Next Article in Journal
Acute and Chronic Exposure to Linagliptin, a Selective Inhibitor of Dipeptidyl Peptidase-4 (DPP-4), Has an Effect on Dopamine, Serotonin and Noradrenaline Level in the Striatum and Hippocampus of Rats
Next Article in Special Issue
Prebiotics and Probiotics: Healthy Biotools for Molecular Integrative and Modulation Approaches 2.0
Previous Article in Journal
Oxidative Stress and Cerebral Vascular Tone: The Role of Reactive Oxygen and Nitrogen Species
Previous Article in Special Issue
Probiotic-Induced Modulation of Microbiota Composition and Antibiotic Resistance Genes Load, an In Vitro Assessment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Lactiplantibacillus (Lactobacillus) plantarum as a Complementary Treatment to Improve Symptomatology in Neurodegenerative Disease: A Systematic Review of Open Access Literature

by
Ana Isabel Beltrán-Velasco
,
Manuel Reiriz
,
Sara Uceda
* and
Víctor Echeverry-Alzate
*
Psychology Department, School of Life and Nature Sciences, Nebrija University, 28240 Madrid, Spain
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(5), 3010; https://doi.org/10.3390/ijms25053010
Submission received: 14 December 2023 / Revised: 28 February 2024 / Accepted: 3 March 2024 / Published: 5 March 2024

Abstract

:
This systematic review addresses the use of Lactiplantibacillus (Lactobacillus) plantarum in the symptomatological intervention of neurodegenerative disease. The existence of gut microbiota dysbiosis has been associated with systemic inflammatory processes present in neurodegenerative disease, creating the opportunity for new treatment strategies. This involves modifying the strains that constitute the gut microbiota to enhance synaptic function through the gut–brain axis. Recent studies have evaluated the beneficial effects of the use of Lactiplantibacillus plantarum on motor and cognitive symptomatology, alone or in combination. This systematic review includes 20 research articles (n = 3 in human and n = 17 in animal models). The main result of this research was that the use of Lactiplantibacillus plantarum alone or in combination produced improvements in symptomatology related to neurodegenerative disease. However, one of the studies included reported negative effects after the administration of Lactiplantibacillus plantarum. This systematic review provides current and relevant information about the use of this probiotic in pathologies that present neurodegenerative processes such as Alzheimer’s disease, Parkinson’s disease and Multiple Sclerosis.

1. Introduction

1.1. The Neurodegenerative Disease

Neurodegenerative diseases refer to a group of pathologies that mainly affect the Nervous System (NS), and which involve tissue degeneration and cell death [1,2]. Lesions can be located in any region of the NS (spinal cord, brain, nerves, etc.), and affect cognitive and/or motor functioning. The impact of these pathologies is very significant [3,4]. For example, it is estimated that in 2050, 153 million people will have dementia [5]. The prevalence of Alzheimer’s disease is particularly relevant and currently affects almost 47 million people worldwide. Regarding Parkinson’s disease, 8.5 million people currently suffer from it [5,6].
One characteristic that is usually present in neurodegenerative diseases is the damage associated with oxidative stress [7,8,9,10]. Thus, it is traditionally studied in relation to cognitive impairment and neurodegeneration processes [11,12]. This phenomenon occurs when there is an imbalance between pro-oxidant and antioxidant chemicals. These pro-oxidants contain free electrons (which are also called free radicals) that have the ability to produce oxidative damage in fractions of a millisecond. However, this damage is sufficient to cause apoptosis and necrosis [13,14]. These mechanisms of action are involved in a number of degenerative pathologies, although it is not possible to confirm exactly whether they are the cause or effect of the clinical disease [15,16,17].
More than 100 neurodegenerative diseases or conditions associated with neuronal death have been described, among which Alzheimer’s Disease (AD), Parkinson’s Disease (PD) and Multiple Sclerosis (MS) stand out due to their prevalence [18,19]. AD is the most common form of dementia among older adults, with a prevalence of more than 46 million people affected worldwide [20,21].
Regarding PD, it is a neurodegenerative disease with premature death of dopaminergic neurons in the substantia nigra pars compacta in the midbrain [22,23]. In addition, atrophy of the basal ganglia is present, as well as dysfunction of communication between these subcortical regions and the prefrontal cortex [24,25,26,27]. Common symptoms in this disease are parkinsonism (bradykinesia and tremor or rigidity), postural instability and gait disturbances [28,29]. In terms of non-motor symptoms, these patients frequently present neuropsychiatric disorders, sleep disorders, autonomic dysfunction and gastrointestinal symptoms, among others [30,31,32].
Finally, MS is an autoimmune disease, although it causes degeneration in the brain and spinal cord. The damage is caused by an inflammatory process, which occurs when the immune system’s own cells attack the NS [33,34]. Because the lesions can be located in different regions, the symptoms are heterogeneous [35,36,37]. The same happens with each flare-up, when new lesions occur anywhere in the brain, optic nerve or spinal cord [38]. Among the most frequent symptoms, we find intestinal symptoms, such as urinary urgency or incontinence; muscular symptoms, such as spasms, stiffness, difficulty in coordination and gait; ocular symptoms such as discomfort, nystagmus or loss of vision; neurological symptoms such as impairment of executive functions such as memory or attention and difficulty in problem solving, among others [39,40,41,42].

1.2. The Impact of Gut Microbiota in Neurodegenerative Disease

There is growing evidence linking the gut microbiota (GM) with neurodegenerative diseases, but it is unclear if it is the consequence or the cause of the disease. The GM is formed by more than 100 million bacteria and more than 300 different species which live in our digestive tract [43,44,45,46]. Bacteria belonging to the phylum Bacillota (Eubacterium, Dorea, Ruminococcus), the phylum Actinomycetota (Bifidobacterium) and the phylum Bacteroidota (Alistipes, Bacteroides) are essential in the gut microbiota [47,48,49]. In addition, the bacteria, the GM is comprised of other microorganisms such as archaea, viruses, fungi, and protozoa [50,51].
It is known that the GM metabolites are involved in the onset and/or evolution of neurodegenerative diseases through different action mechanisms that alter endocrine pathways, immunological signals or neurological factors, among others [52,53]. When the GM is preserved, we talk about eubiosis; however, in some patients there is an alteration of the ratios of different species, and in this case, we speak of dysbiosis [54,55,56,57]. GM dysbiosis has been associated with neurodegenerative processes related to pathologies such as PD, AD or MS [58,59], and is usually accompanied by an increase in Bacteroidota as well as a reduction in the levels of Bacillota and Bifidobacterium [60,61,62].
This field of study is currently seen as an opportunity for intervention in these patients. The results obtained with the use of probiotics (live microorganisms that can be orally administrated as a food supplement or medicine) indicate that it is possible to modulate the microbiota, reducing organic inflammation and the symptoms associated with these neurodegenerative diseases [63,64,65]. Finally, the gut microbiota can modify the expression of certain genes that have been associated with different neurodegenerative diseases, as well as those involved in the mechanisms of cognitive functioning [66,67]. In addition, GM modulates metabolic functions, downregulating the low-grade inflammatory processes [68,69].
Among the most studied probiotics in different pathologies are Bifidobacterium (mainly longum and bifidum spp.) and the Lactobacillaceae family (mainly Lactobacillus paracasei, Lactobacillus casei and Lactoplantibacillus plantarum spp.) [70,71,72]. Bifidobacterium is a genus of gram-positive, anaerobic bacilli, and they utilize the fructose-6-phosphate phosphoketolase pathway. Lactobacillaceae is a family of long, curved or straight, gram-positive, anaerobic, lactic acid-forming bacilli [73,74,75,76,77]. Of these, Lactiplantibacillus (Lactobacillus) plantarum appears to have the most promising results in the intervention of symptoms associated with neurodegeneration [78,79,80]. Several authors have suggested that the use of Lactiplantibacillus plantarum, alone or in combination, improves specific symptoms such as motor, cognitive and psychiatric signs in neurodegenerative processes [81,82,83,84,85,86,87,88,89]. Research regarding these effects has increased in the last decades, making it necessary to compare with the results obtained. It is important to note that the term Lactobacillus plantarum was modified in 2020 for Lactiplantibacillus plantarum. Thus, in this Systematic Review, studies that included both terms were registered to ensure that all potential studies have been included.
The main objective of this Systematic Review was to collect all significant findings from both human and animal studies investigating the impact of Lactiplantibacillus plantarum administration on the improvement of motor and/or cognitive symptomatology in patients with neurodegenerative disease. This review will provide the most current data on the use of Lactiplantibacillus plantarum and intends to facilitate future research in this area.

