synbiotic pomegranate juice, with participants consuming either 2 L per
week or 300 mL daily over periods ranging from 8 to 12 weeks. These interventions were
compared to placebo pomegranate juice or flavored water, allowing for a direct assessment
of the combined effects of probiotics and prebiotics in a liquid medium. The juice form
likely enhances metabolic effects by providing both probiotic benefits and the antioxidant
properties of pomegranate.
Conversely, several studies employed capsule-based interventions. Shoaei et al.,
2021 [39] and Karimi et al., 2018 [45] administered multi-strain probiotics in capsule form,
typically containing strains such as Lactobacillus casei,L. rhamnosus, and Bifidobacterium
longum, delivered in daily doses ranging from 500 mg to 1000 mg over 8 to 12 weeks.
Additionally, studies like Darvishi et al., 2020 [
40
] and Gholizadeh Shamasbi et al., 2018 [
48
]
combined probiotics with prebiotics such as inulin or dextrin in capsule form to enhance
gut flora and overall metabolic health. The prebiotics served as fuel for the probiotics,
potentially increasing their efficacy in modulating the gut microbiota and improving
insulin sensitivity.
Synbiotic capsules were also utilized in studies by Karimi et al., 2020 [
51
] and Nasri
et al., 2018 [
52
], where multi-strain synbiotics were paired with prebiotics like inulin,
administered as two 500 mg doses daily over 12 weeks. These interventions ensured a
controlled dosage, providing participants with a precise amount of beneficial bacteria
and prebiotics. The control groups in these studies typically received placebo capsules
containing starch or maltodextrin, allowing for clear comparisons between active and
inactive treatments.
CFUs varied significantly across studies, influencing the potential efficacy of the
interventions. Studies such as Arab et al., 2022 [
49
] and Karamali et al., 2018 [
50
] employed
high CFU counts, ranging from 10
8
to 10
10
CFU/g for each probiotic strain, potentially
enhancing the probiotic effects compared to studies with lower CFU counts like Nasri
et al., 2018 [
52
]. Dosages also varied, from 500 mg to 1000 mg per capsule, with some
studies administering multiple capsules daily to achieve the desired intake of probiotics
and prebiotics.
The duration of interventions was consistent across most studies, typically lasting
between 8 and 12 weeks, which is generally sufficient to observe meaningful changes in
metabolic and hormonal parameters. All studies included placebo comparison groups to
ensure that observed effects could be attributed to the active interventions. Placebos varied
in form, including starch, maltodextrin, flavored water, or maltodextrin-diluted powders,
depending on the study design.
Despite the variations in probiotic and synbiotic strains, dosages, forms, and CFU
counts, all studies aimed to improve metabolic and hormonal outcomes in women with
PCOS. The diversity in intervention formulations underscores the complexity of determin-
ing the most effective probiotic or synbiotic regimen, as different delivery methods and
strain combinations could lead to varying clinical outcomes. Future research should
aim to standardize these variables to better compare and understand the efficacy of
different formulations.
The reviewed studies present mixed but promising results regarding the effects of pro-
biotics, prebiotics, and synbiotics on insulin resistance, hormonal markers, and metabolic
profiles in women with PCOS. Commonly analyzed metrics included HOMA-IR, fasting
blood glucose, and hormone levels such as testosterone and SHBG.
Esmaeilinezhad et al., 2018 [
38
] (see Table 5) demonstrated that synbiotic pomegranate
juice significantly reduced HOMA-IR, fasting insulin, and glucose, indicating moderate
improvements in insulin sensitivity compared to pomegranate juice or synbiotics alone.
Despite high adherence rates, the study was limited by its small sample size and lack
of long-term follow-up. Similarly, Esmaeilinezhad et al., 2019 [
37
] reported significant
improvements in lipid profiles and cardiovascular markers with synbiotic pomegranate
juice, though they did not measure body composition or gut microbiota changes.
Nutrients 2024,16, 3916 12 of 28
Table 5. Results, adherence, and side effects.
Study
Reference
Post-Intervention
Parameters
Change in
Parameters ()
Comparative Effects
Adherence to
the Intervention Side Effects Primary
Outcomes
Secondary
Outcomes
Measurement
Methods Key Findings Author
Conclusions
Study
Limitations
Esmaeilinezhad
et al., 2018 [
38
]
Synbiotic pomegranate
juice: HOMA-IR:
5.75 ±1.22; pomegranate
juice: HOMA-IR:
6.20 ±1.23; Synbiotic
beverage: HOMA-IR:
5.61 ±0.99
FBS: 111.47 ±6.58 mg/dL
Insulin:
20.36 ±3.35 µIU/mL
QUICKI: 0.29 ±0.007
FBS:
113.68 ±10.63 mg/dL
Insulin:
22.07 ±3.74 µIU/mL
QUICKI: 0.29 ±0.007
FBS: 110.36 ±6.57 mg/dL
Insulin:
21.03 ±3.94 µIU/mL
QUICKI: 0.29 ±0.008;
Control: HOMA-IR:
7.33 ±0.92
FBS: 115.00 ±7.85 mg/dL
Insulin:
25.89 ±3.11 µIU/mL
QUICKI: 0.28 ±0.004
Synbiotic
pomegranate juice:
HOMA-IR: 0.57
FBS: 1.68 mg/dL
Insulin:
1.77 µIU/mL;
pomegranate juice:
HOMA-IR: +0.04
FBS: +0.86 mg/dL
Insulin:
0.08 µIU/mL
QUICKI: 0.00;
synbiotic beverage:
HOMA-IR: 0.50
FBS: 1.18 mg/dL
Insulin:
1.66 µIU/mL
QUICKI: 0.00;
Control: HOMA-IR:
+0.38
FBS: +0.44 mg/dL
Insulin:
+1.23 µIU/mL
QUICKI: 0.01
Synbiotic
pomegranate juice:
significant
improvement
(p< 0.05);
pomegranate juice:
no significant
change; synbiotic
beverage: moderate
improvement
(p< 0.05); control:
no significant
improvement
95% adherence,
as most
participants
completed the
study
None
Insulin
resistance
(HOMA-IR),
fasting
glucose
Testosterone,
insulin
sensitivity,
lipid profile
ELISA for
insulin and
HOMA-IR,
standard
biochemical
analysis
Significant
reduction in
HOMA-IR,
increased
insulin
sensitivity,
decreased
testosterone
Synbiotic
pomegranate
juice improves
insulin
resistance and
hormone levels
in PCOS
Small sample
size, lack of
long-term
follow-up
Shoaei et al.,
2021 [39]
Probiotic: HOMA-IR:
1.9 ±0.2
FBS: 81.5 ±2.1 mg/dL
Insulin:
9.3 ±0.71 µIU/mL
QUICKI: N/A
Placebo: HOMA-IR:
2.00 ±0.22
FBS: 88.3 ±2.7 mg/dL
Insulin: 9.8
±
0.8
µ
IU/mL
QUICKI: N/A
HOMA-IR:
probiotic: 0.21 vs.
placebo: 0.05
FBS: probiotic:
4.15 mg/dL vs.
placebo:
+2.57 mg/dL
Insulin: probiotic:
0.49 µIU/mL vs.
placebo:
+0.34 µIU/mL
Probiotic group
showed
non-significant
changes in FBS,
insulin, and
HOMA-IR (p= 0.7);
however, after
adjusting for
covariates, insulin
reduction was
significant in the
probiotic group
(p= 0.02)
90% adherence,
most
participants
completed the
study
None
Pancreatic
β-cell
function (FBS,
serum insulin,
HOMA-IR,
QUICKI), CRP
(C-reactive
protein)
Insulin, lipid
profile,
hs-CRP
Standard
biochemical
analyses,
immunoassay
for insulin,
HOMA-IR and
QUICKI
calculations
Non-significant
reduction in
FBS, serum
insulin, and
HOMA-IR in
probiotic group;
after adjusting
for covariates,
insulin
reduction was
significant; no
significant
differences in
CRP
Probiotic sup-
plementation
for 8 weeks had
a
non-significant
beneficial effect
on pancreatic
β-cell function
and CRP
Short study
duration, no
glucose
tolerance
tests or
hormonal
evaluations
Nutrients 2024,16, 3916 13 of 28
Table 5. Cont.
Study
Reference
Post-Intervention
Parameters
Change in
Parameters ()
Comparative Effects
Adherence to
the Intervention Side Effects Primary
Outcomes
Secondary
Outcomes
Measurement
Methods Key Findings Author
Conclusions
Study
Limitations
Darvishi et al.,
2020 [40]
Synbiotic: HOMA-IR:
2.58 ±1.15
FBS: 90.08 ±7.90 mg/dL
Insulin:
11.50 ±4.75 µIU/mL
HDL:
47.11 ±12.73 mg/dL;
placebo: HOMA-IR:
3.08 ±1.31
FBS: 94.44 ±9.49 mg/dL
Insulin:
13.17 ±5.29 µIU/mL
HDL:
44.23 ±10.73 mg/dL
HOMA-IR:
synbiotic: 0.47 vs.
placebo: +0.98
FBS: synbiotic:
1.24 mg/dL vs.
placebo:
+5.42 mg/dL
Insulin: synbiotic:
1.86 µIU/mL vs.
placebo:
+3.71 µIU/mL
HDL: synbiotic:
+1.32 mg/dL vs.
placebo:
3.91 mg/dL
Synbiotic group
showed significant
improvement in
HOMA-IR, FBS,
insulin, and HDL
levels (p< 0.05)
compared to placebo
95% adherence,
all participants
completed the
study
None
Glycemic
indices, lipid
profile, obesity
values
Serum apelin
levels
Standard
biochemical
analysis, ELISA,
anthropometric
measurements
Significant
improvements
in glycemic
indices, lipid
profile, and
obesity values;
no changes in
apelin
Synbiotic sup-
plementation
improves
metabolic
factors and
obesity in
women with
PCOS
Short study
duration, no
evaluation of
bacterial
flora or
SCFAs, only
over-
weight/obese
patients
included
Karimi et al.,
2018 [45]
Synbiotic: HOMA-IR:
3.82 ±2.27
FBS: 92 ±11 mg/dL
Apelin 36:
14.4 ±4.5 nmol/L
CRP: 5.2 ±3.9 mg/L;
Placebo: HOMA-IR:
3.8 ±2.46
FBS: 91 ±10 mg/dL
Apelin 36:
18.4 ±9.2 nmol/L
CRP: 4.9 ±4.8 mg/L
HOMA-IR:
synbiotic: +0.05 vs.
placebo: +0.2
FBS: synbiotic:
+0.6 mg/dL vs.
placebo:
+0.95 mg/dL
Apelin 36: synbiotic:
12.6 nmol/L vs.
placebo:
7.6 nmol/L
CRP: synbiotic:
1.7 mg/L vs.
placebo: 0.24 mg/L
Synbiotic group
showed a significant
decrease in apelin 36
levels (p= 0.004)
compared to
placebo. No
significant changes
in metabolic
parameters such as
HOMA-IR, FBS, or
CRP
Approx. 90%
adherence, with
11 participants
lost to follow-up
None
Metabolic
parameters
(fasting
glucose, 2 h
plasma
glucose,
HbA1c,
HOMA-IR,
QUICKI),
fasting insulin,
C-reactive
protein (CRP),
apelin 36
levels
QUICKI, CRP
Standard
biochemical
analysis,
immunotur-
bidimetry for
HbA1c and CRP,
ELISA for apelin
36, HOMA-IR
and QUICKI
calculations
No significant
differences in
metabolic
parameters,
fasting insulin,
or CRP after 12
weeks;
significant
decrease in
apelin 36
Synbiotic sup-
plementation
had no
significant
effects on
metabolic and
inflammatory
parameters;
decrease in
apelin 36
No
examination
of bacterial
flora changes,
potential
reporting
biases
Samimi et al.,
2018 [46]
Synbiotic: FPG:
88.0 ±7.2 mg/dL
Insulin:
10.1 ±3.9 µIU/mL
HOMA-IR: 2.3 ±0.9
Triglycerides:
130.3 ±39.3 mg/dL
VLDL: 26.0 ±7.9 mg/dL
AIP: 0.43 ±0.16
Placebo: FPG:
92.8 ±8.1 mg/dL
Insulin:
13.9 ±5.2 µIU/mL
HOMA-IR: 3.2 ±1.2
Triglycerides:
144.0 ±47.2 mg/dL
VLDL: 28.8 ±9.4 mg/dL
AIP: 0.43 ±0.22
FPG: synbiotic:
4.1 mg/dL vs.
placebo: 1.2 mg/dL
Insulin: synbiotic:
2.8 µIU/mL vs.
placebo:
+1.8 µIU/mL
HOMA-IR:
synbiotic: 0.7 vs.
placebo: +0.4
Triglycerides:
synbiotic:
16.2 mg/dL vs.
placebo: +5.8 mg/dL
VLDL: synbiotic:
3.3 mg/dL vs.
placebo: +1.1 mg/dL
AIP: synbiotic:
0.05 vs. placebo:
0.003
Significant reduction
in insulin,
HOMA-IR,
triglycerides,
VLDL-cholesterol,
and AIP in the
synbiotic group
(p< 0.05) compared
to placebo. No
significant
differences observed
in total cholesterol,
LDL-cholesterol, or
HDL-cholesterol
Approx. 95%
adherence;
4 participants (2
from each group)
were lost to
follow-up due to
personal reasons
None
Glycemic
control
markers
(insulin,
HOMA-IR,
QUICKI)
Lipid profile
(triglycerides,
VLDL-C, AIP)
Standard
biochemical
analyses, ELISA
for insulin,
HOMA-IR and
QUICKI
calculations
Significant
decrease in
serum insulin,
HOMA-IR,
triglycerides,
VLDL
cholesterol, and
AIP; significant
increase in
QUICKI in
synbiotic group
Improvement
in insulin
resistance
markers and
some lipid
parameters
Short
follow-up, no
SCFAs
measured in
stool
Nutrients 2024,16, 3916 14 of 28
Table 5. Cont.
