Skip to main content

REVIEW article

Front. Cell. Infect. Microbiol., 10 November 2022
Sec. Microbiome in Health and Disease
Volume 12 - 2022 | https://doi.org/10.3389/fcimb.2022.1038928

Probiotics for constipation in Parkinson’s: A systematic review and meta-analysis of randomized controlled trials

  • 1Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
  • 2Traditional Chinese Medicine Department, Zigong First People’s Hospital, Zigong, China

Background: Parkinson’s disease (PD)-related constipation may affects both disease occurrence and disease progression. Probiotics, as a potential therapeutic intervention, have attracted the attention of researchers, but the evidence of their efficacy and safety has not been systematically reviewed.

Aim: A systematic review and meta-analysis of randomized controlled trials of probiotics in the treatment of PD constipation was conducted to determine the efficacy and safety of probiotics in the treatment of PD constipation.

Methods: Four databases (The Cochrane Central Register of Controlled Trials, Embase, PubMed, and Web of Science) were searched from their establishment to June 1, 2022. We included randomized controlled trials of probiotics for the treatment of constipation in patients with PD, with probiotics in the experimental group and a placebo, another treatment, or no treatment in the control group. The primary outcome was the number of bowel movements per week. Secondary outcomes included nonmotor symptoms (NMS), gut transit time (GTT), abdominal pain, abdominal distention, constipation, and quality of life scores. Stata15.1 was used to generate a summary of the data and perform a descriptive analysis if necessary. The GRADE tool was used to assess the quality of the evidence and the Cochrane guidelines to assess the risk of bias for each study.

Results: Finally, four qualified RCTs were included, comprising 287 participants. Compared with the control group, probiotics could effectively increase the frequency of defecation per week in PD patients (WMD = 1.02. 95%CI: 0.56–1.48, and P < 0.00001), but the heterogeneity was high, and the quality of the evidence was low. There was no significant difference in average stool consistency between patients with PD treated with probiotics and those given a placebo in (WMD = –0.08. 95%CI: –1.42–1.26, and P = 0.908). In addition, the results suggested that probiotics have no obvious effect on additional indicators of gastrointestinal dysfunction, such as GTT, abdominal pain, and abdominal distension, and there is insufficient evidence on their ability to improve NMS and Parkinson’s disease Questionnaire 39 summary indices (PDQ39-SI). Safety issues should be carefully explained.

Conclusion: There is insufficient evidence supporting the use of probiotics to treat constipation in patients with PD. Taking all the results together, probiotics have potential value in the treatment of PD-related constipation.

Systematic Review Registration: PROSPERO CRD42022331325.

Introduction

Parkinson’s disease (PD) is the second most common neurodegenerative disorder after Alzheimer’s disease, and dyskinesia is a major feature of PD. In fact, a range of nonmotor symptoms (NMS) associated with autonomic nervous dysfunction, especially dysfunction of the gastrointestinal tract (Borek et al., 2006; Cloud and Greene, 2011; Fasano et al., 2015), may occur at all stages of PD (Chaudhuri and Schapira, 2009) and may even be closely related to the pathogenesis of PD (Braak et al., 2003; Holmqvist et al., 2014). NMS has a significant negative impact on clinical care and health-related quality of life (Hr-QoL) in patients with PD (Li et al., 2010; Lyons and Pahwa, 2011).

Constipation is one of the most common NMS in PD patients with autonomic system and gastrointestinal disorders (Sakakibara et al., 2003; Verbaan et al., 2007; Gao et al., 2011), even before the onset of motor symptoms (Abbott et al., 2001; Savica et al., 2009). It affects about 50%–80% of PD patients (Ashraf et al., 1997; Verbaan et al., 2007). The evidence shows that constipation is related to the duration and severity of PD (Krogh et al., 2008), and the frequency and severity of constipation are accelerated by the progression of PD (Edwards et al., 1993). Clearly, constipation and PD have reciprocal effects (Fu et al., 2022).

PD-related constipation is an active research field. Various studies have evaluated different drugs for the treatment of PD-related constipation, but there is no clear guideline recommendation so far (Poirier et al., 2016). Clearly, it is still necessary to explore effective and safe emerging drugs (Pohl et al., 2008). Previous studies have shown that probiotics can significantly improve the stool consistency and bowel habits of PD patients (Cassani et al., 2011); increase the number of complete bowel movements per week (CBM) and gut transit time (GTT) (Barichella et al., 2016; Ibrahim et al., 2020; Tan et al., 2021); and reduce abdominal pain, abdominal distension, and incomplete emptying in PD patients (Perez-Lloret et al., 2013; Georgescu et al., 2016).

However, evidence for the positive effects of probiotics on PD constipation is inconclusive. Therefore, this systematic review and meta-analysis included data from the results of several clinical trials evaluating the efficacy and safety of probiotics in the treatment of PD-related constipation. We aim to provide a comprehensive update of the clinical data for evidence-based guideline development.