2. Methods

2.1. Literature Search

The review followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement [90,91]. We searched for articles published between 2000 and 2024 in the following databases: ScienceDirect, Scopus and Web of Science. We used the following search: (Lactobacillus plantarum OR Lactiplantibacillus plantarum) AND Neurodegenerative disease. Two authors independently conducted the literature search in January 2024 (A.I.B.-V and S.U.), including the initial review of titles and abstracts, and the evaluation of retrievable articles for comprehensive review. Included articles were original research studies in English and Open Access.

2.2. Study Selection

In both human and animal studies, the inclusion criteria required: (i) a direct relationship between the use of Lactobacillus plantarum OR Lactiplantibacillus plantarum and changes in the symptomatology of neurodegenerative disease; (ii) the use of Lactobacillus plantarum OR Lactiplantibacillus plantarum as a therapeutic target in the process of neurodegeneration was addressed; (iii) the studies included any of the pathologies studied in this systematic review, which involve neuronal death associated with AD, PD, MCI and MS, oxidative stress and other neurodegenerative processes.

3. Results

A total of 907 articles were retrieved (Figure 1), of which only 29 articles were identified that fulfilled the eligibility criteria. Once duplicates were removed, the full titles and abstracts of all articles were examined for eligibility. Studies that addressed Lactobacillus plantarum OR Lactiplantibacillus plantarum unrelated to the neurodegeneration process were excluded: 5 were studies that did not include intervention with Lactobacillus plantarum OR Lactiplantibacillus plantarum and 4 were studies that focused on other pathologies not studied. Following this detailed review, a total of 20 studies were included. After the screening phase, all the selected articles were retrieved for comprehensive review, based on the established inclusion criteria.

3.1. Data Extraction

The data extracted from the included studies were as described below (Table 1):
Type of study (human or animal models);
Type of probiotic used (Lactobacillus plantarum or Lactiplantibacillus plantarum alone or in combination);
Neurodegenerative pathology addressed in the research;
Population (description);
Methodology of the research carried out;
Intervention (dose administered, time);
Results obtained after the intervention with Lactobacillus plantarum or Lactiplantibacillus plantarum.

3.2. Human Model

Three studies were conducted in humans: two address PD and the impact of the use of Lactobacillus plantarum or Lactiplantibacillus plantarum in the intervention of neurodegenerative processes in PD patients [92,93] (n = 2), and 1 of them explored MCI (Mild Cognitive Impairment) [94] (n = 1).

3.2.1. Parkinson’s Disease

Ghyselinck et al. (2021) used stool samples from three PD patients to create an in vitro intestinal model and dosed Symprove, a probiotic consisting of Lactobacillus acidophilus NCIMB 30175, Lactobacillus plantarum NCIMB 30173, Lactobacillus rhamnosus NCIMB 30174 and Enterococcus faecium NCIMB 30176 [92]. After 48 h of probiotic dose, the microbial community analysis showed significant changes, with elevated levels of Firmicutes, Actinobacteria and Bacteroidetes. In addition, the production of short-chain fatty acids (SCFA) and lactate was stimulated. Levels of anti-inflammatory cytokines (IL-6, IL-10) were also increased and levels of pro-inflammatory cytokines and chemokines (MCP-1, IL-8) were decreased (TEER-transepithelial electrical resistance (110.3 ± 1.3, p < 0.001)).
Another study conducted by Lu et al. (2021) analyzed the use of Lactobacillus plantarum PS128 for 12 weeks. The results showed that after 12 weeks of PS128 supplementation, UPDRS (Unified Parkinson’s Disease Rating Scale) motor scores improved significantly in both the OFF (−0.80 ± 1.85, p = 0.04) and ON (−2.56 ± 5.36, p = 0.007). In addition, PS128 intervention significantly improved ON and OFF period duration as well as PDQ-39 (Parkinson’s Disease Questionnaire) values (−5.68 ± 8.55, p = 0.031). However, no apparent effect of PS128 on non-motor symptoms of PD patients was observed [93].
It can be concluded that Lactiplantibacillus (Lactobacillus) plantarum improves motor symptomatology in PD due to its neuroprotective action through its microbial profile [92]. Although a significant effect on non-motor symptomatology could not be established, microbial modulation allows for the modification of the protective effects of anti-inflammatory cytokines [93].

3.2.2. Mild Cognitive Impairment

Fei et al. (2023) conducted a study with 42 patients diagnosed with ICM to whom they administered a probiotic mixture (Lactobacillus plantarum BioF-228, Lactococcus lactis BioF-224, Bifidobacterium lactis CP-9, Lactobacillus rhamnosus Bv-77, Lactobacillus johnsonii MH-68, Lactobacillus paracasei MP137, Lactobacillus salivarius AP-32, Lactobacillus acidophilus TYCA06, Lactococcus lactis LY-66, Bifidobacterium lactis HNO19, Lactobacillus rhamnosus HNO01, Lactobacillus paracasei GL-156, Bifidobacterium animalis BB-115, Lactobacillus casei CS-773, Lactobacillus reuteri TSR332, Lactobacillus fermentum TSF331, Bifidobacterium infantis BLI-02 and Lactobacillus plantarum CN2018), for 12 weeks. The results showed that the probiotic mixture improved cognitive function as well as sleep quality (Mini-Mental State Examination-MMSE (24.75 ± 2.47); Montreal Cognitive Assessment Scale-MoCA (22.05 ± 2.14 vs. a 20.10 ± 1.45); Pittsburgh Sleep Quality Index-PSQI (5.35 ± 2.78 vs. 8.40 ± 1.76,) (* p < 0.001).
In conclusion, the evidence suggests that treatment with this mixture of bacterial strains, which includes different species of Lactobacillaceae, can improve mild cognitive impairment and sleep quality [94].

3.3. Animal Model

A total of 17 studies were conducted using different animal models. Of these, 4 focused on AD [95,96,97,98] (n = 4); 7 focused on PD [99,100,101,102,103,104,105] (n = 7); 1 analyzed MS [106], (n = 1); 3 studied oxidative stress [107,108,109] (n = 3); 1 on cognitive impairment and oxidative stress [110] (n = 1); and 1 on neurodegenerative process [111] (n = 1). All of them addressed the intervention with Lactobacillus plantarum or Lactiplantibacillus plantarum for the improvement of symptoms associated with neurodegenerative pathologies.