Study
Reference
Post-Intervention
Parameters
Change in
Parameters ()
Comparative Effects
Adherence to
the Intervention Side Effects Primary
Outcomes
Secondary
Outcomes
Measurement
Methods Key Findings Author
Conclusions
Study
Limitations
Esmaeilinezhad
et al., 2019 [
37
]
SPJ: TGs: 26.4 mg/dL
TC: 13.4 mg/dL
LDL-C: 18.9 mg/dL
HDL-C: +10.7 mg/dL
SBP: 5.6 mmHg
Placebo: TGs: +4.0 mg/dL
TC: +4.3 mg/dL
LDL-C: +7.2 mg/dL
HDL-C: 3.7 mg/dL
SBP: +1.5 mmHg
TGs: SPJ:
26.4 mg/dL vs.
placebo: +4.0 mg/dL
TC: SPJ:
13.4 mg/dL vs.
placebo: +4.3 mg/dL
LDL-C: SPJ:
18.9 mg/dL vs.
placebo: +7.2 mg/dL
HDL-C: SPJ:
+10.7 mg/dL vs.
placebo: 3.7 mg/dL
SBP: SPJ:
5.6 mmHg vs.
placebo: +1.5 mmHg
Significant
improvement in
TGs, LDL-C, HDL-C,
and SBP in the SPJ
group compared to
placebo. Increases in
antioxidant capacity
(TAC) and
reductions in
oxidative stress
(MDA) were also
noted
High adherence
(90%);
reminder
messages were
sent weekly, and
empty bottles
were returned to
ensure
compliance
None
Lipid profile,
oxidative
stress (MDA,
TAC), hs-CRP,
blood pressure
Not specified
Standard
biochemical
analysis, ELISA
for hs-CRP,
MDA and TAC
measurements,
blood pressure
Significant
improvements
in lipid profile,
oxidative stress,
inflammation,
and blood
pressure in SPJ,
PJ, and SB
groups
compared to
placebo
Synbiotic
pomegranate
juice improved
metabolic,
oxidative, and
inflammatory
outcomes
No measure-
ment of gut
microbiota
changes or
body
composition
Gholizadeh
Shamasbi
et al., 2018 [
48
]
Prebiotic: LDL-C:
87.35 mg/dL
HDL-C: 46.15 mg/dL
Total cholesterol:
154.71 mg/dL
TGs: 94.22 mg/dL
FBS: 67.68 mg/dL
hs-CRP: 3.11 mg/dL
Free testosterone:
1.06 pg/mL
DHEA-S: 2.77 µg/mL
LDL-C:
29.79 mg/dL
HDL-C:
+5.82 mg/dL
Total cholesterol:
29.98 mg/dL
TGs:
38.50 mg/dL
FBS: 11.24 mg/dL
hs-CRP:
1.75 mg/dL
Free testosterone:
0.32 pg/mL
DHEA-S:
0.7 µg/mL
Significant reduction
in LDL-C, total
cholesterol,
triglycerides, FBS,
hs-CRP, DHEA-S,
and free testosterone
in the prebiotic
group compared to
placebo. HDL-C
increased
significantly in the
prebiotic group
High adherence
(weekly
follow-up calls
ensured
compliance)
Two
participants
experienced
mild
allergies
and discon-
tinued
interven-
tion
Lipid levels,
fasting
glucose,
hs-CRP,
DHEA-S, free
testosterone
Hirsutism,
menstrual
irregularity
Standard
biochemical
analyses, ELISA
for hormones,
Ferriman–
Gallwey scale
Significant
decrease in
LDL-C, total
cholesterol,
triglycerides,
FBS, hs-CRP,
DHEA-S, free
testosterone,
and hirsutism
score;
significant
increase in
HDL-C
Resistant
dextrin
regulates
metabolic
parameters and
androgen levels
in PCOS
Small sample
size,
participants
only over-
weight/obese
Arab et al.,
2022 [49]
Probiotic: SHBG:
40.06 ±9.14 nmol/mL
Total testosterone:
0.41 ±0.15 ng/mL
FAI: 3.22 ±1.2
DHEA-S:
6.84 ±2.9 nmol/L
SHBG: +3.95
nmol/mL
Total testosterone:
0.01 ng/mL
FAI: 0.02
DHEA-S:
0.06 nmol/L
Probiotic
supplementation
significantly
increased SHBG
compared to the
placebo group, but
no significant
changes were
observed in total
testosterone, FAI,
DHEA-S, or clinical
outcomes (acne,
hirsutism)
High adherence:
compliance
monitored via
phone calls, text
messages, and
capsule return
None
Hormonal and
clinical
parameters:
SHBG, LH,
FSH, DHEA-S,
TT, FAI
Acne,
hirsutism
Hormone
profiles by
electrochemilu-
minescence
immunoassays,
clinical signs
evaluated by
standardized
scales
Significant
increase in
SHBG; no
significant
improvements
in other
hormonal or
clinical
parameters
Probiotic sup-
plementation
improved
SHBG but not
other hormonal
or clinical
parameters
Self-report
instead of
bacterial
stool analysis,
short
duration
Nutrients 2024,16, 3916 15 of 28
Table 5. Cont.
Study
Reference
Post-Intervention
Parameters
Change in
Parameters ()
Comparative Effects
Adherence to
the Intervention Side Effects Primary
Outcomes
Secondary
Outcomes
Measurement
Methods Key Findings Author
Conclusions
Study
Limitations
Karamali et al.,
2018 [50]
Probiotic: SHBG:
72.2 ±31.9 nmol/L
Total testosterone:
1.1 ±0.8 ng/mL
mF-G scores: 12.4 ±3.8
hs-CRP:
2396.7 ±1588.6 ng/mL
TAC:
948.3 ±380.2 mmol/L
MDA: 1.9 ±0.6 µmol/L
SHBG:
+25.9 nmol/L
Total testosterone:
0.2 ng/mL
mF-G scores: 1.7
hs-CRP:
1150 ng/mL
TAC: +8.8 mmol/L
MDA:
0.2
µ
mol/L
Probiotic
supplementation
significantly
increased SHBG,
decreased total
testosterone, mF-G
scores, hs-CRP, and
MDA levels, and
increased TAC
compared to the
placebo group. No
significant effects on
DHEA-S or other
metabolic profiles
Compliance
monitored via
capsule count
and daily SMS
reminders
None
Hormonal and
clinical
parameters:
SHBG, LH,
FSH, DHEA-S,
TT, FAI
Acne,
hirsutism
Hormonal
profile:
electrochem-
iluminescence-
based
immunometric
assays,
biomarkers and
clinical signs
evaluated
Significant
improvements
in SHBG,
decrease in
total
testosterone,
and hs-CRP
and TAC
Improvements
in SHBG,
testosterone,
and
inflammatory
markers
Short
duration,
other strain
combina-
tions or
prebiotics not
evaluated
Karimi et al.,
2020 [51]
Synbiotic: LDL:
92 ±19 mg/dL
HDL: 45 ±8 mg/dL
TC: 170 ±24 mg/dL
TGs: 141 ±78 mg/dL
LDL: 5.27 mg/dL
HDL: +1.71 mg/dL
TC: 5.2 mg/dL
(not significant)
TGs: 2.2 mg/dL
(not significant)
Synbiotic
supplementation
significantly
decreased LDL
levels and increased
HDL levels
compared to the
placebo group. No
significant effects
were found for total
cholesterol or
triglycerides
Compliance
monitored via
capsule count
and daily SMS
reminders
None
Lipids and an-
thropometric
measures:
LDL, HDL,
TC, TGs
Anthropometric
indicators:
weight, BMI,
WC, HC,
WHR
Lipid profile: TC,
TGs, HDL
measured by
colorimetric
methods,
anthropometric
indicators
measured with
digital scale
Significant
decrease in
LDL, increase
in HDL; no
differences in
other
anthropometric
measures
Improvements
in LDL and
HDL, no
changes in
other
parameters
Short
duration
limited to 12
weeks,
dietary
reporting
biases
Nasri et al.,
2018 [52]
Synbiotic: SHBG:
57.1 ±48.6 nmol/L
Total testosterone:
2.4 ±0.9 ng/mL
mF-G scores: 14.0 ±4.9
hs-CRP:
1970 ±1442.0 ng/mL
NO: 44.5 ±5.0 µmol/L
MDA: 2.1 ±0.4 µmol/L
SHBG:
+19.8 nmol/L
Total testosterone:
0.4 ng/mL
mF-G scores: 1.3
hs-CRP:
950 ng/mL
NO: +5.5 µmol/L
MDA:
0.2
µ
mol/L
Synbiotic
supplementation
significantly
increased SHBG,
decreased mF-G
scores, FAI, hs-CRP,
and NO levels
compared to the
placebo group. No
significant effects
were found for other
hormonal markers
and biomarkers of
oxidative stress
Compliance
monitored via
capsule count
and daily SMS
reminders.
None
Hormonal,
inflammation,
and oxidative
stress: SHBG,
LH, FSH,
DHEA-S, TT,
FAI
Inflammation
biomarkers:
hs-CRP
Hormonal
profile: ELISA
kits (DiaMetra,
Italy),
biomarkers:
spectrophoto-
metric methods
for NO, TAC,
GSH, MDA
Significant
increase in
SHBG,
significant
decrease in
hs-CRP, NO,
and mF-G
scores
Synbiotics
improved
SHBG, NO,
hs-CRP, and
mF-G scores
Short
duration,
small sample
size, no
comparison
of different
combina-
tions
Abbreviations: HOMA-IR = Homeostatic Model Assessment of Insulin Resistance; FBS = fasting blood glucose; QUICKI = Quantitative Insulin Sensitivity Check Index; HDL-C =
high-density lipoprotein cholesterol; LDL-C = low-density lipoprotein cholesterol; TGs = triglycerides; VLDL = very low-density lipoprotein; CRP = C-reactive protein; hs-CRP =
high-sensitivity C-reactive protein; SHBG = sex hormone-binding globulin; FAI = free androgen index; DHEA-S = dehydroepiandrosterone sulfate; mF-G = modified Ferriman–Gallwey
scores; TAC = total antioxidant capacity; MDA = malondialdehyde; NO = nitric oxide; SPJ = synbiotic pomegranate juice; PJ = pomegranate juice; SB = synbiotic beverage; FPG = fasting
plasma glucose; ELISA = Enzyme-Linked Immunosorbent Assay; SBP = systolic blood pressure; LH = luteinizing hormone; FSH = follicle-stimulating hormone; TT = total testosterone;
WC = waist circumference; HC = hip circumference; WHR = waist-to-hip ratio; GSH = glutathione.
Nutrients 2024,16, 3916 16 of 28
Shoaei et al., 2021 [
39
] found no significant reduction in HOMA-IR; however, a deeper
analysis revealed a potential reduction in insulin levels, which may have been obscured by
the study’s short duration. Darvishi et al., 2020 [
40
] noted improvements in glycemic indices
and HDL cholesterol with synbiotics, though the absence of gut microbiota data limited
mechanistic insights. Conversely, Karimi et al., 2018 [
45
] observed no significant changes in
HOMA-IR or FBS but found a decrease in apelin 36, suggesting an anti-inflammatory effect
rather than direct metabolic impacts.
Samimi et al., 2018 [
46
] highlighted synbiotics’ positive effects on insulin sensitivity
and lipid metabolism, although total cholesterol and LDL levels remained unchanged.
Gholizadeh Shamasbi et al., 2018 [
48
] demonstrated prebiotics’ ability to lower LDL, total
cholesterol, and inflammatory markers, while also reducing free testosterone, indicat-
ing potential benefits for hyperandrogenism. Arab et al., 2022 [
49
] and Karamali et al.,
2018 [
50
] focused on hormonal regulation, showing mixed results in clinical symptoms of
hyperandrogenism but clear improvements in SHBG and testosterone levels.
Karimi et al., 2020 [
51
] and Nasri et al., 2018 [
52
] supported the positive effects of
synbiotics on lipid profiles and inflammation, although further research with longer follow-
ups is needed to confirm these findings across diverse populations. Across the studies,
adherence rates were generally high, and side effects were minimal, with only a few
instances of mild allergies reported.
Key findings across the studies include significant reductions in HOMA-IR, improve-
ments in lipid profiles, enhanced hormonal balance, and reductions in inflammatory
markers. However, limitations such as small sample sizes, short durations, and the absence
of comprehensive gut microbiota assessments were common. For instance, Esmaeilinezhad
et al., 2018 [
38
] and Darvishi et al., 2020 [
40
] were limited by their small sample sizes and
lack of long-term follow-up, while studies like Arab et al., 2022 [
49
] and Nasri et al., 2018 [
52
]
faced challenges such as short durations and non-standardized measurement methods.
Figure 2is organized into three sections: the distribution of intervention types, their
impact on specific clinical markers, and the relationship between intervention duration and
achieved effects.
In Figure 2A, a pie chart illustrates the distribution of intervention types across the
studies. Notably, combined probiotics and prebiotics (synbiotics) were the most frequently
used interventions, comprising 70% of the total. Probiotics alone accounted for 20%, while
prebiotics alone represented only 10%. This distribution highlights a strong preference
for synbiotic treatments, likely due to the anticipated synergistic benefits of combining
probiotics with prebiotics for broader metabolic improvements.
Figure 2B depicts the changes in three key clinical parameters related to insulin
resistance: HOMA-IR, FBS, and insulin levels. Synbiotics demonstrated the most significant
impact, with notable reductions in all three parameters. Specifically, HOMA-IR decreased
by
0.57, indicating enhanced insulin sensitivity. Additionally, the synbiotic group showed
the greatest reductions in both insulin and FBS, underscoring its potential effectiveness.
Probiotic interventions also yielded improvements, albeit to a lesser extent, with moderate
reductions in HOMA-IR and insulin levels. Prebiotics alone had a smaller yet beneficial
effect. In contrast, the placebo group experienced an increase in HOMA-IR by +1.23,
reflecting a rise in insulin resistance without any therapeutic intervention.
Lastly, Figure 2C explores the relationship between the duration of the interventions
and the changes in clinical outcomes. The data reveal that longer intervention periods,
typically around 12 weeks, are associated with greater improvements in HOMA-IR, FBS,
and insulin levels. This suggests that extended treatment durations are crucial for achieving
substantial metabolic benefits. While shorter interventions, usually lasting 8 weeks, also
resulted in improvements, the effects were less pronounced compared to longer durations.
Therefore, the figure underscores the importance of sustained treatment to maximize
improvements in insulin sensitivity and glycemic control.
Overall, Figure 2emphasizes the superior effectiveness of synbiotic interventions in
improving metabolic markers in women with PCOS, particularly when administered over
Nutrients 2024,16, 3916 17 of 28
longer periods. The preference for combined probiotic and prebiotic treatments, along with
the observed dose–response relationship between intervention duration and metabolic
outcomes, highlights the potential of synbiotics as a comprehensive approach to managing
PCOS-related metabolic disturbances.