Methods

Eligibility criteria

This study included all randomized controlled trials of probiotics in the treatment of PD-related constipation. PD participants meet internationally recognized diagnostic criteria and had constipation or gastrointestinal dysfunction. The experimental group was treated with probiotics, while the control group was treated with a placebo, other treatments, or no intervention. The main outcome was the number of bowel movements per week; secondary outcomes included average stool consistency, NMS, GTT, abdominal pain, abdominal distension, constipation, and quality of life scores.

Reviews, conference papers, comments, animal studies, retrospective studies, case-control studies, and self-controlled studies were excluded. RCTs that did not include constipation-related outcomes were also excluded.

Search strategy

RCTs were searched in The Cochrane Central Register of Controlled Trials, Embase, PubMed, and Web of Science from inception to June 1, 2022. In addition, a list of references included in the study was manually searched to identify relevant trials. There were no restrictions on language, year of publication, etc. Grey literature and data on the research registry platform were not within the scope of the search because we do not have access to these. Detailed search strategies are available in the Supplementary Materials.

Study selection

According to the strict retrieval strategy, reviewers used Endnote X9 and manual procedures to delete duplicate documents. Initial study selection was performed according to the title and abstract, followed by full-text reading to determine the final included studies. Two reviewers completed the literature search and screening independently. Any disagreement between the two reviewers was resolved by discussion. If no agreement was reached, the final decision was made by a third reviewer.

Data extraction

Data extraction was performed independently and cross-checked by two examiners according to a standardized form developed in advance. The main contents included the publication year, first author, country, study design, participants (age, sex), sample size, intervention details (formulation, dose, duration), results, etc. Any disagreement between the two reviewers was resolved by discussion. If no agreement was reached, the final decision was made by a third reviewer.

Data synthesis and statistical analysis

A meta-analysis was conducted on the same outcome indicators in two or more RCTs. Continuous variables were represented by the weighted mean difference (WMD) and 95% confidence interval (CI), a result was considered statistically significant at P < 0.05, and if I2 ≥ 50%, a random effect model was used, and sensitivity analysis was conducted to observe the stability of the results. Due to the small number of RCTs included in this study, we did not test for publication bias. For individual outcome measures, data were summarized, and descriptive analysis was performed.

Assessment of the risk of bias and quality of evidence

A risk of bias assessment was conducted for each RCT according to the Cochrane Handbook. The evaluation areas included random sequence generation, allocation concealment, blinding of participants and personnel, blinding of outcome assessment, incomplete outcome data, selective reporting, and other sources of bias. Each area was rated as a high, low, or unclear risk. Evaluation of the quality of evidence against Grading of Recommendations Assessment, Development, and Evaluations (GRADE) consists of five main factors: risk of bias, inconsistency, imprecision, indirectness, and publication bias. All evaluations were conducted independently by two reviewers, with unresolved differences determined by a third reviewer.

Results

Results of literature search and selection

We retrieved 53 related articles from 4 databases and removed 27 duplicates. Then, four qualified studies were included through title, abstract, and full text evaluation. The detailed flow chart is shown in Figure 1. The excluded list in the “full-text assessed for eligibility” phase are outlined in the Supplementary Materials.

FIGURE 1
www.frontiersin.org

Figure 1 Flow chart of study selection.

Characteristics of included studies

Four RCTs with 287 participants were included in the study, which was conducted in Italy, Romania, and Malaysia between 2016 and 2021. Participants were over 60 years old on average, there were more men than women, and the duration of treatment ranged from four weeks to three months. Except for one study in which trimebutine was used in the control group, all the participants in control groups took a placebo with the same characteristics as the treatment given to the intervention group but without probiotics. Three RCTs showed adverse reactions, mainly manifested as abdominal pain, abdominal distension, and dizziness, and the experimental group was larger than the control group. Detailed literature features are shown in Table 1.

TABLE 1
www.frontiersin.org

Table 1 Detailed literature features.

At the same time, we summarized the baseline information of severity of PD symptoms of participations and PD drugs used in the three RCTs included in the meta-analysis. Ibrahim et al. (2020) included idiopathic PD patients in Hoehn and Yahr stages 1–4, The proportion of participations with stage 3 and below in the experimental group and the control group was 59.3%/64.3%, and the proportion of participations with levodopa in the two groups was 92.6%/89.3% and dopamin agonist were 63%/57.1%; Barichella et al. (2016) also included idiopathic PD patients in Hoehn and Yahr stages 1–4, the proportion of participations with stage 3 and below in the experimental group and the control group was 76.3%/75%, and the proportion of participations who received dopamine-agonist therapy in the two groups was 63.8%/62.5%, and the daily dose of levodopa in the two groups was 691mg ± 315mg/624mg ± 289mg. Tan et al. (2021) used Movement Disorder Society Unified Parkinson’s Disease Rating Scale to evaluate the severity of participations, the score of experimental group and control group was 27.9 ± 12.8/27.5 ± 12.6. The comparison of the proportion of participations taking drugs in the two groups was as follows: levodopa (97.1%/97.4%), caudate agonist (38.2%/39.5%), and anticholinergics (17.6%/13.2%). There was no significant difference in the severity of PD symptoms of participations and PD drugs used between the experimental group and the control group in the three RCTs.