3.3.1. Alzheimer’s Disease

Bonfili et al. (2020) conducted a study with a transgenic mouse model and administered the probiotic SLAB51 for a period of 16 to 48 weeks [95]. It contained eight different live bacterial strains: Streptococcus thermophilus DSM 32245, Bifidobacterium lactis DSM 32246, Bifidobacterium lactis DSM 32247, Lactobacillus acidophilus DSM 32241, Lactobacillus helveticus DSM 32242, Lactobacillus paracasei DSM 32243, Lactobacillus plantarum DSM 32244 and Lactobacillus brevis DSM 27961. The results obtained indicate that the ingestion of probiotics leads to an improvement in the altered brain glucose metabolism associated with the pathogenesis of AD, thereby delaying the progression of the disease (SLAB51 (* p < 0.05) vs. control and Veh.).
The study conducted by Lee et al. in 2021 in a transgenic mouse model addressed the impact on AD symptomatology of a probiotic composed by Lactobacillus plantarum NK151; NK173 and Bifidobacterium longum NK173 [96]. The results showed that the intervention reduced memory impairment and modulated gut bacterial composition in mice with AD cognitive impairment.
Mallikarjuna, Praveen, and Yellamma (2016) conducted a study in D-galactose-induced AD rats, and administered Lactobacillus plantarum MTCC1325 for 60 days [97]. The results demonstrated that, within a 30-day period, rats in the probiotic group exhibited a reversal of all ATPase enzyme components to normal levels. These data indicates that these probiotics exerts a protective action on the ATPase system in the brain, reducing the neurodegeneration (Alleviated Escherichia coli K1-induced cognitive impairment (# p < 0.05 vs. NC. * p < 0.05 vs. EC.).
A study conducted by Kaur et al. in 2021 in an AppNL-G-F (AD) mouse model analyzed the impact of the probiotic compound VSL#3® (Lactobacillus plantarum, Lactobacillus delbrueckii subsp. Bulgaricus, Lactobacillus paracasei, Lactobacillus acidophilus, Bifidobacterium breve, Bifidobacterium longum, Bifidobacterium infantis and Streptococcus salivarius subsp. Thermophilus) for 8 weeks [98]. This research was focused on finding differences by gender, and the results indicated that female mice had improved mnesic function, although no improvement was found in male mice. Similarly, a reduction in TNF-α levels in the brain was found in female mice (*Significant difference at p < 0.05 vs. control group. #Significant difference at p < 0.05 vs. AD group).
These results suggest that Lactiplantibacillus plantarum improves mnesic function, acts as a neuroprotectant in hippocampal cells delaying the neurodegenerative process [95,96], and reverses the action of ATPases in the brain [97]. Moreover, the study that focused on gender differences underscores the importance of conducting studies to analyze potential differences in the effects of probiotic use based on gender and to comprehend the mechanisms involved in these effects [98].

3.3.2. Parkinson’s Disease

Liao et al. (2020) conducted an investigation with a mouse model of MPTP-induced PD [99]. They administered Lactobacillus plantarum PS128 for 28 days and an increase in DA and norepinephrine were found in the striata. In addition, Lactobacillus plantarum PS128 was shown to reduce the death of nigrostriatal dopaminergic neurons. Similarly, Lactobacillus plantarum PS128 reduced MPTP-induced glial reactivity, and increased striatal neurotrophic factors (PS128 increased the DA levels and significantly improved the DOPAC (p < 0.05), HVA (p < 0.001), NE (p < 0.01) and MHPG (p < 0.001) levels in the MPTP-treated group).
Lee et al. (2023) conducted a study in a mouse model of PD [100]. Lactobacillus plantarum PS128 was administered for 6 weeks and the results showed a significant reduction in motor deficits and higher dopamine levels (PS128 significantly increased DA levels vs. ROT group (p < 0.05)). Similarly, a reduction in the loss of dopaminergic neurons and microglial activation was found. Levels of inflammatory factors were also reduced and an increase in the expression of neurotrophic factor was shown in the brain. Ingestion of Lactobacillus plantarum PS128 modified the microbial profile of PD mice and sustained neuroprotective effects by increasing the expression of the suppressor cytokine signaling 1 (SOCS1).
Along the same lines, Wang et al. in their 2021 study in a mouse model of PD induced by MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) [101], administered Lactobacillus plantarum DP189 for 14 days. The results indicated that this product exerted a significant neuroprotective effect on dopaminergic neurons, which improved motor symptoms (activities increased by 13.8% vs. model group (p < 0.05)). In addition, an increase in the levels of monoamines in the nervous system, namely 5-HT (5-hydroxytryptamine) and DA (dopamine) was also described. Another interesting result was that Lactobacillus plantarum DP189 promoted neuronal survival through a modulation of the ERK2 and AKT/mTOR pathways.
Pérez Visñuk, et al. conducted a study in 2020 [102], in a mouse model of PD induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. They administered Lactobacillus plantarum CRL 2130, Streptococcus thermophilus CRL 807 and Streptococcus thermophilus CRL 808 alone or in combination. The results revealed a notable enhancement in the motor skills of mice under both individual and combined administrations; however, the combined form appeared to be more effective. Moreover, the mechanisms of action did not appear to be entirely clear.
Pérez Visñuk et al. in 2022 conducted another study in a mouse model with PD induced by MPTP and probenecid, to whom they administered Lactobacillus plantarum CRL2130 [103]. The results showed that motor deficits were reduced, in addition to preventing the death of dopaminergic neurons. A reduction in proinflammatory cytokines and an increase in IL-10 were also observed compared to the control group, showing a neuroprotective effect. In this same study an in vitro analysis was conducted in which N2a cells were incubated with an intracellular extract of Lactobacillus plantarum CRL2130 and showed that viability was maintained and IL-6 release and reactive oxygen species (ROS) formation, all affected by MPP+, were significantly reduced (In vitro: CRL2130 reduced the oxidative stress and IL-6 (p < 0.05); In vivo: CRL2130 improved the motor deficits (p < 0.05)).
The study conducted by Xu et al. in 2020 addressed the impact of individual strains of Lactobacillus and Acetobacter in a Drosophila model with PINK mutations, and administered the prebiotic EGCG (eigallocatechin-3-gallate) [104]. The individual strains used were Lactobacillus plantarum KJ01, Acetobacter pomorum KJ02, Lactobacillus brevis KJ03, and Acetobacter pasteurianus KJ04. The prebiotic EGCG substantially modified the intestinal microbiota, restoring bacterial abundance, in addition to ameliorating existing dopaminergic deficits. On the other hand, the administration of individual strains of Lactobacillus or Acetobacter exacerbated the neuronal dysfunction reduced by EGCG (prolonged climbing latency in the PINK1B9 (p = 0.013) and PINK1B9+EGCG (p = 0.005) flies).
Another recent study was carried out in 2022 by Ilie et al. In this research, an adult zebrafish model (wild-type AB; WT, genetic line) with ROT-rotenone-induced motor impairment was used [105]. Then, the administration of different compounds was included to study their impact on these deficits. The PROBIOTIC group (Lactobacillus casei W56, Lactobacillus acidophilus W22, Lactobacillus paracasei W20, Lactobacillus salivarius W24, Lactobacillus lactis W19, Lactobacillus plantarum W62, Bifidobacterium lactis W51, Bifidobacterium lactis W52, and Bifidobacterium bifidum W23) and LEV+CARB (levodopa and carbidopa) group showed similar neuroprotective effects, decreasing the toxic effects of ROT, favoring neurogenesis and angiogenesis and their maintenance (PROBIO group: positive marking for PCNA, S100b, and GFAP, p53 and cox4i1; ROT+VPA, ROT+LEV/CARB, and ROT+PROBIO: increase in all IHC markers; ROT+PROBIO: PCNA marked a small number of cells).
Evidence suggests that the use of Lactiplantibacillus plantarum significantly reduces motor deficits. It also reduces levels of inflammatory factors and maintains neuroprotective effects on dopaminergic neurons, both in vivo and in vitro [99,100,101,102,103,104,105].

3.3.3. Multiple Sclerosis

Research carried out by Mestre et al. in 2020, in an experimental model of MS infected with TMEV-IDD (induced demyelinating disease), analyzed the impact of intervention with Vivomixx (probiotic composed of Lactobacillus paracasei DSM 24734, Lactobacillus plantarum DSM 24730, Lactobacillus acidophilus DSM 24735, Lactobacillus delbruckeii subspecies bulgaricus DSM 24734, Bifidobacterium longum DSM 24736, Bifidobacterium infantis DSM 24737, Bifidobacterium breve 24732 and Streptococcus thermophilus DSM 24731) during the chronic stage of the disease. It was administered to mice for 70 to 85 days [106].
Consumption of this probiotic improved motor function, showing an increase in horizontal and vertical activity of mice as opposed to the more limited activity of the vehicle-treated mice (p < 0.01 vs. TMEV-mice). In addition, it was shown that mice treated with Vivomixx underwent subtle changes in the composition of the gut microbiota. Regarding Vivomixx-TMEV mice group, the relative abundance of Anaerostipes, Dorea, Oscillospira, Enterobacteraceae or Ruminococcus decreased while Bacteroides, Odoribacter, Lactobacillus or Sutterella was increased.
This study concluded that the use of a multi-strain probiotic, which includes Lactobacillus plantarum, improves motor function [106].