3.5. Risk-of-Bias Assessment Results
The risk-of-bias assessment for the studies, taken from the RevMan 5.4 analysis, shows
that the overall quality of the included trials was robust. Most domains in Figure 3present
a low risk of bias, and this is especially true for domains like the generation of random
sequences, where almost all studies had appropriate randomization methods to avoid
selection bias. Allocation concealment was also ingeniously handled, as most of the studies
scored as having low risk, meaning the actual process of allocating participants to either
intervention or control was well-blinded to reduce selection bias. However, in some
instances, allocation concealment was rated as unclear due to the need for further explicit
reporting in the studied papers about how this process was conducted; hence, a slight
doubt on selection bias remained.
Nutrients 2024, 16, x FOR PEER REVIEW 20 of 31
3.5. Risk-of-Bias Assessment Results
The risk-of-bias assessment for the studies, taken from the RevMan 5.4 analysis,
shows that the overall quality of the included trials was robust. Most domains in Figure 3
present a low risk of bias, and this is especially true for domains like the generation of
random sequences, where almost all studies had appropriate randomization methods to
avoid selection bias. Allocation concealment was also ingeniously handled, as most of the
studies scored as having low risk, meaning the actual process of allocating participants to
either intervention or control was well-blinded to reduce selection bias. However, in some
instances, allocation concealment was rated as unclear due to the need for further explicit
reporting in the studied papers about how this process was conducted; hence, a slight
doubt on selection bias remained.
Figure 3. Cochrane risk-of-bias assessment for randomized studies of interventions in this
systematic review. (A) Risk-of-bias summary: Review of the authors’ judgments about each risk-of-
bias item for each included study. The symbol “+” indicates a low risk of bias, the symbol “?”
indicates an unclear risk of bias. The colors used are green for low risk of bias, yellow for unclear
risk of bias [37–40,45,46,48–52]. (B) Risk-of-bias graph: Review of the authors’ judgments about each
risk-of-bias item presented as percentages across all included studies. Figure created by RevMan 5
(accessed on 24 August 2024).
Performance bias, or blinding of participants and personnel, was a less easily
surmountable problem. A number of this review’s component studies, including many of
the most robustly designed, were considered to be at high risk. This was largely due to
the inherent nature of the clinical trials in this eld, since maintaining blinding for
participants and personnel was dicult because of obvious dierences between active
interventions (probiotics, synbiotics, or prebiotics) and placebos. Lack of blinding may
Figure 3. Cochrane risk-of-bias assessment for randomized studies of interventions in this systematic
review. (A) Risk-of-bias summary: Review of the authors’ judgments about each risk-of-bias item
for each included study. The symbol “+” indicates a low risk of bias, the symbol “?” indicates
an unclear risk of bias. The colors used are green for low risk of bias, yellow for unclear risk of
bias
[3740,45,46,4852]
. (B) Risk-of-bias graph: Review of the authors’ judgments about each risk-of-
bias item presented as percentages across all included studies. Figure created by RevMan 5 (accessed
on 24 August 2024).
Nutrients 2024,16, 3916 18 of 28
Performance bias, or blinding of participants and personnel, was a less easily sur-
mountable problem. A number of this review’s component studies, including many of
the most robustly designed, were considered to be at high risk. This was largely due
to the inherent nature of the clinical trials in this field, since maintaining blinding for
participants and personnel was difficult because of obvious differences between active
interventions (probiotics, synbiotics, or prebiotics) and placebos. Lack of blinding may
introduce performance bias, which could potentially influence the behavior of participants
or the expectations of the researchers [53].
On the other hand, blinding of outcome assessment or detection bias was generally
well managed in most studies. In most trials, the assessors of clinical outcomes were blinded
to the intervention groups, which minimized the occurrence of biased measurements of
outcome. Attrition bias or incomplete outcome data showed consistent low risk across the
studies. Most trials reported participant dropouts and addressed missing data adequately,
with no studies displaying a high risk of bias in this domain. Selective reporting was
consistently rated as low risk across all studies, indicating comprehensive reporting of
both positive and negative outcomes. The studies demonstrated minimal external factors
influencing the results, as reflected by the low risk of other biases.
When we combine these results with the risk-of-bias assessment using the Jadad scale
(see Table 6), it is clear that the included studies are methodologically strong and have
minimal risk of bias that could undermine their findings. Every study scored a perfect 5
out of 5 on the Jadad scale, demonstrating excellent quality. This top score highlights their
rigorous randomization, effective blinding, and transparent handling of withdrawals and
dropouts, all of which make their results more reliable and trustworthy.
Table 6. Jadad scale assessment.
Study Name Randomization
(0–2) Blinding (0–2)
Withdrawals/Dropouts
(0–1)
Total Score (Out of 5)
Esmaeilinezhad et al., 2018 [38] 2 2 1 5
Shoaei et al., 2021 [39] 2 2 1 5
Darvishi et al., 2020 [40] 2 2 1 5
Karimi et al., 2018 [45] 2 2 1 5
Samimi et al., 2018 [46] 2 2 1 5
Esmaeilinezhad et al., 2019 [37] 2 2 1 5
Gholizadeh Shamasbi et al., 2018 [48] 2 2 1 5
Arab et al., 2022 [49] 2 2 1 5
Karamali et al., 2018 [50] 2 2 1 5
Karimi et al., 2020 [51] 2 2 1 5
Nasri et al., 2018 [52] 2 2 1 5
4. Discussion
4.1. Main Findings
This systematic review sought to answer the following question: How does sup-
plementation with probiotics, prebiotics, or synbiotics, compared to placebo or standard
treatments, affect insulin resistance and hormonal balance in women with PCOS, influenc-
ing their metabolic health and quality of life? The relevance of this study lies in the need
to identify effective and safe interventions that address the characteristic metabolic and
hormonal imbalances of PCOS, a condition that significantly impacts the quality of life of
women of reproductive age.
The results demonstrated multiple benefits associated with probiotic and synbiotic-
based interventions in women with PCOS. Significant reductions were observed in the
HOMA-IR index, indicating substantial improvements in insulin sensitivity. Studies
such as those by Esmaeilinezhad et al., 2018 [
38
], Shoaei et al., 2021 [
39
], and Samimi
et al., 2018 [
46
] reported notable decreases in fasting glucose and insulin, reflecting better
metabolic function.
Nutrients 2024,16, 3916 19 of 28
Additionally, there were improvements in lipid profiles, with reductions in LDL
cholesterol and triglycerides and increases in HDL cholesterol. Research by Esmaeilinezhad
et al., 2019 [
37
] and Karimi et al., 2020 [
51
] supports these findings, suggesting a decrease
in the cardiovascular risk associated with PCOS.
Regarding hormonal balance, increases in SHBG levels and decreases in total testos-
terone were recorded, suggesting a reduction in the hyperandrogenemia typical of PCOS.
Studies such as those by Arab et al., 2022 [
49
] and Karamali et al., 2018 [
50
] demonstrated
improvements in clinical symptoms of hyperandrogenism, which could translate into a
better quality of life for patients. However, the limited number of studies assessing SHBG
and hirsutism requires careful interpretation of these findings, as the evidence is not yet
conclusive. Future research should aim to clarify the effect of probiotics and synbiotics
on SHBG and clinical symptoms such as hirsutism to better understand the therapeutic
potential of these interventions in managing hyperandrogenism in PCOS. According to
the meta-analysis by Shamasbi et al., 2020 [
54
], the use of probiotics and synbiotics in
women with PCOS led to a significant increase in SHBG levels compared to the placebo
group, suggesting an improvement in the hormonal profile. However, regarding hirsutism
symptoms, the same study did not find significant differences between the intervention
and control groups. This indicates that probiotics and synbiotics may not have a direct
impact on reducing hirsutism, and their effect on this condition remains inconclusive based
on current evidence.
Reductions were also observed in inflammatory markers like hs-CRP, highlighting
the anti-inflammatory potential of these interventions [
45
,
52
]. Reducing systemic inflam-
mation is crucial given its role in the pathophysiology of PCOS and its impact on overall
metabolic health.
It is important to note that interventions with synbiotics consistently showed the most
significant benefits compared to probiotics and prebiotics administered separately. Combi-
nations of probiotics and prebiotics offered more robust and comprehensive improvements,
possibly due to the synergy between both components that enhances modulation of the gut
microbiota and optimizes metabolic and hormonal functions [38,40].
The most commonly used probiotic strains included Lactobacillus acidophilus,Lacto-
bacillus casei,Lactobacillus rhamnosus,Bifidobacterium longum,Streptococcus thermophilus, and
Bacillus coagulans. The most common pharmaceutical forms were capsules and synbiotic
juices, with durations ranging between 8 and 12 weeks. Synbiotic combinations produced
more pronounced effects on both insulin resistance and hormonal balance.
Finally, the high adherence rate and minimal reported side effects reinforce the viability
and safety of these interventions. Most studies reported adherence rates above 90%, and
adverse effects were insignificant or nonexistent [
38
,
39
]. This suggests that probiotics and
synbiotics can be effectively integrated into existing treatment regimens, offering a viable
alternative to conventional pharmacological therapies with lower associated risks.
4.2. Comparison with Previous Studies
Our findings are consistent with previous research highlighting the beneficial effects
of probiotics, prebiotics, and synbiotics on metabolic and hormonal parameters in women
with PCOS. Meta-analyses and systematic reviews have demonstrated improvements in
insulin resistance, lipid profiles, and hormonal balance.
For example, Miao et al., 2021 [
55
] concluded that supplementation with probiotics
and synbiotics improved insulin resistance in women with PCOS, supporting the idea that
modulation of the gut microbiota positively influences metabolic and endocrine functions.
Musazadeh et al., 2023 [
56
] found that synbiotics significantly improved lipid profiles and
anthropometric parameters, suggesting a beneficial effect on the management of obesity
and related disorders.
Studies such as those by Cozzolino and Vitagliano, 2019 [
57
] and Karamali et al.,
2018 [
50
] reported that probiotic supplementation was associated with improvements
in metabolic, hormonal, and inflammatory parameters. Probiotics have been found to
Nutrients 2024,16, 3916 20 of 28
increase levels of SHBG, decrease total testosterone, and reduce hs-CRP and MDA. Synbiotic
supplementation also showed beneficial effects on SHBG and inflammatory markers (Nasri
et al., 2018 [52]).
Although some research, such as that by Angoorani et al., 2023 [
41
], suggests that
probiotics are more effective than synbiotics on certain parameters, our findings indicate
that synbiotics may have a broader impact on metabolism. These discrepancies high-
light the need for more research to determine the most effective intervention and under
what circumstances.
Inconsistencies in the literature underscore the importance of strain specificity, dosage,
and treatment duration. McFarland et al., 2018 [
58
] emphasized that different strains within
the same species can have variable effects on health, underscoring the need for specific
clinical guidelines. Our study addresses this gap by identifying probiotic strains that show
greater efficacy in improving metabolic and hormonal parameters in women with PCOS.
4.3. Impact of Probiotics on Insulin Resistance and Hormonal Balance in Polycystic
Ovary Syndrome
Insulin resistance is fundamental in the pathogenesis of PCOS [
59
]. There is a link
between gut microbiota dysbiosis and the development of PCOS, especially related to
insulin resistance and obesity [
60
]. In this context, probiotics and synbiotics have shown
promising effects in improving insulin sensitivity.
These microorganisms modify the composition and diversity of the gut microbiota,
restoring a healthy balance of bacteria that positively impacts metabolic and hormonal path-
ways. Strains such as Lactobacillus and Bifidobacterium have demonstrated improvements
in gut dysbiosis, correlating with more favorable sexual hormone levels and metabolic
parameters [59].
Probiotics increase the production of short-chain fatty acids (SCFAs) like butyrate,
propionate, and acetate, which have anti-inflammatory properties and improve insulin
sensitivity. SCFAs maintain the integrity of the intestinal barrier and modulate immune
responses [
59
,
61
,
62
], activate G protein-coupled receptors, and promote the release of
peptides such as GLP-1 and PYY, contributing to glucose homeostasis [63].
Reducing systemic inflammation is another key mechanism. Chronic low-grade in-
flammation in PCOS interferes with insulin signaling. Probiotics decrease pro-inflammatory
cytokines like TNF-
α
and IL-6, strengthen the intestinal barrier, and reduce the translocation
of lipopolysaccharides into the bloodstream, improving insulin sensitivity [54].
Additionally, probiotics can influence bile acid metabolism, which acts as signaling
molecules in glucose and lipid metabolism through receptors like FXR and TGR5 [
64
].
By modifying this metabolism, they enhance metabolic health [
65
].
In vitro
studies have
shown that probiotic complexes can alter bile acid profiles and microbial composition,
increasing beneficial bacteria and reducing harmful metabolites [66].
Regarding hormonal regulation, probiotics can affect levels of sex hormones such as
testosterone, LH, and FSH. Strains of Lactobacillus and Bifidobacterium have demonstrated
reductions in testosterone and improvements in hormonal profiles, possibly mediated by
the gut–brain axis and microbial modulation of hormonal metabolism [
54
,
59
]. By improving
insulin resistance, they reduce hyperinsulinemia, decreasing ovarian androgen production
and increasing levels of SHBG, which reduces free testosterone [67].
Probiotics also enhance antioxidant capacity by elevating total glutathione and total
antioxidant capacity, mitigating oxidative stress in PCOS [
54
]. They improve lipid pro-
files by reducing triglycerides, total cholesterol, and LDL, benefiting the management of
associated dyslipidemia [68,69].
Clinical evidence supports these mechanisms. Studies have reported significant
reductions in fasting glucose, insulin levels, and HOMA-IR in women with PCOS receiving
probiotic and synbiotic supplementation [
55
]. Decreases in testosterone, increases in SHBG,
and improvements in symptoms like hirsutism and menstrual irregularities were also
observed [50].
Nutrients 2024,16, 3916 21 of 28
Therefore, probiotics positively impact insulin resistance and hormonal balance in
PCOS by modulating the gut microbiota, increasing SCFAs, reducing systemic inflamma-
tion, and improving insulin signaling. These combined effects contribute to better glycemic
and hormonal control, playing a crucial role in the comprehensive management of PCOS.
4.4. Limitations of the Included Studies
Despite these promising findings, it is essential to consider the limitations and risk of
bias present in the reviewed studies to interpret the results accurately.
A key limitation is the small sample size in most studies, which can decrease statistical
power and increase the risk of type II errors [
29
,
70
]. Additionally, all studies were conducted
in Iran and focused on overweight or obese women of reproductive age, limiting the
generalizability of the findings to more diverse populations.