Risk of bias

We assessed the risk of bias for four RCTs using the Cochrane Collaboration Handbook. The study by Georgescu et al. (2016) did not mention the allocation of hidden schemes, and the blinding was not sufficiently informative, so its risk of bias was rated as unclear. The risk of bias of all other studies was rated as low (Figure 2).

FIGURE 2
www.frontiersin.org

Figure 2 The risk of bias of studies.

Results of the meta-analysis

The number of bowel movements per week was reported in three RCTs (Barichella et al., 2016; Ibrahim et al., 2020; Tan et al., 2021). A Meta-analysis suggested that probiotics could effectively increase the number of bowel movements per week in PD patients compared with the control group (WMD = 1.02, 95%CI: 0.56–1.48, and P < 0.00001), but the heterogeneity was high (I2 = 71.5%, P = 0.030), as shown in Figure 3. Two RCTs (Barichella et al., 2016; Tan et al., 2021) calculated the changes in the average stool consistency in PD patients, but the results were inconsistent, and meta-analysis results showed no statistically significant difference between the probiotic group and the control group (WMD = –0.08, 95%CI: –1.42 to 1.26, and P = 0.908), with high heterogeneity (I2 = 93.6%, P = 0.000). Detailed results are shown in Figure 4.

FIGURE 3
www.frontiersin.org

Figure 3 Meta-analysis of the number of bowel movements per week.

FIGURE 4
www.frontiersin.org

Figure 4 Meta-analysis of average stool consistency.

Summary of the outcomes

Georgescu et al. (2016) assessed gastrointestinal function (GI) in the NMS of PD patients. The results showed that probiotics showed the potential to relieve abdominal pain and abdominal distention in PD patients but had no significant effect on relieving constipation symptoms in PD patients. Overall, there was no statistically significant difference between the probiotic group and the trimebutine group. The results of a study by Ibrahim et al. (2020) showed that probiotics can shorten GTT and reduce the Non-motor Symptom Scale (NMSS) score in PD patients. There was a potential improvement in the scores on Parkinson’s disease Questionnaire 39 Summary Indices (PDQ39-SI), but there was no significant difference compared with the placebo group. Tan et al. (2021) showed that probiotics could significantly relieve the degree of constipation in PD patients. More detailed results are shown in Table 2.

TABLE 2
www.frontiersin.org

Table 2 Summary of the outcomes.

Adverse reactions

A total of 7 adverse reactions were noted in the three RCTs, 6 of which occurred in the probiotic group, mainly abdominal pain, abdominal bloating, dizziness, and lethargy. Among them, Ibrahim et al. noted that side effects such as abdominal bloating and dizziness were transient, and symptoms resolved when probiotics were discontinued. No serious adverse reactions related to probiotic treatment were observed.

Sensitivity analysis and publication bias

We performed sensitivity analysis on the meta-analysis results of the number of bowel movements per week, and the results were stable (Supplementary Materials). Due to the small number of RCTs included in this study, we did not conduct a publication bias assessment.

Grade

Due to the high heterogeneity and small sample size in the meta-analysis of the number of bowel movements per week and average stool consistency in Parkinson’s patients, the evidence level of the results was rated as low, as shown in Table 3.

TABLE 3
www.frontiersin.org

Table 3 The result of the GRADE.

Discussion

A total of four RCTs evaluating probiotics for PD constipation were included in this systematic review. A meta-analysis showed that probiotics increased the number of bowel movements per week in PD patients but had no effect on average stool consistency. Although the sensitivity analysis showed that the results were stable, subgroup analysis could not be carried out due to the small number of RCTs included, and the source of heterogeneity could not be found. Different diagnostic criteria, different doses and types of probiotics taken, and the small number of included literatures may be the reasons for the high heterogeneity. High heterogeneity in the results and the small sample size of the meta-analysis resulted in a low quality of evidence. In addition, receiving probiotics or a placebo showed no significant difference in terms of alleviating abdominal pain, abdominal distention, GTT, and other gastrointestinal disorders in PD patients.

The gut-brain axis refers to the dynamic bidirectional interaction between the intestinal flora and the central nervous system. The interaction between the central nervous system and the gut mainly connects peripheral intestinal function to the emotional and cognitive brain centers through various neuro-immune-endocrine mediators (Naomi et al., 2021). An imbalance in the intestinal flora affects the occurrence and progression of neurodegenerative diseases and mental disorders, while supplementation with dietary fiber and probiotics can improve various cognitive functions (Barbosa and Vieira-Coelho, 2020; Barrio et al., 2022). Despite the popularity of probiotics as a treatment for neurodegenerative diseases in recent years, the results of studies on probiotics have been inconsistent.