3.3.4. Oxidative Stress

Shang et al. (2022) conducted a study with Luciobarbus capito (L. capito) in which they investigated the role of Selenium (Se)-enriched Lactobacillus plantarum on Cadmium (Cd)-induced oxidative stress [107]. Compared to the control group, the L. capito that received the product showed a significant decrease in Cd toxicity after one month of diet (p < 0.05), a decrease in oxidative capacity, and improved memory processes.
Xu et al. (2022) conducted a study with a mice model of alcohol-induced cognitive dysfunction. They administered Lactobacillus plantarum ST-III (LP-cs) for 28 days [108]. The results showed that LP-cs is able to significantly improve cognitive dysfunction and improved memory and learning. In addition, modifications were found in hippocampal morphology and synaptic dysfunction, reducing the impact of oxidative stress (Control vs. AE and AE vs. AE/LP-cs, * p < 0.05, ** p < 0.01, *** p < 0.001, n = 3–6).
The study by Westfall, Lomis and Prakash in 2018 addressed the impact of a probiotic compound (Lactobacillus plantarum NCIMB 8826 (Lp8826), Lactobacillus fermentum NCIMB 5221 (Lf5221) and Bifidobacteria longum spp. infantis NCIMB 702,255 (Bi702255)) and a symbiotic compound (probiotic formulation + 0.5% of TFLA power (Emblica officinalis, Terminalia bellirica and Terminalia chebula)) in an aged male Drosophila melanogaster model [109]. The results obtained indicated that both the probiotic compound alone and the symbiotic compound had beneficial effects on metabolic markers associated with aging in this animal model. Specifically, improvements in insulin-like signaling were found, reducing oxidative stress (p < 0.01).
Evidence suggests that the use of Lactiplantibacillus plantarum improves cognitive function, memory processes and learning, reducing the effect of oxidative stress [107,108,109].

3.3.5. Cognitive Decline and Oxidative Stress

Zaydi et al. (2020) conducted a study in a D-galactose-induced aged rat model [110]. They administered Lactobacillus plantarum DR7 for 12 weeks and studied its impact on cognitive impairment and oxidative stress. The results showed that the ingestion of Lactobacillus plantarum DR7 improved memory capacity. Anxiety level also was reduced in the DR7-treated group. In addition, the probiotic improved the serotoninergic pathway (MD 5.62; 95% CI 1.78 to 6.11; p < 0.05).
This study suggests that the use of Lactiplantibacillus plantarum improves cognitive impairment and oxidative stress, memory and anxiety symptomatology [110].

3.3.6. Neurodegenerative Process

Xia et al. (2024) conducted a study in a mice model of D-galactose-induced ageing. They administered Lactobacillus plantarum AR113 as a pre-treatment [111]. The results indicated that Lactobacillus plantarum AR113 significantly reduced D-galactose-induced oxidative stress injury, reducing cytotoxicity while maintaining cell membrane integrity and enhancing antioxidant enzyme activity. Furthermore, the results suggested that the expression of G protein-coupled receptor 78 (GPR78) and C/EBP homologous protein (CHOP) could be reduced, facilitating the restoration of endoplasmic reticulum (ER) homeostasis, thus activating cellular anti-apoptotic pathways (mRNA expression levels of the marker proteins GPR78 and CHOP were reduced to 0.04 and 0.26 times that of the Mod group, respectively, while the expression of PERK increased to 2.09 times that of the Mod group (p < 0.05)).
In conclusion, the use of Lactiplantibacillus plantarum reduced the damage caused by oxidative stress associated with the neurodegeneration process [111].

4. Discussion

The main objective of this systematic review was to compile all significant findings from human and animal studies investigating the impact of Lactiplantibacillus plantarum administration on the improvement of symptomatology in neurodegenerative disease. The results obtained indicated that Lactiplantibacillus plantarum either alone or in combination, improves these symptoms.
Regarding PD, motor symptomatology is the diagnostic mainstay of this condition, and the reduction of these symptoms results in an improvement in the patient’s quality of life [70,112,113,114]. Previous studies have shown that the quality of life in PD patients is not only directly related to motor symptoms but also to cognitive impairment, behavioral disturbances, and sleep disorders such as REM sleep disturbance [115,116,117,118,119,120]. The use of probiotics could have positive effects on the quality of life of PD patients by improving motor and non-motor symptomatology.
Human studies showed that Lactiplantibacillus plantarum, alone or in combination, improves motor symptoms and increases the proliferation of beneficial bacteria in the gut. These strains have shown neuro-modulatory abilities that delay the progression of PD [92,93]. Similarly, Lactobacillus plantarum improved cognitive function in people with MCI, as well as the quality and regularity of sleep [94]. Studies in the same line using different probiotics, such as Lactobacillus acidophilus, Bifidobacterium bifidum, Lactobacillus reuteri, and Lactobacillus fermentum, have shown a similar result. Thus, the use of this species produced a reduction in the oxidative stress and showed a neuroprotective effect. Moreover, the patient who took these probiotics showed a better score on the Movement Disorders Society–Unified Parkinson’s Disease Rating Scale. Therefore, the use of probiotics, especially those who belong to the Lactobacillus genus, seems to be useful to treat the motor and cognitive symptoms associated with PD.
Regarding the use of probiotics in PD animal models, studies showed a significant improvement in motor deficits following the administration of Lactiplantibacillus plantarum alone or in combination. Similarly, it showed neuroprotective effects, reducing dopaminergic neuronal death and glial reactivity, and increasing striatal neurotrophic factors. In addition, neurogenesis and angiogenesis were increased after the probiotic use [99,100,101,102]. Moreover, an in vitro study [103] showed a decrease in ROS. The study performed by Xu et al. in 2020 with a Drosophila model showed opposite results indicating that the PROBIO combination exacerbated neuronal disfunction that has been previously alleviated with EGCG (eigallocatechin-3-gallate) [104].
Analyzing other probiotics that could be used in the PD treatment, a recent study using an established Caenorhabditis elegans (roundworm) model of synucleinopathy was able to show that worms that received the probiotic strain Bacillus subtilis PXN21 hardly formed any alpha-synuclein aggregates, reducing the inflammatory response [121]. In other study conducted by Liu et al. in 2022 in a mouse model of chronic PD, they administered polymannuronic acid (PM) or Lacticaseibacillus rhamnosus GG (LGG), or their combination, showing neuroprotective effects independently, and a greater effect when administered in combination. Neurotrophic factor expression was enhanced and the level of Clostridial bacteria was increased. In addition, blood–brain barrier integrity was improved, and apoptosis in the striatum was inhibited [122]. It is therefore necessary to identify other strains that can be administered alone or in combination to enhance the neuroprotective effects.
Analyzing the results obtained from studies addressing AD, the use of Lactiplantibacillus plantarum alone or in combination showed benefits. Specifically, it improved AD-associated memory function [96], and neurodegeneration was reduced through processes in the brain’s ATPase system as well as the neuroprotective role exerted by the administered probiotic [97]. In addition, brain glucose metabolism, involved in the degenerative processes associated with AD, was improved [95]. Moreover, neuronal survival and synapse function were also protected. Finally, modulation of the gut microbiota led to a modification of the microbial profile, which exerts a neuroprotective function [95].
On the other hand, the study focused on gender differences, showing that Lactiplantibacillus plantarum improves memory in female mice and reduced TNF-a levels, although these improvements were not found in male mice [98].
In this line, other probiotics that have shown positive effects in ameliorating the symptomatology of Alzheimer’s disease (AD) include Bifidobacterium longum, which improved memory and plasticity in rats. Interventions with Lactobacillus acidophilus, Lactobacillus fermentum, Bifidobacterium lactis, and Bifidobacterium longum have demonstrated positive effects in both human and animal models [97,123,124].
The study of the impact of Lactiplantibacillus plantarum in MS intervention showed a significant improvement in the motor symptoms associated with the pathology, as well as an increase in anti-inflammatory activity in microglia, reducing oxidative stress. Furthermore, in the study of cognitive function, the administration of Lactiplantibacillus plantarum showed a decrease in memory loss [106].
Regarding the use of other probiotics in the MS treatment, others Lactobacillus spp., Bifidobacteriums (such as B. animalis) and Streptococcus showed a modulation in the CNS inflammation by T helper type 1 (Th1) and Th17 cells, which produce pro-inflammatory cytokines that damage the blood–brain barrier [125,126,127,128].
Moreover, studies analyzing oxidative stress, cognitive impairment, and neurodegenerative processes, along with the impact of various strains of Lactiplantibacillus plantarum, have shown significant improvements in reducing the loss of mitochondrial integrity; improvements in cognitive processes such as learning and memory, an increase in the activity of antioxidant enzymes, and a reduction in metabolic stress and inflammation [129,130,131,132,133]. As in other studies, the administration of Lactiplantibacillus plantarum was shown to modify hippocampal morphology and improve synaptic dysfunction [124,134,135].
While the utilization of probiotics appears promising in the treatment of neurodegenerative diseases, it is important to note that it is not the sole option available. An alternative approach involves the modulation of gut microbiota through fecal transplantation from healthy individuals, which has demonstrated efficacy in restoring eubiosis. This restoration enhances anti-inflammatory mechanisms and mitigates cellular oxidative processes. Notably, the application of this technique has proven to be a valuable treatment for a spectrum of neurodegenerative conditions, including PD, AD, MS, and neurodegenerative processes associated with oxidative stress [136,137,138,139].