The short duration of the interventions, generally between 8 and 12 weeks, might not
be sufficient to observe their long-term effects or the sustainability of the benefits. Longer
studies are needed to determine whether the improvements are maintained over time and
to evaluate their impact on clinical outcomes such as menstrual regularity and fertility, also
with probiotic doses 1010 CFU/day and without concurrent energy restriction [71].
The lack of comprehensive evaluations of the gut microbiota and body composition
measurements limits the understanding of underlying mechanisms. Moreover, variability
in doses, strains, and forms of the interventions makes it difficult to determine the most
effective regimen and complicates comparisons between studies [
29
,
72
,
73
]. For example,
some studies report an increase in Bacteroides spp. and Lactobacillus spp. in PCOS patients,
while others highlight a decrease in Prevotella spp. and Lachnospira spp. [
29
,
73
]. This
inconsistency in the bacterial strains studied and reported makes it challenging to identify
specific microbial patterns associated with PCOS.
The absence of control and detailed reporting on dietary intake and physical activity
is another limitation, as these factors can influence metabolic and hormonal parameters,
introducing biases in interpretation.
4.5. Limitations of the Review
Despite efforts to conduct a thorough and rigorous systematic review, this research
presents several limitations that should be considered when interpreting the results and
planning future research in this field.
Firstly, there is the possibility of having excluded unpublished studies or studies
published in languages other than English and Spanish. This review was based on searches
in international databases and the literature accessible in these languages, which may have
omitted relevant research published in other languages. This linguistic bias may limit the
completeness of the evidence collected and lead to an overestimation or underestimation
of the effects of the interventions.
Additionally, the inability to perform a quantitative meta-analysis due to heterogeneity
among the included studies is a significant limitation. Differences in study designs, partici-
pant populations, interventions (types and doses of probiotics, prebiotics, and synbiotics),
intervention duration, and outcome measures made it difficult to statistically combine the
data. This heterogeneity impedes the quantitative synthesis of results and limits the ability
to draw more general and robust conclusions about the efficacy of the interventions [
29
,
73
].
Another important limitation is the geographic and demographic homogeneity of the
included studies. Most clinical trials were conducted in Iran and focused on overweight
or obese women of reproductive age diagnosed with PCOS according to the Rotterdam
criteria. This lack of diversity in the studied populations limits the generalizability of the
findings to women of different ethnic, cultural, and socioeconomic backgrounds, as well as
those with different clinical characteristics of PCOS.
Additionally, the variable methodological quality of the included studies represents
a limitation. Although many studies presented low risk of bias in areas such as random
sequence generation and allocation concealment, others showed risks in aspects like blind-
Nutrients 2024,16, 3916 22 of 28
ing, sample size, and follow-up duration. These methodological limitations can influence
the internal validity of the studies and affect confidence in the reported findings.
Finally, the lack of standardized outcome measures and comprehensive evaluations of
the gut microbiota hinders the comparison of studies and the understanding of underlying
mechanisms. The bacterial strains and taxa examined vary across studies, with researchers
often focusing on different bacterial families, genera, or species. Additionally, inconsis-
tencies in methods of assessing insulin resistance, hormonal profiles, and inflammatory
markers further complicate the ability to draw robust and comparable conclusions.
4.6. Clinical Implications
The findings of this systematic review suggest that synbiotic supplementation may be
a valuable complementary strategy in the management of PCOS. Improvements in insulin
resistance, lipid profiles, and hormonal balance indicate that synbiotics could address
central metabolic and endocrine alterations of PCOS.
Variability in individual responses to probiotics and prebiotics highlights the im-
portance of personalized treatment approaches. Factors such as initial gut microbiota
composition, metabolic status, hormonal profiles, dietary habits, and genetic predisposi-
tions can influence the effectiveness of these interventions. Personalizing probiotic and
prebiotic regimens according to each patient’s individual profile can maximize therapeutic
outcomes [7476].
Clinicians should consider the following factors [7478]:
Selection of specific strains: different probiotic strains can have varied effects on
metabolic and hormonal parameters. Choosing strains with demonstrated efficacy
could enhance treatment outcomes.
Dosage and formulation: adjusting the dosage and choosing the appropriate formu-
lation (capsules, powders, functional foods) according to patient preferences and
tolerance can improve adherence and effectiveness.
Comprehensive evaluation: assessing the patient’s overall health status, including
metabolic markers, hormonal levels, and lifestyle factors, can help develop a tailored
supplementation plan.
The personalization of synbiotic interventions could be especially beneficial for pa-
tients who have not responded adequately to standard treatments or who experience
adverse effects from pharmacological therapies. Collaboration among healthcare profes-
sionals is essential to effectively implement personalized strategies [74,79,80].
Moreover, the management of PCOS often requires a multidisciplinary approach that
addresses metabolic, reproductive, and psychological aspects. Synbiotic supplementation
can be integrated into this care model as a complementary therapy. Healthcare professionals
should educate patients about the potential benefits and limitations of probiotics and
prebiotics, emphasizing that these supplements do not replace conventional treatments but
can enhance overall management when used in conjunction [41,81].
Regular monitoring of metabolic and hormonal parameters is necessary to evaluate the
effectiveness of the intervention and adjust treatment as needed. Promoting a balanced diet,
regular physical activity, and adherence to medical therapies will optimize outcomes [
41
,
82
].
Although evidence supports the potential benefits of synbiotic supplementation, clini-
cians should be cautious due to variability in available products. They should consider the
following factors:
Quality and standardization: selecting high-quality products from reputable man-
ufacturers is crucial, as efficacy depends on the viability and concentration of the
strains used.
Regulation: dietary supplements are not always subject to the same regulatory stan-
dards as medications. Clinicians should guide patients toward products proven in
safety and efficacy.
Nutrients 2024,16, 3916 23 of 28
Patient education: informing patients about the importance of adherence, possible
side effects, and realistic expectations will enhance satisfaction and compliance with
the supplementation regimen.
Advances in microbiome research and personalized medicine could offer tools for
more precise interventions. Microbiota profiling could identify specific dysbiosis patterns
in women with PCOS, allowing for targeted probiotic and prebiotic therapies. Additionally,
larger and longer clinical trials are required to establish standardized guidelines and
determine optimal strains, doses, and durations of supplementation [41,8386].
4.7. Recommendations for Future Research
More research is needed to address the current limitations. Future studies should
be larger and multicentric randomized trials including diverse populations. Extending
the duration of trials to six months or more will allow for the evaluation of the long-term
efficacy and sustainability of the benefits.
Incorporating detailed analyses of the gut microbiota and metabolomic evaluations
will help understand the biological mechanisms behind the observed clinical effects. Ad-
vanced techniques such as 16S rRNA gene sequencing and metagenomics can provide
valuable information on how interventions alter microbial composition and metabolic
pathways. Standardizing probiotic strains, doses, and administration forms will facilitate
comparison between studies and the development of evidence-based clinical recommenda-
tions [8789].
Including participants with different manifestations of PCOS will allow us to under-
stand how interventions affect each subgroup and personalizing treatments. Evaluating
clinical outcomes that directly impact quality of life, such as menstrual regularity and
improvement of symptoms like hirsutism, is fundamental.
It is crucial to assess long-term safety and explore strategies to improve adherence,
such as education and support programs. Investigating combination with other therapies
may reveal additional benefits and optimize treatment protocols.
Conducting economic evaluations will help determine the value of these interventions
compared to existing treatments, informing healthcare policy decisions. Incorporating
patient perspectives in the study design will ensure that interventions address their needs,
enhancing comprehensive care.
5. Conclusions
The studies reviewed in this review provide promising evidence on the use of synbi-
otics in the management of PCOS. The results indicate that synbiotic supplementation can
significantly improve insulin resistance, lipid profiles, and hormonal balance in women
with PCOS, suggesting a therapeutic potential to address the metabolic and endocrine
alterations associated with this condition. These improvements can have a positive impact
on the metabolic health and quality of life of patients, offering an alternative or complement
to conventional therapies. However, greater validation is required through future research
that addresses the current limitations.
Although the studies show encouraging results, the current body of evidence is
limited by methodological constraints such as small sample sizes, homogeneity in the
populations studied, relatively short intervention durations, and a lack of standardization
in interventions and measurements. These limitations affect the generalization of the results
and underscore the need to conduct additional studies with more robust and diversified
designs. Only through larger, longer-term studies with standardized methodologies can
the efficacy and safety of synbiotics in this population be confirmed, allowing for their
evidence-based integration into clinical practice.
Nutrients 2024,16, 3916 24 of 28
Author Contributions: Conceptualization, D.M.G. and Y.L.; methodology, D.M.G. and Y.L.; software,
J.G.; validation, D.M.G., M.E. and A.G.; formal analysis, D.M.G.; investigation, D.M.G.; resources,
Y.L. and J.G.; data curation, S.V.C.; writing—original draft preparation, Y.L., S.V.C., D.M.G., I.P., A.G.
and M.E.; writing—review and editing, D.M.G. and I.P.; visualization, I.P.; supervision, Y.L.; project
administration, Y.L.; funding acquisition, Y.L. All authors have read and agreed to the published
version of the manuscript.
Funding: This research has been funded by Dirección General de Investigaciones de la Universidad
Santiago de Cali, Convocatoria Interna No. 01–2024.
Data Availability Statement: Data are contained within the article.
Acknowledgments: This research has been funded by Dirección General de Investigaciones de la
Universidad Santiago de Cali, Convocatoria Interna No. 01–2024. We thank the Universidad Santiago
de Cali for their financial and academic support in the preparation of this manuscript. All individuals
included in this section have given their consent to be acknowledged.
Conflicts of Interest: The authors declare no conflicts of interest.
References
1.
Azziz, R.; Carmina, E.; Chen, Z.; Dunaif, A.; Laven, J.S.E.; Legro, R.S.; Lizneva, D.; Natterson-Horowtiz, B.; Teede, H.J.; Yıldız,
B.O. Polycystic Ovary Syndrome. Nat. Rev. Dis. Primer 2016,2, 16057. [CrossRef]
2.
Salari, N.; Nankali, A.; Ghanbari, A.; Jafarpour, S.; Ghasemi, H.; Dokaneheifard, S.; Mohammadi, M. Global Prevalence of
Polycystic Ovary Syndrome in Women Worldwide: A Comprehensive Systematic Review and Meta-Analysis. Arch. Gynecol.
Obstet. 2024,310, 1303–1314. [CrossRef] [PubMed]
3.
Azziz, R. Controversy in Clinical Endocrinology: Diagnosis of Polycystic Ovarian Syndrome: The Rotterdam Criteria Are
Premature. J. Clin. Endocrinol. Metab. 2006,91, 781–785. [CrossRef] [PubMed]
4.
Teede, H.J.; Tay, C.T.; Laven, J.J.E.; Dokras, A.; Moran, L.J.; Piltonen, T.T.; Costello, M.F.; Boivin, J.; Redman, L.M.; Boyle, J.A.; et al.
Recommendations from the 2023 International Evidence-Based Guideline for the Assessment and Management of Polycystic
Ovary Syndrome. J. Clin. Endocrinol. Metab. 2023,108, 2447–2469. [CrossRef]
5.
Joham, A.E.; Norman, R.J.; Stener-Victorin, E.; Legro, R.S.; Franks, S.; Moran, L.J.; Boyle, J.; Teede, H.J. Polycystic Ovary Syndrome.
Lancet Diabetes Endocrinol. 2022,10, 668–680. [CrossRef] [PubMed]
6.
Wen, X.; Wang, L.; Bai, E. Metabolic Characteristics of Different Phenotypes in Reproductive-Aged Women with Polycystic Ovary
Syndrome. Front. Endocrinol. 2024,15, 1370578. [CrossRef] [PubMed]
7.
Krentowska, A.; Kowalska, I. Metabolic Syndrome and Its Components in Different Phenotypes of Polycystic Ovary Syndrome.
Diabetes Metab. Res. Rev. 2022,38, e3464. [CrossRef]
8.
Moghetti, P.; Tosi, F.; Bonin, C.; Di Sarra, D.; Fiers, T.; Kaufman, J.-M.; Giagulli, V.A.; Signori, C.; Zambotti, F.; Dall’Alda, M.; et al.
Divergences in Insulin Resistance Between the Different Phenotypes of the Polycystic Ovary Syndrome. J. Clin. Endocrinol. Metab.
2013,98, E628–E637. [CrossRef]
9.
Khorshidi, A.; Azami, M.; Tardeh, S.; Tardeh, Z. The Prevalence of Metabolic Syndrome in Patients with Polycystic Ovary
Syndrome: A Systematic Review and Meta-Analysis. Diabetes Metab. Syndr. 2019,13, 2747–2753. [CrossRef]
10.
Lana, M.B.P.; Demayo, S.; Giannone, L.; Nolting, M.; D’isa, E.; Servetti, V.; Rolo, G.; Gutierrez, G.; Jarlip, M. Metabolic Compromise
in Women with PCOS: Earlier than Expected. Rev. Assoc. Med. Bras. 2020,66, 1225–1228. [CrossRef]
11.
Zinnat ara nasreen, S.; Shahreen, S.; Huq, S.; Rahman, S. Polycystic ovary syndrome with metabolic syndrome. J. Med. Case Rep.
Rev. 2021,4, 921–929.
12.
DeUgarte, C.M.; Bartolucci, A.A.; Azziz, R. Prevalence of Insulin Resistance in the Polycystic Ovary Syndrome Using the
Homeostasis Model Assessment. Fertil. Steril. 2005,83, 1454–1460. [CrossRef] [PubMed]
13.
Kim, J.J.; Hwang, K.R.; Oh, S.H.; Chae, S.J.; Yoon, S.H.; Choi, Y.M. Prevalence of Insulin Resistance in Korean Women with
Polycystic Ovary Syndrome According to Various Homeostasis Model Assessment for Insulin Resistance Cutoff Values. Fertil.
Steril. 2019,112, 959–966.e1. [CrossRef]
14.
Stepto, N.K.; Cassar, S.; Joham, A.E.; Hutchison, S.K.; Harrison, C.L.; Goldstein, R.F.; Teede, H.J. Women with Polycystic Ovary
Syndrome Have Intrinsic Insulin Resistance on Euglycaemic-Hyperinsulaemic Clamp. Hum. Reprod. 2013,28, 777–784. [CrossRef]
15.
Pateguana, N.B.; Janes, A.W. The Contribution of Hyperinsulinemia to the Hyperandrogenism of Polycystic Ovary Syndrome. J.
Insul. Resist. 2019,4, 3. [CrossRef]
16.
Moghetti, P. The Multifarious Role of Insulin in PCOS: From Pathophysiology to Therapeutic Management. In Polycystic Ovary
Syndrome; Elsevier: Amsterdam, The Netherlands, 2022.