The FAO/WHO defines probiotics as “living microorganisms beneficial to the health of the host when ingested in an appropriate amount” (Hill et al., 2014). A study summarized the evidence of the relationship between the intestinal microflora, cognitive function, and dementia pathology in the elderly, and its conclusion supported the impact of intestinal microorganisms on cognitive function. In animal studies, prebiotics and probiotics had a positive effect on cognitive function (Neta et al., 2022; de Rijke et al., 2022), but the existing evidence is insufficient to support a clinical application (Ticinesi et al., 2018).

Gastrointestinal tract is closely related to the central nervous system, environmental pathogens may enter the central nervous system through the vagal connections in the gut, and eventually accelerate the progression of PD (Travagli et al., 2020). Constipation is a prevalen non-motor symptom in PD, its underlying mechanism and pathophysiology is complex, such as accumulation of alpha-synuclein originate from the myenteric plexus in the intestine may be one of the reasons (Fasano et al., 2015; Barrenschee et al., 2017). At the same time, the use of anti-parkinsonism drugs can also result in slow colonic transport or puborectalis dyssynergia and aggravate constipation symptoms (Stocchi and Torti, 2017). Moreover, PD patients are associated with lower short chain fatty acids (SCFAs), which have anti-inflammatory properties and are essential for gut mucosal lining repair, regulation of intestinal nervous system activity, and enhancement of gut motility (Unger et al., 2016; Aho et al., 2021). The mechanism by which probiotics improve PD constipation may be through the increase of SCFAs and mucin production in the gut thereby repairing the gut mucosal lining and enhancing gut motility (Dimidi et al., 2017; Suez et al., 2019). Whether and to what extent probiotics, while relieving constipation, also slow the progression of PD, remains to be investigated.

The authors of several systematic reviews and meta-analyses evaluating the use of probiotics for Alzheimer’s disease (AD), mild cognitive impairment (MCI), and PD believe that probiotics and synbiotics supplements improve cognitive function in patients with AD, while no positive effect was seen in other biomarkers of oxidative stress or lipid profiles. Only insulin resistance could be improved in patients with AD (Krüger et al., 2021; Li et al., 2021), and dietary probiotics could improve cognitive function in MCI patients, but in another study, the effect on AD patients was limited (Zhu et al., 2021). However, studies by Leta et al. (2021) highlighted that probiotic therapy can increase glucose metabolism, reduce peripheral and central inflammatory responses (e.g., reduction of interleukin-6 (IL-6), hs-CRP, and tumor necrosis factor -α (TNF-α) in PD patients, and increase motor and non-motor function. The results of a meta-analysis by Xiang et al. (2022) suggest that probiotics can enhance the cognitive function of AD and MCI patients and improve the gastrointestinal symptoms of PD patients, for example, by relieving abdominal pain, abdominal distention, and constipation and increasing the number of bowel movements per week, with no significant effect on stool consistency. In addition, probiotics can also reduce biomarkers of inflammation and oxidative stress. The results of gastrointestinal symptoms were similar to those of this study.

Based on the gut-brain axis connection, patients with neurological diseases have a much higher risk of intestinal dysfunction. How to effectively manage intestinal disorders has always been a focus of the medical field, while intestinal management in the past was empirical with very little research basis (Coggrave et al., 2006). In the updated Cochrane Systematic Review, interventions to address constipation remain limited, and the quality of the evidence is very low due to differences in intervention and control approaches. At present, common methods to improve constipation mainly include catharsis, abdominal massage, electrical stimulation, an anticholinesterase anticholinergic drug combination (neostigmine glycopyrrolate), anal flushing, oral carbonated water, and lifestyle modification (Coggrave et al., 2014). Probiotic therapy has been well documented in patients with simple functional constipation, with multistrain probiotics significantly reducing GTT, increasing stool removal frequency, and improving stool consistency. Therefore, probiotics are considered safe and natural remedies for the relief of functional constipation in adults (Zhang et al., 2020).