Limitations and Future Research

The main limitation of this systematic review was the lack of clinical studies, which is consistent with the novelty of this area of study. However, this review is relevant and significant, as it provides the latest information currently available, focusing on the probiotic interventions. The inclusion of these details enables us to identify starting points for future research with a solid and rigorous empirical foundation.
Additional limitations arise from the heterogeneity of the utilized probiotics, as well as variations in quantities of CFUs and combinations of strains within probiotic formulations. Moreover, the mixture of probiotics with other substances further contributes to the complexity of these limitations and makes it difficult to reach solid conclusions on the beneficial effects of Lactiplantibacillus plantarum use alone or in combination. It is essential to emphasize that further research is necessary to elucidate the mechanisms of action for each probiotic. Additionally, determining the most appropriate administration protocol in each case is crucial to ensure the neuroprotective effects of the probiotic under consideration.
There is also a reduced number of studies indicating non-beneficial or non-existent effects of probiotics administration and it is necessary to also have information about these cases, to improve interventions.
This systematic review is the first to report and highlight the effects of Lactiplantibacillus (Lactobacillus) plantarum, alone or in combination, on the neurodegeneration processes present in neurodegenerative diseases. This information supports the development of interventions incorporating this probiotic to alleviate motor, cognitive, and behavioral symptoms linked to these pathologies.