17.
Kayampilly, P.P.; Wanamaker, B.L.; Stewart, J.A.; Wagner, C.L.; Menon, K.M.J. Stimulatory Effect of Insulin on 5alpha-Reductase
Type 1 (SRD5A1) Expression through an Akt-Dependent Pathway in Ovarian Granulosa Cells. Endocrinology 2010,151, 5030–5037.
[CrossRef]
Nutrients 2024,16, 3916 25 of 28
18.
Munir, I.; Yen, H.-W.; Geller, D.H.; Torbati, D.; Bierden, R.M.; Weitsman, S.R.; Agarwal, S.K.; Magoffin, D.A. Insulin Augmentation
of 17alpha-Hydroxylase Activity Is Mediated by Phosphatidyl Inositol 3-Kinase but Not Extracellular Signal-Regulated Kinase-1/2
in Human Ovarian Theca Cells. Endocrinology 2004,145, 175–183. [CrossRef]
19.
Nestler, J.E.; Jakubowicz, D.J.; de Vargas, A.F.; Brik, C.; Quintero, N.; Medina, F. Insulin Stimulates Testosterone Biosynthesis by
Human Thecal Cells from Women with Polycystic Ovary Syndrome by Activating Its Own Receptor and Using Inositolglycan
Mediators as the Signal Transduction System. J. Clin. Endocrinol. Metab. 1998,83, 2001–2005. [CrossRef]
20.
Halperin, I.J.; Kumar, S.S.; Stroup, D.F.; Laredo, S.E. The Association Between the Combined Oral Contraceptive Pill and Insulin
Resistance, Dysglycemia and Dyslipidemia in Women with Polycystic Ovary Syndrome: A Systematic Review and Meta-Analysis
of Observational Studies. Hum. Reprod. 2011,26, 191–201. [CrossRef]
21. Mosorin, M.-E.; Piltonen, T.; Rantala, A.S.; Kangasniemi, M.; Korhonen, E.; Bloigu, R.; Tapanainen, J.S.; Morin-Papunen, L. Oral
and Vaginal Hormonal Contraceptives Induce Similar Unfavorable Metabolic Effects in Women with PCOS: A Randomized
Controlled Trial. J. Clin. Med. 2023,12, 2827. [CrossRef]
22.
Guedes, J.V.M.; Nunes, N.; Ferreira, L.G.R.; Vilar, T.; Pinheiro, M.D.B.; Domingueti, C.P. Evaluation of Lipid Profile, High-
Sensitivity C-Reactive Protein and D-Dimer in Users of Oral Contraceptives of Different Types. J. Bras. Patol. E Med. Lab. 2018,54,
14–20. [CrossRef]
23.
Ferreira, J.R.D.; Aleluia, M.M.; Figueiredo, C.V.B.; Vieira, L.C.D.L.; Santiago, R.P.; Guarda, C.C.D.; Barbosa, C.G.; Oliveira, R.R.;
Adorno, E.V.; Gonçalves, M. de S. Evaluation of Cardiometabolic Parameters among Obese Women Using Oral Contraceptives.
Front. Endocrinol. 2017,8, 256. [CrossRef] [PubMed]
24.
Adeniji, A.A.; Essah, P.A.; Nestler, J.E.; Cheang, K.I. Metabolic Effects of a Commonly Used Combined Hormonal Oral Contracep-
tive in Women with and Without Polycystic Ovary Syndrome. J. Womens Health 2016,25, 638–645. [CrossRef] [PubMed]
25.
Vrbíková, J.; Cibula, D. Combined Oral Contraceptives in the Treatment of Polycystic Ovary Syndrome. Hum. Reprod. Update
2005,11, 277–291. [CrossRef]
26.
He, F.; Li, Y. Role of Gut Microbiota in the Development of Insulin Resistance and the Mechanism Underlying Polycystic Ovary
Syndrome: A Review. J. Ovarian Res. 2020,13, 73. [CrossRef]
27.
Liu, R.; Zhang, C.; Shi, Y.; Zhang, F.; Li, L.; Wang, X.; Ling, Y.; Fu, H.; Dong, W.; Shen, J.; et al. Dysbiosis of Gut Microbiota
Associated with Clinical Parameters in Polycystic Ovary Syndrome. Front. Microbiol. 2017,8, 324. [CrossRef]
28.
Zeng, B.; Lai, Z.; Sun, L.; Zhang, Z.; Yang, J.; Li, Z.; Lin, J.; Zhang, Z. Structural and Functional Profiles of the Gut Microbial
Community in Polycystic Ovary Syndrome with Insulin Resistance (IR-PCOS): A Pilot Study. Res. Microbiol. 2019,170, 43–52.
[CrossRef]
29.
Li, P.; Shuai, P.; Shen, S.; Zheng, H.; Sun, P.; Zhang, R.; Lan, S.; Lan, Z.; Jayawardana, T.; Yang, Y.; et al. Perturbations in Gut
Microbiota Composition in Patients with Polycystic Ovary Syndrome: A Systematic Review and Meta-Analysis. BMC Med. 2023,
21, 302. [CrossRef]
30.
Gurung, M.; Li, Z.; You, H.N.; Rodrigues, R.R.; Jump, D.B.; Morgun, A.; Shulzhenko, N. Role of Gut Microbiota in Type 2 Diabetes
Pathophysiology. eBioMedicine 2020,51, 102590. [CrossRef]
31.
Huda, M.N.; Kim, M.G.; Bennett, B.J. Modulating the Microbiota as a Therapeutic Intervention for Type 2 Diabetes. Front.
Endocrinol. 2021,12, 632335. [CrossRef]
32.
Panwar, H.; Rashmi, H.M.; Batish, V.K.; Grover, S. Probiotics as Potential Biotherapeutics in the Management of Type 2 Diabetes—
Prospects and Perspectives. Diabetes Metab. Res. Rev. 2013,29, 103–112. [CrossRef] [PubMed]
33.
Tonucci, L.B.; Santos, K.M.O.D.; Ferreira, C.L.D.L.F.; Ribeiro, S.M.R.; Oliveira, L.L.D.; Martino, H.S.D. Gut Microbiota and
Probiotics: Focus on Diabetes Mellitus. Crit. Rev. Food Sci. Nutr. 2017,57, 2296–2309. [CrossRef] [PubMed]
34. Florowska, A.; Hilal, A.; Florowski, T. Prebiotics and Synbiotics. In Probiotics; Academic Press: New York, NY, USA, 2022.
35.
Yadav, M.; Mandeep; Shukla, P. Probiotics of Diverse Origin and Their Therapeutic Applications: A Review. J. Am. Coll. Nutr.
2020,39, 469–479. [CrossRef]
36. Erkmen, O.; Bozo˘glu, T. Probiotics and Prebiotics; John Wiley & Sons: New York, NY, USA, 2016.
37.
Esmaeilinezhad, Z.; Barati-Boldaji, R.; Brett, N.R.; De Zepetnek, J.O.T.; Bellissimo, N.; Babajafari, S.; Sohrabi, Z. The Effect of
Synbiotics Pomegranate Juice on Cardiovascular Risk Factors in PCOS Patients: A Randomized, Triple-Blinded, Controlled Trial.
J. Endocrinol. Investig. 2020,43, 539–548. [CrossRef]
38.
Esmaeilinezhad, Z.; Babajafari, S.; Sohrabi, Z.; Eskandari, M.-H.; Amooee, S.; Barati-Boldaji, R. Effect of Synbiotic Pomegranate
Juice on Glycemic, Sex Hormone Profile and Anthropometric Indices in PCOS: A Randomized, Triple Blind, Controlled Trial.
Nutr. Metab. Cardiovasc. Dis. 2019,29, 201–208. [CrossRef] [PubMed]
39.
Shoaei, T.; Heidari-Beni, M.; Tehrani, H.; Feizi, A.; Esmaillzadeh, A.; Askari, G. Effects of Probiotic Supplementation on
Pancreatic
β
-Cell Function and c-Reactive Protein in Women with Polycystic Ovary Syndrome: A Randomized Double-Blind
Placebo-Controlled Clinical Trial. Int. J. Prev. Med. 2015,6, 27. [CrossRef]
40.
Darvishi, S.; Rafraf, M.; Asaghari Jafarabadi, M.; Farzadi, L. Synbiotic Supplementation Improves Metabolic Factors and Obesity
Values in Women with Polycystic Ovary Syndrome Independent of Affecting Apelin Levels: A Randomized Double-Blind
Placebo-Controlled Clinical Trial. Int. J. Fertil. Steril. 2021,15, 51. [CrossRef]
41.
Angoorani, P.; Ejtahed, H.-S.; Marvasti, F.E.; Taghavi, M.; Ahranjani, B.M.; Hasani-Ranjbar, S.; Larijani, B. The Effects of Probiotics,
Prebiotics, and Synbiotics on Polycystic Ovarian Syndrome: An Overview of Systematic Reviews. Front. Med. 2023,10, 1141355.
[CrossRef]
Nutrients 2024,16, 3916 26 of 28
42.
Calcaterra, V.; Rossi, V.; Massini, G.; Casini, F.; Zuccotti, G.V.; Fabiano, V. Probiotics and Polycystic Ovary Syndrome: A
Perspective for Management in Adolescents with Obesity. Nutrients 2023,15, 3144. [CrossRef]
43.
Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.;
Brennan, S.E.; et al. The PRISMA 2020 Statement: An Updated Guideline for Reporting Systematic Reviews. BMJ 2021,372, n71.
[CrossRef]
44.
Haddaway, N.R.; Page, M.J.; Pritchard, C.C.; McGuinness, L.A. PRISMA2020: An R Package and Shiny App for Producing
PRISMA 2020-Compliant Flow Diagrams, with Interactivity for Optimised Digital Transparency and Open Synthesis. Campbell
Syst. Rev. 2022,18, e1230. [CrossRef] [PubMed]
45.
Karimi, E.; Moini, A.; Yaseri, M.; Shirzad, N.; Sepidarkish, M.; Hossein-Boroujerdi, M.; Hosseinzadeh-Attar, M.J. Effects of
Synbiotic Supplementation on Metabolic Parameters and Apelin in Women with Polycystic Ovary Syndrome: A Randomised
Double-Blind Placebo-Controlled Trial. Br. J. Nutr. 2018,119, 398–406. [CrossRef] [PubMed]
46.
Samimi, M.; Dadkhah, A.; Haddad Kashani, H.; Tajabadi-Ebrahimi, M.; Seyed Hosseini, E.; Asemi, Z. The Effects of Synbiotic
Supplementation on Metabolic Status in Women with Polycystic Ovary Syndrome: A Randomized Double-Blind Clinical Trial.
Probiotics Antimicrob. Proteins 2019,11, 1355–1361. [CrossRef] [PubMed]
47.
Cochrane Cochrane Handbook for Systematic Reviews of Interventions. Available online: https://training.cochrane.org/
handbook (accessed on 9 June 2023).
48.
Gholizadeh Shamasbi, S.; Dehgan, P.; Mohammad-Alizadeh Charandabi, S.; Aliasgarzadeh, A.; Mirghafourvand, M. The Effect of
Resistant Dextrin as a Prebiotic on Metabolic Parameters and Androgen Level in Women with Polycystic Ovarian Syndrome: A
Randomized, Triple-Blind, Controlled, Clinical Trial. Eur. J. Nutr. 2019,58, 629–640. [CrossRef]
49.
Arab, A.; Hossein-Boroujerdi, M.; Moini, A.; Sepidarkish, M.; Shirzad, N.; Karimi, E. Effects of Probiotic Supplementation
on Hormonal and Clinical Outcomes of Women Diagnosed with Polycystic Ovary Syndrome: A Double-Blind, Randomized,
Placebo-Controlled Clinical Trial. J. Funct. Foods 2022,96, 105203. [CrossRef]
50.
Karamali, M.; Eghbalpour, S.; Rajabi, S.; Jamilian, M.; Bahmani, F.; Tajabadi, M.; Keneshlou, F.; Mirhashemi, S.M.; Chamani, M.;
Gelougerdi, S.H.; et al. Effects of Probiotic Supplementation on Hormonal Profiles, Biomarkers of Inflammation and Oxidative
Stress in Women with Polycystic Ovary Syndrome: A Randomized, Double-Blind, Placebo-Controlled Trial. Arch. Iran. Med. 2018,
21, 1–7.
51.
Karimi, E.; Heshmati, J.; Shirzad, N.; Vesali, S.; Hosseinzadeh-Attar, M.J.; Moini, A.; Sepidarkish, M. The Effect of Synbiotics
Supplementation on Anthropometric Indicators and Lipid Profiles in Women with Polycystic Ovary Syndrome: A Randomized
Controlled Trial. Lipids Health Dis. 2020,19, 60. [CrossRef]
52.
Nasri, K.; Jamilian, M.; Rahmani, E.; Bahmani, F.; Tajabadi-Ebrahimi, M.; Asemi, Z. The Effects of Synbiotic Supplementation on
Hormonal Status, Biomarkers of Inflammation and Oxidative Stress in Subjects with Polycystic Ovary Syndrome: A Randomized,
Double-Blind, Placebo-Controlled Trial. BMC Endocr. Disord. 2018,18, 21. [CrossRef]
53.
Rogers, N.; Mousa, S.A. The Shortcomings of Clinical Trials Assessing the Efficacy of Probiotics in Irritable Bowel Syndrome.
J. Altern. Complement. Med. 2012,18, 112–119. [CrossRef]
54.
Shamasbi, S.G.; Ghanbari-Homayi, S.; Mirghafourvand, M. The Effect of Probiotics, Prebiotics, and Synbiotics on Hormonal and
Inflammatory Indices in Women with Polycystic Ovary Syndrome: A Systematic Review and Meta-Analysis. Eur. J. Nutr. 2020,
59, 433–450. [CrossRef]
55.
Miao, Q.; Ma, Y.; Ling, Y.; Jin, W.; Su, Y.; Wang, Q.; Pan, J.; Zhang, Y.; Chen, H.; Yuan, J.; et al. Evaluation of Superinfection,
Antimicrobial Usage, and Airway Microbiome with Metagenomic Sequencing in COVID-19 Patients: A Cohort Study in Shanghai.
J. Microbiol. Immunol. Infect. 2021,54, 808–815. [CrossRef] [PubMed]
56.
Musazadeh, V.; Mohammadi Anilou, M.; Vajdi, M.; Karimi, A.; Sedgh Ahrabi, S.; Dehghan, P. Effects of Synbiotics Supplementa-
tion on Anthropometric and Lipid Profile Parameters: Finding from an Umbrella Meta-Analysis. Front. Nutr. 2023,10, 1121541.