However, The International Parkinson and Movement Disorder Society (MDS) Evidence‐Based Medicine (EBM) Committee only recommended Macrogol, Lubiprostone, and Probiotics/Prebiotic fibers as three medicines/foods used to treat PD-related constipation (Hatano et al., 2022). Chronic constipation is the earliest symptom of PD prodrome and one of the universal NMS in PD (Kalia and Lang, 2015). This systematic review focused on the evaluation of probiotics for the treatment of constipation in PD patients. Probiotics increased the number of weekly defecations in PD patients compared with a placebo, but with high heterogeneity and a low quality of evidence. Our results also suggest that probiotics have no significant beneficial effect on stool consistency, GTT, NMSS, and PDQ39-SI, and there is no clear evidence that probiotics have a significant effect on additional symptoms of gastrointestinal dysfunction, such as abdominal pain and bloating. In terms of safety, clinical studies have reported adverse reactions such as abdominal pain and abdominal distention in the probiotic group. In fact, gastrointestinal dysfunction in PD patients, as one of the common NMS, may include clinical symptoms such as abdominal pain and abdominal distension. Whether adverse reactions are caused by drugs requires careful consideration. In addition, the included studies also reported two adverse reactions of lethargy and dizziness in the probiotics group. Although the authors indicated that the symptoms disappeared after the cessation of probiotics and no serious adverse reactions occurred, the safety of probiotics still needs to be verified in subsequent studies. Meanwhile, clinical studies are limited, the overall sample size is small, and whether probiotics synthesized by different strains have different effects on intestinal function still needs further research. A large sample size and high-quality clinical evidence are still the top priority to clarify the efficacy and safety of probiotics in the treatment of PD-related constipation.

Strengths and limitations

The problem of constipation in PD patients is closely related to the progression of their own disease. With the gradual emergence of probiotics, it is clearly important to determine the effectiveness and safety of probiotics on PD-related constipation for clinical selection. Here, we must point out that the systematic review has some limitations. First, the number of clinical studies was limited. We only included four RCTs involving 287 participants and only conducted a meta-analysis on the two main results, the number of bowel movements per week and thin stool consistency, which had high heterogeneity. Secondly, due to the small number of RCTs included in this study, the composition, dosage, and frequency of probiotics were different, so we did not conduct publication bias assessment and subgroup analysis. In addition, the sensitivity analysis of the number of bowel movements per week was stable, but the results should be interpreted carefully. However, the advantages of our study are that (1) This is the first meta-analysis and systematic review of existing evidence to clarify the efficacy and safety of probiotics for constipation in PD patients, which will provide favorable evidence for evidence-based medicine. (2) The reviewers discussed the limitations of the included studies and proposed specific suggestions for future studies to provide reliable research results.

Conclusion

Although the evidence in this systematic review only supports the notion that probiotics have a significant effect on increasing the number of bowel movements per week in patients with PD constipation, probiotics have potential value in the treatment of PD-related constipation based on the overall results of existing clinical observational studies, animal research reviews, and clinical experience.

Author contributions

FZ and SY: study conception and design, data analysis, interpretation, and manuscript writing. All authors: final approval of manuscript.

Acknowledgments

Thanks to LYL, DYC and ZL for their contribution to the data collection and assembly of this manuscript.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Supplementary material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fcimb.2022.1038928/full#supplementary-material

References

Abbott, R. D., Petrovitch, H., White, L. R., Masaki, K. H., Tanner, C. M., Curb, J. D., et al. (2001). Frequency of bowel movements and the future risk of parkinson's disease. Neurology 57 (3), 456–462. doi: 10.1212/wnl.57.3.456

PubMed Abstract | CrossRef Full Text | Google Scholar

Aho, V. T. E., Houser, M. C., Pereira, P. A. B., Chang, J., Rudi, K., Paulin, L., et al. (2021). Relationships of gut microbiota, short-chain fatty acids, inflammation, and the gut barrier in parkinson's disease. Mol. Neurodegener. 16 (1), 6. doi: 10.1186/s13024-021-00427-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Ashraf, W., Pfeiffer, R. F., Park, F., Lof, J., Quigley, E. M. (1997). Constipation in parkinson's disease: objective assessment and response to psyllium. Mov Disord. 12 (6), 946–951. doi: 10.1002/mds.870120617

PubMed Abstract | CrossRef Full Text | Google Scholar

Barbosa, R. S. D., Vieira-Coelho, M. A. (2020). Probiotics and prebiotics: focus on psychiatric disorders - a systematic review. Nutr. Rev. 78 (6), 437–450. doi: 10.1093/nutrit/nuz080

PubMed Abstract | CrossRef Full Text | Google Scholar

Barichella, M., Pacchetti, C., Bolliri, C., Cassani, E., Iorio, L., Pusani, C., et al. (2016). Probiotics and prebiotic fiber for constipation associated with Parkinson disease: An RCT. Neurology 87 (12), 1274–1280. doi: 10.1212/wnl.0000000000003127

PubMed Abstract | CrossRef Full Text | Google Scholar

Barrenschee, M., Zorenkov, D., Böttner, M., Lange, C., Cossais, F., Scharf, A. B., et al. (2017). Distinct pattern of enteric phospho-alpha-synuclein aggregates and gene expression profiles in patients with parkinson's disease. Acta Neuropathol. Commun. 5 (1), 1. doi: 10.1186/s40478-016-0408-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Barrio, C., Arias-Sánchez, S., Martín-Monzón, I. (2022). The gut microbiota-brain axis, psychobiotics and its influence on brain and behaviour: A systematic review. Psychoneuroendocrinology 137, 105640. doi: 10.1016/j.psyneuen.2021.105640