Author Contributions

Conceptualization, all authors; methodology, all authors; writing—original draft preparation, all authors; writing—review and editing, all authors; visualization, all authors; supervision, S.U. and V.E.-A.; project administration, S.U. and V.E.-A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Tam, O.H.; Ostrow, L.W.; Hammell, M.G. Diseases of the nERVous system: Retrotransposon activity in neurodegenerative disease. Mob. DNA 2019, 10, 32. [Google Scholar] [CrossRef]
  2. Guzman-Martinez, L.; Maccioni, R.B.; Andrade, V.; Navarrete, L.P.; Pastor, M.G.; Ramos-Escobar, N. Neuroinflammation as a Common Feature of Neurodegenerative Disorders. Front. Pharmacol. 2019, 10, 1008. [Google Scholar] [CrossRef]
  3. Muddapu, V.R.; Dharshini, S.A.P.; Chakravarthy, V.S.; Gromiha, M.M. Neurodegenerative Diseases—Is Metabolic Deficiency the Root Cause? Front. Neurosci. 2020, 14, 213. [Google Scholar] [CrossRef]
  4. Rekatsina, M.; Paladini, A.; Piroli, A.; Zis, P.; Pergolizzi, J.V.; Varrassi, G. Pathophysiology and Therapeutic Perspectives of Oxidative Stress and Neurodegenerative Diseases: A Narrative Review. Adv. Ther. 2020, 37, 113–139. [Google Scholar] [CrossRef]
  5. Nichols, E.; Steinmetz, J.D.; Vollset, S.E.; Fukutaki, K.; Chalek, J.; Abd-Allah, F.; Abdoli, A.; Abualhasan, A.; Abu-Gharbieh, E.; Akram, T.T.; et al. Estimation of the global prevalence of dementia in 2019 and forecasted prevalence in 2050: An analysis for the Global Burden of Disease Study 2019. Lancet Public Health 2022, 7, e105–e125. [Google Scholar] [CrossRef] [PubMed]
  6. Harciarek, M.; Kertesz, A. The Prevalence of Misidentification Syndromes in Neurodegenerative Diseases. Alzheimer Dis. Assoc. Disord. 2008, 22, 163–169. [Google Scholar] [CrossRef] [PubMed]
  7. Wang, X.; Zhou, Y.; Gao, Q.; Ping, D.; Wang, Y.; Wu, W.; Lin, X.; Fang, Y.; Zhang, J.; Shao, A. The Role of Exosomal microRNAs and Oxidative Stress in Neurodegenerative Diseases. Oxidative Med. Cell. Longev. 2020, 2020, 1–17. [Google Scholar] [CrossRef] [PubMed]
  8. Teleanu, D.M.; Niculescu, A.G.; Lungu, I.I.; Radu, C.I.; Vladâcenco, O.; Roza, E.; Costăchescu, B.; Grumezescu, A.M.; Teleanu, R.I. An Overview of Oxidative Stress, Neuroinflammation, and Neurodegenerative Diseases. Int. J. Mol. Sci. 2022, 23, 5938. [Google Scholar] [CrossRef] [PubMed]
  9. Rehman, M.U.; Sehar, N.; Dar, N.J.; Khan, A.; Arafah, A.; Rashid, S.; Rashid, S.M.; Ganaie, M.A. Mitochondrial dysfunctions, oxidative stress and neuroinflammation as therapeutic targets for neurodegenerative diseases: An update on current advances and impediments. Neurosci. Biobehav. Rev. 2023, 144, 104961. [Google Scholar] [CrossRef]
  10. Clemente-Suárez, V.J.; Redondo-Flórez, L.; Beltrán-Velasco, A.I.; Ramos-Campo, D.J.; Belinchón-deMiguel, P.; Martinez-Guardado, I.; Dalamitros, A.A.; Yáñez-Sepúlveda, R.; Martín-Rodríguez, A.; Tornero-Aguilera, J.F. Mitochondria and Brain Disease: A Comprehensive Review of Pathological Mechanisms and Therapeutic Opportunities. Biomedicines 2023, 11, 2488. [Google Scholar] [CrossRef]
  11. Cenini, G.; Lloret, A.; Cascella, R. Oxidative Stress in Neurodegenerative Diseases: From a Mitochondrial Point of View. Oxidative Med. Cell. Longev. 2019, 2019, 1–18. [Google Scholar] [CrossRef] [PubMed]
  12. Konovalova, J.; Gerasymchuk, D.; Parkkinen, I.; Chmielarz, P.; Domanskyi, A. Interplay between MicroRNAs and Oxidative Stress in Neurodegenerative Diseases. Int. J. Mol. Sci. 2019, 20, 6055. [Google Scholar] [CrossRef] [PubMed]
  13. Singh, A.; Kukreti, R.; Saso, L.; Kukreti, S. Oxidative Stress: A Key Modulator in Neurodegenerative Diseases. Molecules 2019, 24, 1583. [Google Scholar] [CrossRef] [PubMed]
  14. Wu, Y.; Chen, M.; Jiang, J. Mitochondrial dysfunction in neurodegenerative diseases and drug targets via apoptotic signaling. Mitochondrion 2019, 49, 35–45. [Google Scholar] [CrossRef]
  15. Callizot, N.; Combes, M.; Henriques, A.; Poindron, P. Necrosis, apoptosis, necroptosis, three modes of action of dopaminergic neuron neurotoxins. PLoS ONE 2019, 14, e0215277. [Google Scholar] [CrossRef]
  16. Saleem, S. Apoptosis, Autophagy, Necrosis and Their Multi Galore Crosstalk in Neurodegeneration. Neuroscience 2021, 469, 162–174. [Google Scholar] [CrossRef]
  17. Shandilya, S.; Kumar, S.; Jha, N.K.; Kesari, K.K.; Ruokolainen, J. Interplay of gut microbiota and oxidative stress: Perspective on neurodegeneration and neuroprotection. J. Adv. Res. 2022, 38, 223–244. [Google Scholar] [CrossRef]
  18. Shang, H.; Zhao, X.; Zhang, X. Neurodegenerative Diseases. In Pediatric Neuroimaging; Springer: Singapore, 2022; pp. 211–214. [Google Scholar]
  19. Mohammadi, A.; Colagar, A.H.; Khorshidian, A.; Amini, S.M. The Functional Roles of Curcumin on Astrocytes in Neurodegenerative Diseases. Neuroimmunomodulation 2022, 29, 4–14. [Google Scholar] [CrossRef]
  20. Litke, R.; Garcharna, L.C.; Jiwani, S.; Neugroschl, J. Modifiable Risk Factors in Alzheimer Disease and Related Dementias: A Review. Clin. Ther. 2021, 43, 953–965. [Google Scholar] [CrossRef] [PubMed]
  21. World Health Organization. Fact Sheets of Dementia; World Health Organization: Geneva, Switzerland, 2021; Available online: https:// www.who.int/news-room/fact-sheets/detail/dementia (accessed on 2 March 2024).
  22. Shahnawaz, M.; Mukherjee, A.; Pritzkow, S.; Mendez, N.; Rabadia, P.; Liu, X.; Hu, B.; Schmeichel, A.; Singer, W.; Wu, G.; et al. Discriminating α-synuclein strains in Parkinson’s disease and multiple system atrophy. Nature 2020, 578, 273–277. [Google Scholar] [CrossRef] [PubMed]
  23. Mus, L.; Siani, F.; Giuliano, C.; Ghezzi, C.; Cerri, S.; Blandini, F. Development and biochemical characterization of a mouse model of Parkinson’s disease bearing defective glucocerebrosidase activity. Neurobiol. Dis. 2019, 124, 289–296. [Google Scholar] [CrossRef]
  24. Meoni, S.; Cury, R.G.; Moro, E. New players in basal ganglia dysfunction in Parkinson’s disease. Prog. Brain Res. 2020, 252, 307–327. [Google Scholar]
  25. Navarro-López, E.M.; Çelikok, U.; Şengör, N.S. A dynamical model for the basal ganglia-thalamo-cortical oscillatory activity and its implications in Parkinson’s disease. Cogn. Neurodyn. 2021, 15, 693–720. [Google Scholar] [CrossRef]
  26. Adler, A.F.; Cardoso, T.; Nolbrant, S.; Mattsson, B.; Hoban, D.B.; Jarl, U.; Wahlestedt, J.N.; Grealish, S.; Björklund, A.; Parmar, M. hESC-Derived Dopaminergic Transplants Integrate into Basal Ganglia Circuitry in a Preclinical Model of Parkinson’s Disease. Cell Rep. 2019, 28, 3462–3473.e5. [Google Scholar] [CrossRef]
  27. Park, Y.W.; Shin, N.; Chung, S.J.; Kim, J.; Lim, S.M.; Lee, P.H.; Lee, S.; Ahn, K.J. Magnetic Resonance Imaging–Visible Perivascular Spaces in Basal Ganglia Predict Cognitive Decline in Parkinson’s Disease. Mov. Disord. 2019, 34, 1672–1679. [Google Scholar] [CrossRef]
  28. Weintraub, D.; Aarsland, D.; Chaudhuri, K.R.; Dobkin, R.D.; Leentjens, A.F.; Rodriguez-Violante, M.; Schrag, A. The neuropsychiatry of Parkinson’s disease: Advances and challenges. Lancet Neurol. 2022, 21, 89–102. [Google Scholar] [CrossRef]
  29. Armstrong, M.J.; Okun, M.S. Diagnosis and Treatment of Parkinson Disease. JAMA 2020, 323, 548–560. [Google Scholar] [CrossRef] [PubMed]
  30. Peball, M.; Krismer, F.; Knaus, H.; Djamshidian, A.; Werkmann, M.; Carbone, F.; Ellmerer, P.; Heim, B.; Marini, K.; Valent, D.; et al. Non-Motor Symptoms in Parkinson’s Disease are Reduced by Nabilone. Ann. Neurol. 2020, 88, 712–722. [Google Scholar] [CrossRef] [PubMed]