[CrossRef] [PubMed]
57.
Cozzolino, M.; Vitagliano, A. Probiotics and Synbiotics for Polycystic Ovarian Syndrome: A Systematic Review and Meta-Analysis.
Fertil. Steril. 2019,112, e391. [CrossRef]
58.
Mcfarland, L.V.; Evans, C.T.; Goldstein, E.J.C. Strain-Specificity and Disease-Specificity of Probiotic Efficacy: A Systematic Review
and Meta-Analysis. Front. Med. 2018,5, 124. [CrossRef]
59.
He, Y.; Wang, Q.; Li, X.; Wang, G.; Zhao, J.; Zhang, H.; Chen, W. Lactic Acid Bacteria Alleviate Polycystic Ovarian Syndrome by
Regulating Sex Hormone Related Gut Microbiota. Food Funct. 2020,11, 5192–5204. [CrossRef]
60.
Schiattarella, A.; Riemma, G.; Verde, M.L.; Franci, G.; Chianese, A.; Fasulo, D.; Fichera, M.; Gallo, P.; Franciscis, P.D. Polycystic
Ovary Syndrome and Probiotics: A Natural Approach to an Inflammatory Disease. Curr. Women Health Rev. 2021,17, 14–20.
[CrossRef]
61.
Kim, Y.-S.; Keogh, J.B.; Clifton, P.M. Probiotics, Prebiotics, Synbiotics and Insulin Sensitivity. Nutr. Res. Rev. 2017,31, 35–51.
[CrossRef]
62.
Liscano, Y.; Sanchez-Palacio, N. A Critical Look at Omega-3 Supplementation: A Thematic Review. Healthcare 2023,11, 3065.
[CrossRef]
Nutrients 2024,16, 3916 27 of 28
63.
Puddu, A.; Sanguineti, R.; Montecucco, F.; Viviani, G.L. Evidence for the Gut Microbiota Short-Chain Fatty Acids as Key
Pathophysiological Molecules Improving Diabetes. Mediators Inflamm. 2014,2014, 162021. [CrossRef]
64.
Yu, O.; Christ, J.P.; Schulze-Rath, R.; Covey, J.; Kelley, A.; Grafton, J.; Cronkite, D.; Holden, E.; Hilpert, J.; Sacher, F.; et al. Incidence,
Prevalence, and Trends in Polycystic Ovary Syndrome Diagnosis: A United States Population-Based Study from 2006 to 2019.
Am. J. Obstet. Gynecol. 2023,229, 39.e1–39.e12. [CrossRef]
65.
Joyce, S.A.; Gahan, C.G.M. Bile Acid Modifications at the Microbe-Host Interface: Potential for Nutraceutical and Pharmaceutical
Interventions in Host Health. Annu. Rev. Food Sci. Technol. 2016,7, 313–333. [CrossRef] [PubMed]
66.
Liu, W.; Li, Z.; Ze, X.; Deng, C.; Xu, S.; Ye, F. Multispecies Probiotics Complex Improves Bile Acids and Gut Microbiota Metabolism
Status in an In Vitro Fermentation Model. Front. Microbiol. 2024,15, 1314528. [CrossRef] [PubMed]
67.
Nestler, J.E. Role of Hyperinsulinemia in the Pathogenesis of the Polycystic Ovary Syndrome, and Its Clinical Implications. Semin.
Reprod. Endocrinol. 1997,15, 111–122. [CrossRef] [PubMed]
68.
Zafar, U.; Hassan, J.A.; Ismail, K.; Agha, S.; Memon, Z.; Bhatty, S. Effectiveness of Probiotics, Metformin and Their Combination
Therapy in Ameliorating Dyslipidemia Associated with PCOS. J. Pharm. Res. Int. 2019,30, 1–9. [CrossRef]
69.
Gadelha, C.J.M.U.; Bezerra, A.N. Effects of Probiotics on the Lipid Profile: Systematic Review. J. Vasc. Bras. 2019,18, e20180124.
[CrossRef] [PubMed]
70.
Sola-Leyva, A.; Pérez-Prieto, I.; Molina, N.M.; Vargas, E.; Ruiz-Durán, S.; Leonés-Baños, I.; Canha-Gouveia, A.; Altmäe, S.
Microbial Composition Across Body Sites in Polycystic Ovary Syndrome: A Systematic Review and Meta-Analysis. Reprod.
Biomed. Online 2023,47, 129–150. [CrossRef]
71.
Guedes, M.R.; Pontes, K.S.D.S.; Silva, M.I.B.; Neves, M.F.T.; Klein, M.R.S.T. Randomized Controlled Trials Reporting the Effects of
Probiotics in Individuals with Overweight and Obesity: A Critical Review of the Interventions and Body Adiposity Parameters.
Clin. Nutr. 2023,42, 835–847. [CrossRef]
72.
Healey, G.R.; Murphy, R.; Brough, L.; Butts, C.A.; Coad, J. Interindividual Variability in Gut Microbiota and Host Response to
Dietary Interventions. Nutr. Rev. 2017,75, 1059–1080. [CrossRef]
73.
Guo, J.; Shao, J.; Yang, Y.; Niu, X.; Liao, J.; Zhao, Q.; Wang, D.; Li, S.; Hu, J. Gut Microbiota in Patients with Polycystic Ovary
Syndrome: A Systematic Review. Reprod. Sci. 2022,29, 69–83. [CrossRef]
74.
Bubnov, R.V.; Spivak, M.Y. Towards Individualized Use of Probiotics and Prebiotics for Metabolic Syndrome and Associated
Diseases Treatment: Does Pathophysiology-Based Approach Work and Can Anticipated Evidence Be Completed? Probiotics 2018.
[CrossRef]
75.
Ejtahed, H.-S.; Hasani-Ranjbar, S.; Larijani, B. Human Microbiome as an Approach to Personalized Medicine. Altern. Ther. Health
Med. 2017,23, 8–9. [PubMed]
76.
Gibbons, S.M.; Gurry, T.; Lampe, J.W.; Chakrabarti, A.; Dam, V.; Everard, A.; Goas, A.; Gabriele, G.; Kleerebez, M.; Lane, J.; et al.
Perspective: Leveraging the Gut Microbiota to Predict Personalized Responses to Dietary, Prebiotic, and Probiotic Interventions.
Adv. Nutr. 2022,13, 1450–1461. [CrossRef] [PubMed]
77.
Pereira, G.V.D.M.; Coelho, B.D.O.; Júnior, A.I.M.; Thomaz-Soccol, V.; Soccol, C.R. How to Select a Probiotic? A Review and
Update of Methods and Criteria. Biotechnol. Adv. 2018,36, 2060–2076. [CrossRef]
78.
Sniffen, J.; Mcfarland, L.V.; Evans, C.T.; Goldstein, E.J.C. Choosing an Appropriate Probiotic Product for Your Patient: An
Evidence-Based Practical Guide. PLoS ONE 2018,13, e0209205. [CrossRef]
79.
Filippis, F.D.; Vitaglione, P.; Cuomo, R.; Canani, R.B.; Ercolini, D. Dietary Interventions to Modulate the Gut Microbiome—How
Far Away Are We from Precision Medicine. Inflamm. Bowel Dis. 2018,24, 2142–2154. [CrossRef]
80.
Panebianco, C.; Andriulli, A.; Pazienza, V. Pharmacomicrobiomics: Exploiting the Drug-Microbiota Interactions in Anticancer
Therapies. Microbiome 2018,6, 92. [CrossRef] [PubMed]
81.
Tay, C.T.; Moran, L.J.; Wijeyaratne, C.N.; Redman, L.M.; Norman, R.J.; Teede, H.J.; Joham, A.E. Integrated Model of Care for
Polycystic Ovary Syndrome. Semin. Reprod. Med. 2018,36, 86–94. [CrossRef]
82.
Kochar, D.N.; Solanke, M.S.; Chandewar, D.A. Probiotic Interventions for Polycystic Ovarian Syndrome—A Comprehensive
Review. Sch. Int. J. Obstet. Gynecol. 2024,7, 235–245. [CrossRef]
83.
Yurtda¸s, G.; Akdevelio˘glu, Y. A New Approach to Polycystic Ovary Syndrome: The Gut Microbiota. J. Am. Coll. Nutr. 2019,39,
371–382. [CrossRef]
84.
Batra, M.; Bhatnager, R.; Kumar, A.; Suneja, P.; Dang, A.S. Interplay Between PCOS and Microbiome: The Road Less Travelled.
Am. J. Reprod. Immunol. 2022,88, e13580. [CrossRef]
85.
Mosquera, F.E.C.; Guevara-Montoya, M.C.; Serna-Ramirez, V.; Liscano, Y. Neuroinflammation and Schizophrenia: New Thera-
peutic Strategies through Psychobiotics, Nanotechnology, and Artificial Intelligence (AI). J. Pers. Med. 2024,14, 391. [CrossRef]
[PubMed]
86.
Mosquera, F.E.C.; Lizcano Martinez, S.; Liscano, Y. Effectiveness of Psychobiotics in the Treatment of Psychiatric and Cognitive
Disorders: A Systematic Review of Randomized Clinical Trials. Nutrients 2024,16, 1352. [CrossRef] [PubMed]
87.
Dong, S.; Jiao, J.; Jia, S.; Li, G.; Zhang, W.; Yang, K.; Wang, Z.; Liu, C.; Li, D.; Wang, X. 16S rDNA Full-Length Assembly Sequencing
Technology Analysis of Intestinal Microbiome in Polycystic Ovary Syndrome. Front. Cell. Infect. Microbiol. 2021,11, 634981.
[CrossRef] [PubMed]
Nutrients 2024,16, 3916 28 of 28
88.
Yang, T.; Li, G.; Xu, Y.; He, X.; Song, B.; Cao, Y. Characterization of the Gut Microbiota in Polycystic Ovary Syndrome with
Dyslipidemia. BMC Microbiol. 2024,24, 169. [CrossRef]
89.
Yang, Z.; Fu, H.; Su, H.; Cai, X.; Wang, Y.; Hong, Y.; Hu, J.; Xie, Z.; Wang, X. Multi-Omics Analyses Reveal the Specific Changes
in Gut Metagenome and Serum Metabolome of Patients with Polycystic Ovary Syndrome. Front. Microbiol. 2022,13, 1017147.
[CrossRef]
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.
... Probiotics have emerged as potential therapeutic agents for PCOS due to their ability to improve gut microbiota composition, reduce inflammation, enhance metabolic parameters, and modulate hormone levels [6][7][8][9]. By improving gut health and inhibiting harmful microbes, they offer a promising approach to managing both metabolic and reproductive aspects of PCOS [10]. ...
Article
Full-text available
This study amed to investigate the combined use of probiotics and metformin on metabolic parameters in patients with polycystic ovary syndrome (PCOS). 138 patients with PCOS were retrospectively included from January to December 2023 and divided into metformin (n = 70) and probiotics-metformin combination (n = 68) groups based on their clinical treatment regimens. After the three-month intervention, the combination group showed significantly greater improvements compared to the metformin group in high-density lipoprotein cholesterol levels (p < 0.001), insulin levels (p < 0.001), and adiponectin levels (p = 0.033). The combination group also exhibited more significant reductions in weight (p = 0.006), waist circumference (p = 0.037), hip circumference (p = 0.044), and waist-hip ratio (p = 0.031). No significant differences were observed between the groups in changes to other metabolic parameters, hormonal profiles, inflammatory markers, or quality of life assessments (p > 0.05). This retrospective pilot study suggests that the addition of probiotics to metformin therapy may improve HDL levels, insulin sensitivity, anthropometric measurements, and adiponectin levels compared to metformin alone in patients with PCOS.
... The integration of these mechanisms, which include competition for colonization sites, production of bacteriocins, and regulation of the immune response through antiinflammatory mediators, supports the hypothesis that careful selection of specific strains and optimization of the administration route are key factors in achieving an optimal preventive effect against AOM. Each of these studies underscores the importance of evaluating not only the effectiveness of probiotics but also the underlying mechanisms that enable their beneficial action in preventing middle ear infections [59][60][61]. ...
Article
Full-text available
Background and aim: Probiotics, prebiotics, and synbiotics have been documented to modulate the microbiota, enhance immunity, and reduce antibiotic resistance, making them a promising alternative in the management of acute otitis media (AOM). Accordingly, the aim of this study was to determine their effectiveness in the prevention and treatment of AOM in patients. Methods: A systematic review and meta-analysis of randomized controlled trials published between 2000 and 2024 was conducted using Science Direct, PubMed, LILACS, SCOPUS, Web of Science, and Cochrane Clinical Trials, following PRISMA guidelines. The methodological quality was evaluated using the Jadad scale, and the meta-analysis was performed with RevMan 5.4® and Jamovi 2.3.28®. Results: A total of 16 trials with 4034 patients were included. The meta-analysis showed that the intervention did not affect the time to AOM presentation (MD: -7.98; 95% CI: -19.74 to 3.78; p = 0.18), the recurrence of the disease (RR: 0.99; 95% CI: 0.74-1.33; p = 0.96), or the requirement for antibiotics (RR: 1.31; 95% CI: 0.92 to 1.84; p = 0.13); however, it was associated with a reduced probability of developing AOM (RR: 0.80; 95% CI: 0.66 to 0.96; p = 0.02). Subgroup analysis suggests that the effect of probiotic supplementation on AOM incidence is influenced by treatment duration, patient age, and the number of probiotic strains in the product. Conclusions: Supplementation with probiotics, prebiotics, or synbiotics is associated with a significant reduction in the incidence of AOM in children, although no significant impact was observed on other key clinical parameters. These interventions may be considered as a complementary strategy to conventional treatments; however, further high-quality, standardized trials are needed to confirm these findings and to define optimal protocols.
... Lipid metabolism regulation: Decreased levels of low-density lipoprotein cholesterol and triglycerides, accompanied by elevated high-density lipoprotein cholesterol. Sex hormone homeostasis: Increased serum sex hormone-binding globulin levels coupled with reduced total testosterone concentrations, indicative of attenuated hyperandrogenemia (66). In terms of blood glucose regulation, Probiotics can increase the production of SCFA to maintain the integrity of the intestinal barrier or regulate the immune response (67), activate G protein-coupled receptors, and promote the release of peptides such as GLP-1 and PYY to lower blood glucose (68). ...