PubMed Abstract | CrossRef Full Text | Google Scholar

Borek, L. L., Amick, M. M., Friedman, J. H. (2006). Non-motor aspects of parkinson's disease. CNS Spectr. 11 (7), 541–554. doi: 10.1017/s1092852900013560

PubMed Abstract | CrossRef Full Text | Google Scholar

Braak, H., Rüb, U., Gai, W. P., Del Tredici, K. (2003). Idiopathic parkinson's disease: possible routes by which vulnerable neuronal types may be subject to neuroinvasion by an unknown pathogen. J. Neural Transm (Vienna) 110 (5), 517–536. doi: 10.1007/s00702-002-0808-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Cassani, E., Privitera, G., Pezzoli, G., Pusani, C., Madio, C., Iorio, L., et al. (2011). Use of probiotics for the treatment of constipation in parkinson's disease patients. Minerva Gastroenterol. Dietol 57 (2), 117–121.

PubMed Abstract | Google Scholar

Chaudhuri, K. R., Schapira, A. H. (2009). Non-motor symptoms of parkinson's disease: dopaminergic pathophysiology and treatment. Lancet Neurol. 8 (5), 464–474. doi: 10.1016/s1474-4422(09)70068-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Cloud, L. J., Greene, J. G. (2011). Gastrointestinal features of parkinson's disease. Curr. Neurol. Neurosci. Rep. 11 (4), 379–384. doi: 10.1007/s11910-011-0204-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Coggrave, M., Norton, C., Cody, J. D. (2014). Management of faecal incontinence and constipation in adults with central neurological diseases. Cochrane Database Syst. Rev. 1), Cd002115. doi: 10.1002/14651858.CD002115.pub5

CrossRef Full Text | Google Scholar

Coggrave, M., Wiesel, P. H., Norton, C. (2006). Management of faecal incontinence and constipation in adults with central neurological diseases. Cochrane Database Syst. Rev. 2), Cd002115. doi: 10.1002/14651858.CD002115.pub3

CrossRef Full Text | Google Scholar

de Rijke, T. J., Doting, M. H. E., van Hemert, S., De Deyn, P. P., van Munster, B. C., Harmsen, H. J. M., et al. (2022). A systematic review on the effects of different types of probiotics in animal alzheimer's disease studies. Front. Psychiatry 13. doi: 10.3389/fpsyt.2022.879491

CrossRef Full Text | Google Scholar

Dimidi, E., Christodoulides, S., Scott, S. M., Whelan, K. (2017). Mechanisms of action of probiotics and the gastrointestinal microbiota on gut motility and constipation. Adv. Nutr. 8 (3), 484–494. doi: 10.3945/an.116.014407

PubMed Abstract | CrossRef Full Text | Google Scholar

Edwards, L., Quigley, E. M., Hofman, R., Pfeiffer, R. F. (1993). Gastrointestinal symptoms in Parkinson disease: 18-month follow-up study. Mov Disord. 8 (1), 83–86. doi: 10.1002/mds.870080115

PubMed Abstract | CrossRef Full Text | Google Scholar

Fasano, A., Visanji, N. P., Liu, L. W., Lang, A. E., Pfeiffer, R. F. (2015). Gastrointestinal dysfunction in parkinson's disease. Lancet Neurol. 14 (6), 625–639. doi: 10.1016/s1474-4422(15)00007-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Fu, S. C., Shih, L. C., Wu, P. H., Hsieh, Y. C., Lee, C. H., Lin, S. H., et al. (2022). Exploring the causal effect of constipation on parkinson's disease through mediation analysis of microbial data. Front. Cell Infect. Microbiol. 12. doi: 10.3389/fcimb.2022.871710

CrossRef Full Text | Google Scholar

Gao, X., Chen, H., Schwarzschild, M. A., Ascherio, A. (2011). A prospective study of bowel movement frequency and risk of parkinson's disease. Am. J. Epidemiol. 174 (5), 546–551. doi: 10.1093/aje/kwr119

PubMed Abstract | CrossRef Full Text | Google Scholar

Georgescu, D., Ancusa, O. E., Georgescu, L. A., Ionita, I., Reisz, D. (2016). Nonmotor gastrointestinal disorders in older patients with parkinson's disease: Is there hope? Clin. Interv Aging 11, 1601–1608. doi: 10.2147/cia.S106284

PubMed Abstract | CrossRef Full Text | Google Scholar

Hatano, T., Oyama, G., Shimo, Y., Ogaki, K., Nishikawa, N., Fukae, J., et al. (2022). Investigating the efficacy and safety of elobixibat, an ileal bile acid transporter inhibitor, in patients with parkinson's disease with chronic constipation: a multicentre, placebo-controlled, randomised, double-blind, parallel-group stud (CONST-PD). BMJ Open 12 (2), e054129. doi: 10.1136/bmjopen-2021-054129