  31. Babiloni, A.H.; Bellemare, A.; Beetz, G.; Vinet, S.-A.; Martel, M.O.; Lavigne, G.J.; De Beaumont, L. The effects of non-invasive brain stimulation on sleep disturbances among different neurological and neuropsychiatric conditions: A systematic review. Sleep Med. Rev. 2021, 55, 101381. [Google Scholar] [CrossRef]
  32. Maggi, G.; Trojano, L.; Barone, P.; Santangelo, G. Sleep Disorders and Cognitive Dysfunctions in Parkinson’s Disease: A Meta-Analytic Study. Neuropsychol. Rev. 2021, 31, 643–682. [Google Scholar] [CrossRef]
  33. Adiele, R.C.; Adiele, C.A. Metabolic defects in multiple sclerosis. Mitochondrion 2019, 44, 7–14. [Google Scholar] [CrossRef] [PubMed]
  34. Sasso, B.L.; Agnello, L.; Bivona, G.; Bellia, C.; Ciaccio, M. Cerebrospinal Fluid Analysis in Multiple Sclerosis Diagnosis: An Update. Medicina 2019, 55, 245. [Google Scholar] [CrossRef] [PubMed]
  35. Absinta, M.; Sati, P.; Masuzzo, F.; Nair, G.; Sethi, V.; Kolb, H.; Ohayon, J.; Wu, T.; Cortese, I.C.M.; Reich, D.S. Association of Chronic Active Multiple Sclerosis Lesions With Disability In Vivo. JAMA Neurol. 2019, 76, 1474–1483. [Google Scholar] [CrossRef] [PubMed]
  36. Zeng, C.; Gu, L.; Liu, Z.; Zhao, S. Review of Deep Learning Approaches for the Segmentation of Multiple Sclerosis Lesions on Brain MRI. Front. Neurosci. 2020, 14, 610967. [Google Scholar] [CrossRef]
  37. Calvi, A.; Haider, L.; Prados, F.; Tur, C.; Chard, D.; Barkhof, F. In vivo imaging of chronic active lesions in multiple sclerosis. Mult. Scler. J. 2022, 28, 683–690. [Google Scholar] [CrossRef]
  38. Villarreal, J.V.; Abraham, M.J.; Acevedo, J.A.; Rai, P.K.; Thottempudi, N.; Fang, X.; Gogia, B. Tumefactive multiple sclerosis (TMS): A case series of this challenging variant of MS. Mult. Scler. Relat. Disord. 2021, 48, 102699. [Google Scholar] [CrossRef]
  39. Vaddepally, R.; Sodavarapu, S.; Kutadi, A.; Taylor, W.; Kumar, N. Case Report: A case of immune checkpoint inhibitor therapy in a patient with multiple sclerosis. F1000Research 2020, 9, 1167. [Google Scholar] [CrossRef]
  40. McGinley, M.P.; Goldschmidt, C.H.; Rae-Grant, A.D. Diagnosis and Treatment of Multiple Sclerosis. JAMA 2021, 325, 765–779. [Google Scholar] [CrossRef]
  41. Silveira, C.; Guedes, R.; Maia, D.; Curral, R.; Coelho, R. Neuropsychiatric Symptoms of Multiple Sclerosis: State of the Art. Psychiatry Investig. 2019, 16, 877–888. [Google Scholar] [CrossRef]
  42. Rommer, P.S.; Eichstädt, K.; Ellenberger, D.; Flachenecker, P.; Friede, T.; Haas, J.; Kleinschnitz, C.; Pöhlau, D.; Rienhoff, O.; Stahmann, A.; et al. Symptomatology and symptomatic treatment in multiple sclerosis: Results from a nationwide MS registry. Mult. Scler. J. 2019, 25, 1641–1652. [Google Scholar] [CrossRef]
  43. Uceda, S.; Echeverry-Alzate, V.; Reiriz-Rojas, M.; Martínez-Miguel, E.; Pérez-Curiel, A.; Gómez-Senent, S.; Beltrán-Velasco, A.I. Gut Microbial Metabolome and Dysbiosis in Neurodegenerative Diseases: Psychobiotics and Fecal Microbiota Transplantation as a Therapeutic Approach—A Comprehensive Narrative Review. Int. J. Mol. Sci. 2023, 24, 13294. [Google Scholar] [CrossRef]
  44. Rinninella, E.; Raoul, P.; Cintoni, M.; Franceschi, F.; Miggiano, G.A.; Gasbarrini, A.; Mele, M.C. What Is the Healthy Gut Microbiota Composition? A Changing Ecosystem across Age, Environment, Diet, and Diseases. Microorganisms 2019, 7, 14. [Google Scholar] [CrossRef] [PubMed]
  45. Al Bander, Z.; Nitert, M.D.; Mousa, A.; Naderpoor, N. The Gut Microbiota and Inflammation: An Overview. Int. J. Environ. Res. Public Health 2020, 17, 7618. [Google Scholar] [CrossRef] [PubMed]
  46. Verhaar, B.J.H.; Hendriksen, H.M.A.; de Leeuw, F.A.; Doorduijn, A.S.; van Leeuwenstijn, M.; Teunissen, C.E.; Barkhof, F.; Scheltens, P.; Kraaij, R.; van Duijn, C.M.; et al. Gut Microbiota Composition Is Related to AD Pathology. Front. Immunol. 2022, 12, 794519. [Google Scholar] [CrossRef] [PubMed]
  47. Michaudel, C.; Sokol, H. The Gut Microbiota at the Service of Immunometabolism. Cell Metab. 2020, 32, 514–523. [Google Scholar] [CrossRef]
  48. Gomaa, E.Z. Human gut microbiota/microbiome in health and diseases: A review. Antonie van Leeuwenhoek 2020, 113, 2019–2040. [Google Scholar] [CrossRef]
  49. Chen, Y.; Zhou, J.; Wang, L. Role and Mechanism of Gut Microbiota in Human Disease. Front. Cell. Infect. Microbiol. 2021, 11, 86. [Google Scholar] [CrossRef]
  50. Dingeo, G.; Brito, A.; Samouda, H.; Iddir, M.; La Frano, M.R.; Bohn, T. Phytochemicals as modifiers of gut microbial communities. Food Funct. 2020, 11, 8444–8471. [Google Scholar] [CrossRef]
  51. Marć, M.A.; Jastrząb, R.; Mytych, J. Does the Gut Microbial Metabolome Really Matter? The Connection between GUT Metabolome and Neurological Disorders. Nutrients 2022, 14, 3967. [Google Scholar] [CrossRef]
  52. Chen, Z.; Maqbool, J.; Sajid, F.; Hussain, G.; Sun, T. Human gut microbiota and its association with pathogenesis and treatments of neurodegenerative diseases. Microb. Pathog. 2021, 150, 104675. [Google Scholar] [CrossRef]
  53. Ning, J.; Huang, S.Y.; Chen, S.D.; Zhang, Y.R.; Huang, Y.Y.; Yu, J.T. Investigating Casual Associations Among Gut Microbiota, Metabolites, and Neurodegenerative Diseases: A Mendelian Randomization Study. J. Alzheimer’s Dis. 2022, 87, 211–222. [Google Scholar] [CrossRef]
  54. Wallace, T.C.; Guarner, F.; Madsen, K.; Cabana, M.D.; Gibson, G.; Hentges, E.; Sanders, M.E. Human gut microbiota and its relationship to health and disease. Nutr. Rev. 2011, 69, 392–403. [Google Scholar] [CrossRef]
  55. Lozupone, C.A.; Stombaugh, J.I.; Gordon, J.I.; Jansson, J.K.; Knight, R. Diversity, stability and resilience of the human gut microbiota. Nature 2012, 489, 220–230. [Google Scholar] [CrossRef]
  56. Harmsen, H.J.M.; de Goffau Marcus, C. The Human Gut Microbiota. In Microbiota of the Human Body: Implications in Health and Disease; Springer: Berlin/Heidelberg, Germany, 2016; pp. 95–108. [Google Scholar]
  57. Al-Rashidi, H.E. Gut microbiota and immunity relevance in eubiosis and dysbiosis. Saudi J. Biol. Sci. 2022, 29, 1628–1643. [Google Scholar] [CrossRef]
  58. Jurjus, A.; Eid, A.; Al Kattar, S.; Zeenny, M.N.; Gerges-Geagea, A.; Haydar, H.; Hilal, A.; Oueidat, D.; Matar, M.; Tawilah, J.; et al. Inflammatory bowel disease, colorectal cancer and type 2 diabetes mellitus: The links. BBA Clin. 2016, 5, 16–24. [Google Scholar] [CrossRef]
  59. Borrego-Ruiz, A.; Borrego, J.J. An updated overview on the relationship between human gut microbiome dysbiosis and psychiatric and psychological disorders. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2024, 128, 110861. [Google Scholar] [CrossRef]
  60. Aponte, M.; Murru, N.; Shoukat, M. Therapeutic, Prophylactic, and Functional Use of Probiotics: A Current Perspective. Front. Microbiol. 2020, 11, 562048. [Google Scholar] [CrossRef]
  61. Tremblay, A.; Lingrand, L.; Maillard, M.; Feuz, B.; Tompkins, T.A. The effects of psychobiotics on the microbiota-gut-brain axis in early-life stress and neuropsychiatric disorders. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2021, 105, 110142. [Google Scholar] [CrossRef]
  62. Ağagündüz, D.; Kocaadam-Bozkurt, B.; Bozkurt, O.; Sharma, H.; Esposito, R.; Özoğul, F.; Capasso, R. Microbiota alteration and modulation in Alzheimer’s disease by gerobiotics: The gut-health axis for a good mind. Biomed. Pharmacother. 2022, 153, 113430. [Google Scholar] [CrossRef]
  63. Faulin, T.d.E.S.; Estadella, D. Alzheimer’s disease and its relationship with the microbiota-gut-brain axis. Arq. Gastroenterol. 2023, 60, 144–154. [Google Scholar] [CrossRef]