Article
Full-text available
Polycystic ovary syndrome (PCOS) is a prevalent gynecologic endocrine disorder characterized by menstrual irregularities, elevated androgen levels, and ovulatory dysfunction. Its etiology is multifactorial. Emerging evidence indicates that PCOS patients exhibit diminished gut microbiota (GM) diversity and altered microbial ratios, contributing to the metabolic derangements observed in these individuals. This review elucidates the role of GM in the pathogenesis and metabolic disorders of PCOS, encompassing insulin resistance (IR), hormonal imbalances, bile acid metabolic disorders, Interleukin-22-mediated immune dysregulation, and brain-gut axis disturbances. Additionally, it synthesizes current therapeutic strategies targeting the GM, aiming to furnish a theoretical framework for prospective clinical interventions.
... This was consistent with the previous findings [95]. Therefore, probiotics' positive benefits in PCOS are ascribed to their capacity to regulate IR, inflammation, host metabolism, and reproductive function [96]. Furthermore, probiotics may help address the imbalance in GM communities associated with PCOS and its related symptoms, including HA [97]. ...
Article
Full-text available
Polycystic ovarian syndrome (PCOS) has become a common metabolic syndrome among women globally. In recent times, Indian women are more prone to PCOS following unhealthy eating patterns that involve overconsumption of commercially available Western diets (WD), including sugary food and processed food (PF). Ingestion of WDs and PFs causes dysbiosis (DB) in the gut. Dysbiosis (DB)-an imbalance between beneficial and pathogenic gut bacteria is suspected to undermine the health of the intestine by triggering obesity, insulin resistance (IR), disrupted ovulation (DO), and hyperandrogenism (HA). DO is termed anovulation. Pregnant women who consume commercial foods are prone to intergenerational implications for PCOS risk. Following dietary patterns focusing on GM regulation, such as probiotics and prebiotics, are emerging. Consumption of such a diet can regulate GM and revert the associated metabolic disorders (MD), such as IR, inflammation, and HA, which are the leading factors of PCOS. Emphasizing the choice of traditional, home-cooked food can address nutritional deficiencies and support gut health. The systematic review renders the effect of dietary choices on GM in PCOS regulation.
... These SCFAs help maintain gut health and reduce inflammation. Probiotics also strengthen epithelial barriers and may have direct and indirect pulmonary effects through the gut-lung axis, which can potentially decrease exacerbations and enhance local mucosal defense [52][53][54][55]. ...
Article
Full-text available
Background and Objectives: Cystic fibrosis (CF), caused by CFTR gene mutations, primarily affects the respiratory and gastrointestinal systems. Microbiota modulation through probiotics, prebiotics, or synbiotics may help restore microbial diversity and reduce inflammation. This study aimed to evaluate their efficacy in CF. Materials and Methods: A systematic review and meta-analysis of randomized controlled trials (RCTs) published between 2000 and 2024 was conducted in Cochrane, ScienceDirect, Web of Science, LILAC, BMC, PubMed, and SCOPUS following PRISMA guidelines. Methodological quality was assessed using the Jadad scale, and RevMan 5.4® estimated effects on pulmonary function (FEV1), exacerbations, hospitalizations, quality of life, and inflammatory markers. Results: Thirteen RCTs (n = 552), mostly in pediatric populations, were included. Most examined probiotics (e.g., Lactobacillus rhamnosus GG, L. reuteri), while four used synbiotics. Several studies reported reduced fecal calprotectin and proinflammatory interleukins (e.g., IL-6, IL-8), suggesting an anti-inflammatory effect. However, no significant differences were observed regarding hospitalizations or quality of life. Additionally, none of the studies documented serious adverse events associated with the intervention. The meta-analysis showed no significant decrease in exacerbations (RR = 0.81; 95% CI = 0.48–1.37; p = 0.43) or improvements in FEV1 (MD = 4.7; 95% CI = −5.4 to 14.8; p = 0.37), even in subgroup analyses. Sensitivity analyses did not modify the effect of the intervention on pulmonary function or exacerbation frequency, supporting the robustness of the findings. Conclusions: Current evidence suggests that probiotics or synbiotics yield inconsistent clinical benefits in CF, although some reduction in inflammatory markers may occur. Larger, multicenter RCTs with longer follow-up are needed for clearer conclusions. Until more definitive evidence is available, these supplements should be considered experimental adjuncts rather than standard interventions for CF management.
... Unlike antioxidant supplements, which primarily target oxidative stress, probiotics provide comprehensive immune and metabolic regulation [70]. Hormonal therapies, while effective, may have side effects and do not address the systemic imbalances that probiotics can correct [71,72]. Probiotics thus present a multifaceted, non-pharmacological strategy for improving reproductive health, offering distinct advantages over traditional treatments by addressing root causes through gut microbiota modulation and systemic health enhancement [73,74]. ...
Article
Full-text available
Sexual dysfunction, influenced by hormonal imbalances, psychological factors, and chronic diseases, affects a significant portion of the population. Probiotics, known for their beneficial effects on gut microbiota, have emerged as potential therapeutic agents for improving sexual health. This systematic review evaluates the impact of probiotics on sexual function, hormonal regulation, and reproductive outcomes. A comprehensive search identified 3308 studies, with 12 meeting the inclusion criteria—comprising 10 randomized controlled trials (RCTs) and 2 in vivo and in vitro studies. Probiotic interventions were shown to significantly improve sexual function, particularly in women undergoing antidepressant therapy (p < 0.05). Significant improvements in Female Sexual Function Index (FSFI) scores were observed, with combined treatments such as Lactofem with Letrozole and Lactofem with selective serotonin reuptake inhibitors (SSRIs) demonstrating a 10% biochemical and clinical pregnancy rate compared to 0% in the control group (p = 0.05). Probiotic use was also associated with a 66% reduction in menopausal symptoms, increased sperm motility (36.08%), viability (46.79%), and morphology (36.47%). Probiotics also contributed to favorable hormonal changes, including a reduced luteinizing hormone (LH) to follicle-stimulating hormone (FSH) ratio (from 3.0 to 2.5, p < 0.05) and increased testosterone levels. Regarding reproductive outcomes, probiotic use was associated with higher pregnancy rates in women undergoing fertility treatments and improvements in sperm motility, viability, and morphology in men. This review highlights the promising role of probiotics in addressing sexual dysfunction and reproductive health, suggesting their potential as adjunctive treatments for conditions such as depression and infertility. Further research is needed to better understand the underlying mechanisms of these beneficial effects.
... Polycystic ovary syndrome (PCOS) is a common metabolic disease of endocrine disorder caused by multiple factors in women, with an incidence of 5-10% in women of reproductive age [1][2][3]. Patients often have hyperandrogenemia and ovarian polycystic characteristics such as hirsutism, acne, obesity [4][5][6], as well as clinical manifestations such as oligomenorrhea or amenorrhea, infertility, insulin resistance and type 2 diabetes [7][8][9]. PCOS pathogenesis is complex and multifactorial, involving environmental and genetic factors [10]. Mutations, polymorphisms and differential regulation of genes may be the genetic pathogenesis of PCOS [11]. ...
Article
Full-text available
Purpose The objective of this study was to elucidate the relationship between two single nucleotide polymorphisms (SNPs) rs7176005 and rs6493497 in CYP19 gene and the risk of polycystic ovary syndrome (PCOS) in Northern Chinese women. Methods In this case-control study, a total of 340 women with PCOS and 340 matched healthy controls were recruited. Polymerase chain reaction ligase detection reaction (PCR-LDR) method was used to investigate two SNPs (rs7176005 and rs6493497) in the 5’-flanking region of CYP19 gene exon 1. Results We observed a significant association of rs7176005 and rs6493497 with reduced risk of PCOS. Compared with CC genotype, a significant association of CT genotype (p = 0.019), TT genotype (p < 0.001) and combined CT + TT genotype (p < 0.001) with reduced risk of PCOS was observed. The result of linkage disequilibrium analysis showed that these two SNPs are in complete linkage disequilibrium (r² = 1). For rs7176005 SNP, compared with CC genotype, CT, TT and CT + TT genotypes reduced the risk of PCOS. The age, BMI-adjusted OR were 0.650 (95% CI = 0.460–0.917), 0.158 (95% CI = 0.066–0.376) and 0.545(95% CI = 0.391–0.759), respectively. Conclusions These findings highlight a significant association between CYP19 gene polymorphisms and PCOS susceptibility, implying potential protective effects of T and A alleles. Of course, the major limitation of this study is the sample size of the case-control study. Larger cohort studies are needed to confirm these findings and investigate the underlying causes.
Article
Polycystic Ovary Syndrome (PCOS) is a common endocrine disorder that impacts many women of reproductive age, marked by symptoms like irregular menstrual cycles, infertility, hirsutism, and metabolic issues such as insulin resistance [1-3]. Recent studies have shed light on the impact of gut microbiota—the intricate community of microorganisms living in the gastrointestinal tract—on the pathophysiology of PCOS. Dysbiosis, which refers to an imbalance in gut bacteria, has been connected to the worsening of PCOS symptoms and related metabolic disorders. This connection raises significant questions about the potential effectiveness of microbiota-targeted interventions in managing the syndrome. [4][5]. Research indicates that women with PCOS show a decrease in gut microbiota diversity and changes in bacterial composition when compared to healthy individuals. This alteration may play a role in heightened insulin resistance and chronic inflammation, which are two primary characteristics of the condition.[6][7]. As the ways in which gut microbiota connect with metabolic pathways, hormone regulation, and immune responses become more evident, a framework for comprehending the importance of gut health in PCOS management is being established. Certain gut bacteria are specifically linked to the metabolism of branched-chain amino acids, which can affect insulin sensitivity and androgen levels, thereby adding complexity to the clinical presentation of PCOS [8][9]. Potential controversies in this area of research include doubts about the reliability of findings due to limited diversity in study populations, as most studies have primarily focused on individuals of European descent, which may not reflect the global PCOS population [10]. Additionally, although probiotics and dietary interventions appear promising for enhancing gut health and reducing PCOS symptoms, their effectiveness and mechanisms need further exploration to develop standardized treatment methods[11][12]. Grasping t
Article
Full-text available
Objective Polycystic ovary syndrome (PCOS) is an endocrine metabolic disorder in reproductive-aged women. The study was designed to investigate the metabolic characteristics of different phenotypes in women with PCOS of reproductive age. Methods A total of 442 women with PCOS were recruited in this cross-sectional study. According to different phenotypes, all women were divided into three groups: the chronic ovulatory dysfunction and hyperandrogenism group (OD-HA group, n = 138), the chronic ovulatory dysfunction and polycystic ovarian morphology group (OD-PCOM group, n = 161), and the hyperandrogenism and polycystic ovarian morphology group (HA-PCOM group, n = 143). The metabolic risk factors and prevalence rates of metabolic disorders among the three groups were compared. Results The body mass index (BMI), waist circumference, and waist-to-hip ratio (WHR) of women from the OD-HA group and HA-PCOM group were significantly higher than those of women from the OD-PCOM group (p < 0.05). The serum insulin concentration and homeostasis model assessment of insulin resistance (HOMA IR) at 2 h and 3 h after oral glucose powder in women from the OD-HA group and HA-PCOM group were significantly higher than those from the OD-PCOM group (p < 0.05). The serum total cholesterol (TC), triglyceride (TG), and low-density lipoprotein cholesterol (LDL-C) in women from the OD-HA group and HA-PCOM group were significantly higher than those in women from the OD-PCOM group (p < 0.05). The prevalence rates of impaired glucose tolerance (IGT), type 2 diabetes mellitus (T2DM), insulin resistance (IR), metabolic syndrome (MS), nonalcoholic fatty liver disease (NAFLD), and dyslipidemia of women with PCOS were 17.9%, 3.6%, 58.4%, 29.4%, 46.6%, and 43.4%, respectively. The prevalence rates of IGT, IR, MS, NAFLD, and dyslipidemia of women in the OD-HA group and HA-PCOM group were significantly higher than those of women in the OD-PCOM group (p < 0.05). T concentration (>1.67 nmol/L) and Ferriman–Gallwey (F–G) score (>3) significantly increased the risk of metabolic disorders in women with PCOS (p < 0.05). Conclusion The phenotypes of OD-HA and HA-PCOM in women with PCOS were vulnerable to metabolic disorders compared to OD-PCOM. Thus, the metabolic disorders in women with PCOS especially those with the HA phenotype should be paid more attention in order to reduce long-term complications.
Article
Full-text available
Background Polycystic ovary syndrome (PCOS) is the most common metabolic disorder among women of reproductive age. Many factors are involved in the development of PCOS, among which genetic predisposition is probably the main contributor that is also influenced by lifestyle and environmental factors. This study aims to determine the prevalence of PCOS in different continents based on Rotterdam, AES and NIH diagnostic criteria. Methods We conducted a systematic review and meta-analysis to evaluate the prevalence of polycystic ovary syndrome in women according to (Preferred Reporting Items for Systematic Review and Meta-Analysis) PRISMA guidelines. PubMed, Scopus, Science Direct, Web of Science and Google Scholar databases were comprehensively searched until February 2021 for relevant articles. Heterogeneity between the studies was assessed using the I² index. Begg and Mazumdar’s test was used to evaluate publication bias. Results A total of 35 studies with 12,365,646 subjects were retrieved. The mean age ranged from 10–45 years. Global prevalence of PCOS was 9.2% (95% CI: 6.8–12.5%) based on meta-analysis, our results showed that the global prevalence of PCOS was 5.5% (95% CI: 3.9–7.7%) based on NIH criteria, 11.5 (95% CI: 6.6–19.4) based on Rotterdam criteria, and 7.1% (95% CI: 2.3–20.2%) based on AES criteria. According to self-report subgroup analysis, the prevalence of PCOS was found to be 11% (95% CI: 5.2–21.8%). Conclusion Based on the results of the present study, the prevalence of PCOS in the world was 9.2% (95% CI: 6.8–12.5%). According to the results of the present study and the high prevalence of PCOS, especially in the Africa continent, it is necessary for health systems to implement measures to timely prevent and treat this syndrome.