PubMed Abstract | CrossRef Full Text | Google Scholar

Hill, C., Guarner, F., Reid, G., Gibson, G. R., Merenstein, D. J., Pot, B., et al. (2014). Expert consensus document. the international scientific association for probiotics and prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat. Rev. Gastroenterol. Hepatol. 11 (8), 506–514. doi: 10.1038/nrgastro.2014.66

PubMed Abstract | CrossRef Full Text | Google Scholar

Holmqvist, S., Chutna, O., Bousset, L., Aldrin-Kirk, P., Li, W., Björklund, T., et al. (2014). Direct evidence of Parkinson pathology spread from the gastrointestinal tract to the brain in rats. Acta Neuropathol. 128 (6), 805–820. doi: 10.1007/s00401-014-1343-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Ibrahim, A., Ali, R. A. R., Manaf, M. R. A., Ahmad, N., Tajurruddin, F. W., Qin, W. Z., et al. (2020). Multi-strain probiotics (Hexbio) containing MCP BCMC strains improved constipation and gut motility in parkinson's disease: A randomised controlled trial. PLos One 15 (12), e0244680. doi: 10.1371/journal.pone.0244680

PubMed Abstract | CrossRef Full Text | Google Scholar

Kalia, L. V., Lang, A. E. (2015). Parkinson's disease. Lancet 386 (9996), 896–912. doi: 10.1016/s0140-6736(14)61393-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Krogh, K., Ostergaard, K., Sabroe, S., Laurberg, S. (2008). Clinical aspects of bowel symptoms in parkinson's disease. Acta Neurol. Scand. 117 (1), 60–64. doi: 10.1111/j.1600-0404.2007.00900.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Krüger, J. F., Hillesheim, E., Pereira, A., Camargo, C. Q., Rabito, E. I. (2021). Probiotics for dementia: a systematic review and meta-analysis of randomized controlled trials. Nutr. Rev. 79 (2), 160–170. doi: 10.1093/nutrit/nuaa037

PubMed Abstract | CrossRef Full Text | Google Scholar

Leta, V., Ray Chaudhuri, K., Milner, O., Chung-Faye, G., Metta, V., Pariante, C. M., et al. (2021). Neurogenic and anti-inflammatory effects of probiotics in parkinson's disease: A systematic review of preclinical and clinical evidence. Brain Behav. Immun. 98, 59–73. doi: 10.1016/j.bbi.2021.07.026

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, X., Lv, C., Song, J., Li, J. (2021). Effect of probiotic supplementation on cognitive function and metabolic status in mild cognitive impairment and alzheimer's disease: A meta-analysis. Front. Nutr. 8. doi: 10.3389/fnut.2021.757673

CrossRef Full Text | Google Scholar

Li, H., Zhang, M., Chen, L., Zhang, J., Pei, Z., Hu, A., et al. (2010). Nonmotor symptoms are independently associated with impaired health-related quality of life in Chinese patients with parkinson's disease. Mov Disord. 25 (16), 2740–2746. doi: 10.1002/mds.23368

PubMed Abstract | CrossRef Full Text | Google Scholar

Lyons, K. E., Pahwa, R. (2011). The impact and management of nonmotor symptoms of parkinson's disease. Am. J. Manag Care 17 Suppl 12, S308–S314.

PubMed Abstract | Google Scholar

Naomi, R., Embong, H., Othman, F., Ghazi, H. F., Maruthey, N., Bahari, H. (2021). Probiotics for alzheimer's disease: A systematic review. Nutrients 14 (1), 20. doi: 10.3390/nu14010020

PubMed Abstract | CrossRef Full Text | Google Scholar

Neta, F. I., de Souza, F. E. S., Batista, A. L., Pinheiro, F. I., Cobucci, R. N., Guzen, F. P. (2022). Effects of supplementation with probiotics in experimental models of alzheimer's disease: A systematic review of animal experiments. Curr. Alzheimer Res. 19 (3), 188–201. doi: 10.2174/1567205019666220318092003

PubMed Abstract | CrossRef Full Text | Google Scholar

Perez-Lloret, S., Rey, M. V., Pavy-Le Traon, A., Rascol, O. (2013). Emerging drugs for autonomic dysfunction in parkinson's disease. Expert Opin. Emerg. Drugs 18 (1), 39–53. doi: 10.1517/14728214.2013.766168

PubMed Abstract | CrossRef Full Text | Google Scholar

Pohl, D., Tutuian, R., Fried, M. (2008). Pharmacologic treatment of constipation: what is new? Curr. Opin. Pharmacol. 8 (6), 724–728. doi: 10.1016/j.coph.2008.07.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Poirier, A. A., Aubé, B., Côté, M., Morin, N., Di Paolo, T., Soulet, D. (2016). Gastrointestinal dysfunctions in parkinson's disease: Symptoms and treatments. Parkinsons Dis. 2016, 6762528. doi: 10.1155/2016/6762528