Article
Full-text available
Background Polycystic ovary syndrome (PCOS) is an endocrinopathy in childbearing-age females which can cause many complications, such as diabetes, obesity, and dyslipidemia. The metabolic disorders in patients with PCOS were linked to gut microbial dysbiosis. However, the correlation between the gut microbial community and dyslipidemia in PCOS remains unillustrated. Our study elucidated the different gut microbiota in patients with PCOS and dyslipidemia (PCOS.D) compared to those with only PCOS and healthy women. Results In total, 18 patients with PCOS, 16 healthy females, and 18 patients with PCOS.D were enrolled. The 16 S rRNA sequencing in V3-V4 region was utilized for identifying the gut microbiota, which analyzes species annotation, community diversity, and community functions. Our results showed that the β diversity of gut microbiota did not differ significantly among the three groups. Regarding gut microbiota dysbiosis, patients with PCOS showed a decreased abundance of Proteobacteria, and patients with PCOS.D showed an increased abundance of Bacteroidota compared to other groups. With respect to the gut microbial imbalance at genus level, the PCOS.D group showed a higher abundance of Clostridium_sensu_stricto_1 compared to other two groups. Furthermore, the abundances of Faecalibacterium and Holdemanella were lower in the PCOS.D than those in the PCOS group. Several genera, including Faecalibacterium and Holdemanella, were negatively correlated with the lipid profiles. Pseudomonas was negatively correlated with luteinizing hormone levels. Using PICRUSt analysis, the gut microbiota community functions suggested that certain metabolic pathways (e.g., amino acids, glycolysis, and lipid) were altered in PCOS.D patients as compared to those in PCOS patients. Conclusions The gut microbiota characterizations in patients with PCOS.D differ from those in patients with PCOS and controls, and those might also be related to clinical parameters. This may have the potential to become an alternative therapy to regulate the clinical lipid levels of patients with PCOS in the future.
Article
Full-text available
In this study, a systematic review of randomized clinical trials conducted from January 2000 to December 2023 was performed to examine the efficacy of psychobiotics—probiotics beneficial to mental health via the gut–brain axis—in adults with psychiatric and cognitive disorders. Out of the 51 studies involving 3353 patients where half received psychobiotics, there was a notably high measurement of effectiveness specifically in the treatment of depression symptoms. Most participants were older and female, with treatments commonly utilizing strains of Lactobacillus and Bifidobacteria over periods ranging from 4 to 24 weeks. Although there was a general agreement on the effectiveness of psychobiotics, the variability in treatment approaches and clinical presentations limits the comparability and generalization of the findings. This underscores the need for more personalized treatment optimization and a deeper investigation into the mechanisms through which psychobiotics act. The research corroborates the therapeutic potential of psychobiotics and represents progress in the management of psychiatric and cognitive disorders.
Article
Full-text available
The prevalence of schizophrenia, affecting approximately 1% of the global population, underscores the urgency for innovative therapeutic strategies. Recent insights into the role of neuroinflammation, the gut–brain axis, and the microbiota in schizophrenia pathogenesis have paved the way for the exploration of psychobiotics as a novel treatment avenue. These interventions, targeting the gut microbiome, offer a promising approach to ameliorating psychiatric symptoms. Furthermore, advancements in artificial intelligence and nanotechnology are set to revolutionize psychobiotic development and application, promising to enhance their production, precision, and effectiveness. This interdisciplinary approach heralds a new era in schizophrenia management, potentially transforming patient outcomes and offering a beacon of hope for those afflicted by this complex disorder.
Article
Full-text available
The consumption of probiotics has been extensively employed for the management or prevention of gastrointestinal disorders by modifying the gut microbiota and changing metabolites. Nevertheless, the probiotic-mediated regulation of host metabolism through the metabolism of bile acids (BAs) remains inadequately comprehended. The gut-liver axis has received more attention in recent years due to its association with BA metabolism. The objective of this research was to examine the changes in BAs and gut microbiota using an in vitro fermentation model. The metabolism and regulation of gut microbiota by commercial probiotics complex containing various species such as Lactobacillus, Bifidobacterium, and Streptococcus were investigated. The findings indicated that the probiotic strains had produced diverse metabolic profiles of BAs. The probiotics mixture demonstrated the greatest capacity for Bile salt hydrolase (BSH) deconjugation and 7α-dehydroxylation, leading to a significant elevation in the concentrations of Chenodeoxycholic acid, Deoxycholic acidcholic acid, and hyocholic acid in humans. In addition, the probiotic mixtures have the potential to regulate the microbiome of the human intestines, resulting in a reduction of isobutyric acid, isovaleric acid, hydrogen sulfide, and ammonia. The probiotics complex intervention group showed a significant increase in the quantities of Lactobacillus and Bifidobacterium strains, in comparison to the control group. Hence, the use of probiotics complex to alter gut bacteria and enhance the conversion of BAs could be a promising approach to mitigate metabolic disorders in individuals.
Article
Full-text available
Postpartum depression (PPD) affects 10-20% of women. Traditional treatments have raised concerns, but omega-3 fatty acids show potential as an alternative. This thematic review, sourced from databases like PubMed and Scopus between 1 February 2023 and 15 March 2023, seeks to delve into the various perspectives on omega-3 supplementation for PPD. The criteria included studies detailing depressive symptoms, social functioning, and neurobiological variables. The review includes research with women showing PPD symptoms, randomized clinical trials, and articles in Spanish, English, and French. Exclusions were studies lacking proper control comparisons and other interventions besides omega-3. Data extraction was performed independently. Two key studies provide contrasting findings on omega-3's impact on PPD symptoms. In the study comparing DHA supplementation to a placebo, significant differences were not found in the EPDS scale, but differences were observed in the BDI scale. In contrast, another study recorded a significant decrease in depression scores in all dose groups, with reductions of 51.5% in the EPDS scale and 48.8% in the HRSD scale. Other studies, encompassing both prenatal and postpartum periods, underscore the differentiation between prenatal depression and PPD. Despite shared diagnostic criteria, PPD presents unique symptoms like restlessness, emotional lability, and baby-related concerns. It is crucial to address biases and obtain specific results, recommending exclusive PPD-focused studies. This review emphasizes the need for continuous exploration of omega-3's relationship with PPD to enhance the life quality of pregnant women and their families.
Article
Full-text available
Study question: What is the recommended assessment and management of those with polycystic ovary syndrome (PCOS), based on the best available evidence, clinical expertise, and consumer preference? Summary answer: International evidence-based guidelines address prioritized questions and outcomes and include 254 recommendations and practice points, to promote consistent, evidence-based care and improve the experience and health outcomes in PCOS. What is known already: The 2018 International PCOS Guideline was independently evaluated as high quality and integrated multidisciplinary and consumer perspectives from six continents; it is now used in 196 countries and is widely cited. It was based on best available, but generally very low to low quality, evidence. It applied robust methodological processes and addressed shared priorities. The guideline transitioned from consensus based to evidence-based diagnostic criteria and enhanced accuracy of diagnosis, whilst promoting consistency of care. However, diagnosis is still delayed, the needs of those with PCOS are not being adequately met, evidence quality was low and evidence-practice gaps persist. Study design, size, duration: The 2023 International Evidence-based Guideline update reengaged the 2018 network across professional societies and consumer organizations with multidisciplinary experts and women with PCOS directly involved at all stages. Extensive evidence synthesis was completed. Appraisal of Guidelines for Research and Evaluation-II (AGREEII)-compliant processes were followed. The Grading of Recommendations, Assessment, Development, and Evaluation (GRADE) framework was applied across evidence quality, feasibility, acceptability, cost, implementation and ultimately recommendation strength and diversity and inclusion were considered throughout. Participants/materials, setting, methods: This summary should be read in conjunction with the full Guideline for detailed participants and methods. Governance included a six-continent international advisory and management committee, five guideline development groups, and paediatric, consumer, and translation committees. Extensive consumer engagement and guideline experts informed the update scope and priorities. Engaged international society-nominated panels included paediatrics, endocrinology, gynaecology, primary care, reproductive endocrinology, obstetrics, psychiatry, psychology, dietetics, exercise physiology, obesity care, public health and other experts, alongside consumers, project management, evidence synthesis, statisticians and translation experts. Thirty-nine professional and consumer organizations covering 71 countries engaged in the process. Twenty meetings and five face-to-face forums over 12 months addressed 58 prioritized clinical questions involving 52 systematic and 3 narrative reviews. Evidence-based recommendations were developed and approved via consensus across five guideline panels, modified based on international feedback and peer review, independently reviewed for methodological rigour, and approved by the Australian Government National Health and Medical Research Council (NHMRC). Main results and the role of chance: The evidence in the assessment and management of PCOS has generally improved in the past five years, but remains of low to moderate quality. The technical evidence report and analyses (∼6000 pages) underpins 77 evidence-based and 54 consensus recommendations, with 123 practice points. Key updates include: i) further refinement of individual diagnostic criteria, a simplified diagnostic algorithm and inclusion of anti-Müllerian hormone (AMH) levels as an alternative to ultrasound in adults only; ii) strengthening recognition of broader features of PCOS including metabolic risk factors, cardiovascular disease, sleep apnea, very high prevalence of psychological features, and high risk status for adverse outcomes during pregnancy; iii) emphasizing the poorly recognized, diverse burden of disease and the need for greater healthcare professional education, evidence-based patient information, improved models of care and shared decision making to improve patient experience, alongside greater research; iv) maintained emphasis on healthy lifestyle, emotional wellbeing and quality of life, with awareness and consideration of weight stigma; and v) emphasizing evidence-based medical therapy and cheaper and safer fertility management. Limitations, reasons for caution: Overall, recommendations are strengthened and evidence is improved, but remain generally low to moderate quality. Significantly greater research is now needed in this neglected, yet common condition. Regional health system variation was considered and acknowledged, with a further process for guideline and translation resource adaptation provided. Wider implications of the findings: The 2023 International Guideline for the Assessment and Management of PCOS provides clinicians and patients with clear advice on best practice, based on the best available evidence, expert multidisciplinary input and consumer preferences. Research recommendations have been generated and a comprehensive multifaceted dissemination and translation programme supports the Guideline with an integrated evaluation program. Study funding/competing interest(s): This effort was primarily funded by the Australian Government via the National Health Medical Research Council (NHMRC) (APP1171592), supported by a partnership with American Society for Reproductive Medicine, Endocrine Society, European Society for Human Reproduction and Embryology, and the European Society for Endocrinology. The Commonwealth Government of Australia also supported Guideline translation through the Medical Research Future Fund (MRFCRI000266). HJT and AM are funded by NHMRC fellowships. JT is funded by a Royal Australasian College of Physicians (RACP) fellowship. Guideline development group members were volunteers. Travel expenses were covered by the sponsoring organizations. Disclosures of interest were strictly managed according to NHMRC policy and are available with the full guideline, technical evidence report, peer review and responses (www.monash.edu/medicine/mchri/pcos). Of named authors HJT, CTT, AD, LM, LR, JBoyle, AM have no conflicts of interest to declare. JL declares grant from Ferring and Merck; consulting fees from Ferring and Titus Health Care; speaker's fees from Ferring; unpaid consultancy for Ferring, Roche Diagnostics and Ansh Labs; and sits on advisory boards for Ferring, Roche Diagnostics, Ansh Labs, and Gedeon Richter. TP declares a grant from Roche; consulting fees from Gedeon Richter and Organon; speaker's fees from Gedeon Richter and Exeltis; travel support from Gedeon Richter and Exeltis; unpaid consultancy for Roche Diagnostics; and sits on advisory boards for Roche Diagnostics. MC declares travels support from Merck; and sits on an advisory board for Merck. JBoivin declares grants from Merck Serono Ltd.; consulting fees from Ferring B.V; speaker's fees from Ferring Arzneimittell GmbH; travel support from Organon; and sits on an advisory board for the Office of Health Economics. RJN has received speaker's fees from Merck and sits on an advisory board for Ferring. AJoham has received speaker's fees from Novo Nordisk and Boehringer Ingelheim. The guideline was peer reviewed by special interest groups across our 39 partner and collaborating organizations, was independently methodologically assessed against AGREEII criteria and was approved by all members of the guideline development groups and by the NHMRC.
Article
Full-text available
Background The results of human observational studies on the correlation between gut microbiota perturbations and polycystic ovary syndrome (PCOS) have been contradictory. This study aimed to perform the first systematic review and meta-analysis to evaluate the specificity of the gut microbiota in PCOS patients compared to healthy women. Methods Literature through May 22, 2023, was searched on PubMed, Web of Science, Medline, Embase, Cochrane Library, and Wiley Online Library databases. Unreported data in diversity indices were filled by downloading and processing raw sequencing data. Systematic review inclusion: original studies were eligible if they applied an observational case-control design, performed gut microbiota analysis and reported diversity or abundance measures, sampled general pre-menopausal women with PCOS, and are longitudinal studies with baseline comparison between PCOS patients and healthy females. Systematic review exclusion: studies that conducted interventional or longitudinal comparisons in the absence of a control group. Two researchers made abstract, full-text, and data extraction decisions, independently. The Joanna Briggs Institute Critical Appraisal Checklist was used to assess the methodologic quality. Hedge’s g standardized mean difference (SMD), confidence intervals (CIs), and heterogeneity (I²) for alpha diversity were calculated. Qualitative syntheses of beta-diversity and microbe alterations were performed. Results Twenty-eight studies (n = 1022 patients, n = 928 control) that investigated gut microbiota by collecting stool samples were included, with 26 and 27 studies having provided alpha-diversity and beta-diversity results respectively. A significant decrease in microbial evenness and phylogenetic diversity was observed in PCOS patients when compared with control participants (Shannon index: SMD = − 0.27; 95% CI, − 0.37 to − 0.16; phylogenetic diversity: SMD = − 0.39; 95% CI, -− 0.74 to − 0.03). We also found that reported beta-diversity was inconsistent between studies. Despite heterogeneity in bacterial relative abundance, we observed depletion of Lachnospira and Prevotella and enrichment of Bacteroides, Parabacteroides, Lactobacillus, Fusobacterium, and Escherichia/Shigella in PCOS. Gut dysbiosis in PCOS, which might be characterized by the reduction of short-chain fatty acid (SCFA)-producing and bile-acid-metabolizing bacteria, suggests a shift in balance to favor pro-inflammatory rather than anti-inflammatory bacteria. Conclusions Gut dysbiosis in PCOS is associated with decreased diversity and alterations in bacteria involved in microbiota-host crosstalk. Trial registration PROSPERO registration: CRD42021285206, May 22, 2023.
Article
Polycystic ovarian syndrome (PCOS) is a complex endocrine disorder characterized by hormonal dysregulation, metabolic disturbances, and reproductive abnormalities. Probiotics are the gut bacteria which helps in digestion and possess several functionalities positively in body like immunomodulation, hormonal balancing, antihypertensive etc. There are evidences pointing for preventive as well as therapeutic results from the PCOS symptoms by administrating probiotics to the adolescent women. Some triggers causing implications of gut microbiota alterations in PCOS, including modulation of host metabolism, inflammation, insulin resistance, and reproductive function. Present paper reviews the mechanism through which these outcomes are achieved.