PubMed Abstract | CrossRef Full Text | Google Scholar

Sakakibara, R., Odaka, T., Uchiyama, T., Asahina, M., Yamaguchi, K., Yamaguchi, T., et al. (2003). Colonic transit time and rectoanal videomanometry in parkinson's disease. J. Neurol. Neurosurg. Psychiatry 74 (2), 268–272. doi: 10.1136/jnnp.74.2.268

PubMed Abstract | CrossRef Full Text | Google Scholar

Savica, R., Carlin, J. M., Grossardt, B. R., Bower, J. H., Ahlskog, J. E., Maraganore, D. M., et al. (2009). Medical records documentation of constipation preceding Parkinson disease: A case-control study. Neurology 73 (21), 1752–1758. doi: 10.1212/WNL.0b013e3181c34af5

PubMed Abstract | CrossRef Full Text | Google Scholar

Stocchi, F., Torti, M. (2017). Constipation in parkinson's disease. Int. Rev. Neurobiol. 134, 811–826. doi: 10.1016/bs.irn.2017.06.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Suez, J., Zmora, N., Segal, E., Elinav, E. (2019). The pros, cons, and many unknowns of probiotics. Nat. Med. 25 (5), 716–729. doi: 10.1038/s41591-019-0439-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Tan, A. H., Lim, S. Y., Chong, K. K., MAA, A. M., Hor, J. W., Lim, J. L., et al. (2021). Probiotics for constipation in Parkinson disease: A randomized placebo-controlled study. Neurology 96 (5), e772–ee82. doi: 10.1212/wnl.0000000000010998

PubMed Abstract | CrossRef Full Text | Google Scholar

Ticinesi, A., Tana, C., Nouvenne, A., Prati, B., Lauretani, F., Meschi, T. (2018). Gut microbiota, cognitive frailty and dementia in older individuals: a systematic review. Clin. Interv Aging 13, 1497–1511. doi: 10.2147/cia.S139163

PubMed Abstract | CrossRef Full Text | Google Scholar

Travagli, R. A., Browning, K. N., Camilleri, M. (2020). Parkinson Disease and the gut: New insights into pathogenesis and clinical relevance. Nat. Rev. Gastroenterol. Hepatol. 17 (11), 673–685. doi: 10.1038/s41575-020-0339-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Unger, M. M., Spiegel, J., Dillmann, K. U., Grundmann, D., Philippeit, H., Bürmann, J., et al. (2016). Short chain fatty acids and gut microbiota differ between patients with parkinson's disease and age-matched controls. Parkinsonism Relat. Disord. 32, 66–72. doi: 10.1016/j.parkreldis.2016.08.019

PubMed Abstract | CrossRef Full Text | Google Scholar

Verbaan, D., Marinus, J., Visser, M., van Rooden, S. M., Stiggelbout, A. M., van Hilten, J. J. (2007). Patient-reported autonomic symptoms in Parkinson disease. Neurology 69 (4), 333–341. doi: 10.1212/01.wnl.0000266593.50534.e8

PubMed Abstract | CrossRef Full Text | Google Scholar

Xiang, S., Ji, J. L., Li, S., Cao, X. P., Xu, W., Tan, L., et al. (2022). Efficacy and safety of probiotics for the treatment of alzheimer's disease, mild cognitive impairment, and parkinson's disease: A systematic review and meta-analysis. Front. Aging Neurosci. 14. doi: 10.3389/fnagi.2022.730036

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, C., Jiang, J., Tian, F., Zhao, J., Zhang, H., Zhai, Q., et al. (2020). Meta-analysis of randomized controlled trials of the effects of probiotics on functional constipation in adults. Clin. Nutr. 39 (10), 2960–2969. doi: 10.1016/j.clnu.2020.01.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhu, G., Zhao, J., Zhang, H., Chen, W., Wang, G. (2021). Probiotics for mild cognitive impairment and alzheimer's disease: A systematic review and meta-analysis. Foods 10 (7), 1672. doi: 10.3390/foods10071672

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: Parkinson, probiotics, constipation, systematic review, meta-analysis

Citation: Yin S and Zhu F (2022) Probiotics for constipation in Parkinson’s: A systematic review and meta-analysis of randomized controlled trials. Front. Cell. Infect. Microbiol. 12:1038928. doi: 10.3389/fcimb.2022.1038928

Received: 07 September 2022; Accepted: 25 October 2022;
Published: 10 November 2022.

Edited by:

Huang He, Tianjin University, China

Reviewed by:

Kwame Kumi Asare, University of Cape Coast, Ghana
Jie Chen, Huazhong University of Science and Technology, China

Copyright © 2022 Yin and Zhu. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Fengya Zhu, notfounds@foxmail.com

Download