  64. Morella, I.; Negro, M.; Dossena, M.; Brambilla, R.; D’Antona, G. Gut-muscle-brain axis: Molecular mechanisms in neurodegenerative disorders and potential therapeutic efficacy of probiotic supplementation coupled with exercise. Neuropharmacology 2023, 240, 109718. [Google Scholar] [CrossRef]
  65. Kumar, H.; Dhalaria, R.; Guleria, S.; Cimler, R.; Sharma, R.; Siddiqui, S.A.; Valko, M.; Nepovimova, E.; Dhanjal, D.S.; Singh, R.; et al. Anti-oxidant potential of plants and probiotic spp. in alleviating oxidative stress induced by H2O2. Biomed. Pharmacother. 2023, 165, 115022. [Google Scholar] [CrossRef]
  66. Suganya, K.; Koo, B.-S. Gut–Brain Axis: Role of Gut Microbiota on Neurological Disorders and How Probiotics/Prebiotics Beneficially Modulate Microbial and Immune Pathways to Improve Brain Functions. Int. J. Mol. Sci. 2020, 21, 7551. [Google Scholar] [CrossRef]
  67. Shabbir, U.; Arshad, M.S.; Sameen, A.; Oh, D.-H. Crosstalk between Gut and Brain in Alzheimer’s Disease: The Role of Gut Microbiota Modulation Strategies. Nutrients 2021, 13, 690. [Google Scholar] [CrossRef]
  68. Wang, P.; Gao, J.; Ke, W.; Wang, J.; Li, D.; Liu, R.; Jia, Y.; Wang, X.; Chen, X.; Chen, F.; et al. Resveratrol reduces obesity in high-fat diet-fed mice via modulating the composition and metabolic function of the gut microbiota. Free Radic. Biol. Med. 2020, 156, 83–98. [Google Scholar] [CrossRef]
  69. Fan, Y.; Pedersen, O. Gut microbiota in human metabolic health and disease. Nat. Rev. Microbiol. 2021, 19, 55–71. [Google Scholar] [CrossRef]
  70. Leta, V.; Chaudhuri, K.R.; Milner, O.; Chung-Faye, G.; Metta, V.; Pariante, C.M.; Borsini, A. Neurogenic and anti-inflammatory effects of probiotics in Parkinson’s disease: A systematic review of preclinical and clinical evidence. Brain Behav. Immun. 2021, 98, 59–73. [Google Scholar] [CrossRef]
  71. Socała, K.; Doboszewska, U.; Szopa, A.; Serefko, A.; Włodarczyk, M.; Zielińska, A.; Poleszak, E.; Fichna, J.; Wlaź, P. The role of microbiota-gut-brain axis in neuropsychiatric and neurological disorders. Pharmacol. Res. 2021, 172, 105840. [Google Scholar] [CrossRef]
  72. Korf, J.M.; Ganesh, B.P.; McCullough, L.D. Gut dysbiosis and age-related neurological diseases in females. Neurobiol. Dis. 2022, 168, 105695. [Google Scholar] [CrossRef]
  73. Aizawa, E.; Tsuji, H.; Asahara, T.; Takahashi, T.; Teraishi, T.; Yoshida, S.; Koga, N.; Hattori, K.; Ota, M.; Kunugi, H. Bifidobacterium and Lactobacillus Counts in the Gut Microbiota of Patients With Bipolar Disorder and Healthy Controls. Front. Psychiatry 2019, 9, 730. [Google Scholar] [CrossRef]
  74. Monteiro, C.R.A.V.; Carmo, M.S.D.; Melo, B.O.; Alves, M.S.; dos Santos, C.I.; Monteiro, S.G.; Bomfim, M.R.Q.; Fernandes, E.S.; Monteiro-Neto, V. In Vitro Antimicrobial Activity and Probiotic Potential of Bifidobacterium and Lactobacillus against Species of Clostridium. Nutrients 2019, 11, 448. [Google Scholar] [CrossRef] [PubMed]
  75. Zhang, C.; Zhang, Y.; Li, H.; Liu, X. The potential of proteins, hydrolysates and peptides as growth factors for Lactobacillus and Bifidobacterium: Current research and future perspectives. Food Funct. 2020, 11, 1946–1957. [Google Scholar] [CrossRef]
  76. Wang, C.-H.; Yen, H.-R.; Lu, W.-L.; Ho, H.-H.; Lin, W.-Y.; Kuo, Y.-W.; Huang, Y.-Y.; Tsai, S.-Y.; Lin, H.-C. Adjuvant Probiotics of Lactobacillus salivarius subsp. salicinius AP-32, L. johnsonii MH-68, and Bifidobacterium animalis subsp. lactis CP-9 Attenuate Glycemic Levels and Inflammatory Cytokines in Patients With Type 1 Diabetes Mellitus. Front. Endocrinol. (Lausanne) 2022, 13, 754401. [Google Scholar] [CrossRef]
  77. Ni, Y.; Zhang, Y.; Zheng, L.; Rong, N.; Yang, Y.; Gong, P.; Yang, Y.; Siwu, X.; Zhang, C.; Zhu, L.; et al. Bifidobacterium and Lactobacillus improve inflammatory bowel disease in zebrafish of different ages by regulating the intestinal mucosal barrier and microbiota. Life Sci. 2023, 324, 121699. [Google Scholar] [CrossRef]
  78. Mohammadi, G.; Dargahi, L.; Peymani, A.; Mirzanejad, Y.; Alizadeh, S.A.; Naserpour, T.; Nassiri-Asl, M. The Effects of Probiotic Formulation Pretreatment (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) on a Lipopolysaccharide Rat Model. J. Am. Coll. Nutr. 2019, 38, 209–217. [Google Scholar] [CrossRef] [PubMed]
  79. Mohammadi, G.; Dargahi, L.; Naserpour, T.; Mirzanejad, Y.; Alizadeh, S.A.; Peymani, A.; Nassiri-Asl, M. Probiotic mixture of Lactobacillus helveticus R0052 and Bifidobacterium longum R0175 attenuates hippocampal apoptosis induced by lipopolysaccharide in rats. Int. Microbiol. 2019, 22, 317–323. [Google Scholar] [CrossRef]
  80. Un-Nisa, A.; Khan, A.; Zakria, M.; Siraj, S.; Ullah, S.; Tipu, M.K.; Ikram, M.; Kim, M.O. Updates on the Role of Probiotics against Different Health Issues: Focus on Lactobacillus. Int. J. Mol. Sci. 2022, 24, 142. [Google Scholar] [CrossRef] [PubMed]
  81. Cheng, L.-H.; Liu, Y.-W.; Wu, C.-C.; Wang, S.; Tsai, Y.-C. Psychobiotics in mental health, neurodegenerative and neurodevelopmental disorders. J. Food Drug Anal. 2019, 27, 632–648. [Google Scholar] [CrossRef]
  82. Kong, Y.; Wang, L.; Jiang, B. The Role of Gut Microbiota in Aging and Aging Related Neurodegenerative Disorders: Insights from Drosophila Model. Life 2021, 11, 855. [Google Scholar] [CrossRef]
  83. Sun, P.; Su, L.; Zhu, H.; Li, X.; Guo, Y.; Du, X.; Zhang, L.; Qin, C. Gut Microbiota Regulation and Their Implication in the Development of Neurodegenerative Disease. Microorganisms 2021, 9, 2281. [Google Scholar] [CrossRef]
  84. Ojha, S.; Patil, N.; Jain, M.; Kole, C.; Kaushik, P. Probiotics for Neurodegenerative Diseases: A Systemic Review. Microorganisms 2023, 11, 1083. [Google Scholar] [CrossRef]
  85. Patel, C.; Pande, S.; Acharya, S. Potentiation of anti-Alzheimer activity of curcumin by probiotic Lactobacillus rhamnosus UBLR-58 against scopolamine-induced memory impairment in mice. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2020, 393, 1955–1962. [Google Scholar] [CrossRef]
  86. Wang, L.; Zhao, Z.; Zhao, L.; Zhao, Y.; Yang, G.; Wang, C.; Gao, L.; Niu, C.; Li, S. Lactobacillus plantarum DP189 Reduces α-SYN Aggravation in MPTP-Induced Parkinson’s Disease Mice via Regulating Oxidative Damage, Inflammation, and Gut Microbiota Disorder. J. Agric. Food Chem. 2022, 70, 1163–1173. [Google Scholar] [CrossRef]
  87. Bi, M.; Liu, C.; Wang, Y.; Liu, S.-J. Therapeutic Prospect of New Probiotics in Neurodegenerative Diseases. Microorganisms 2023, 11, 1527. [Google Scholar] [CrossRef]
  88. Mateo, D.; Marquès, M.; Domingo, J.L.; Torrente, M. Influence of gut microbiota on the development of most prevalent neurodegenerative dementias and the potential effect of probiotics in elderly: A scoping review. Am. J. Med. Genet. Part B Neuropsychiatr. Genet. 2023, 195, e32959. [Google Scholar] [CrossRef]
  89. Solanki, R.; Karande, A.; Ranganathan, P. Emerging role of gut microbiota dysbiosis in neuroinflammation and neurodegeneration. Front. Neurol. 2023, 14, 1149618. [Google Scholar] [CrossRef]
  90. Moher, D.; Shamseer, L.; Clarke, M.; Ghersi, D.; Liberati, A.; Petticrew, M.; Shekelle, P.; Stewart, L.A.; Prisma-P Group. Preferred reporting items for systematic review and meta-analysis protocols (PRISMA-P) 2015 statement. Syst. Rev. 2015, 4, 1. [Google Scholar] [CrossRef]
  91. Page, M.J.; E McKenzie, J.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Moher, D. Updating guidance for reporting systematic reviews: Development of the PRISMA 2020 statement. J. Clin. Epidemiol. 2021, 134, 103–112. [Google Scholar] [CrossRef]
  92. Ghyselinck, J.; Verstrepen, L.; Moens, F.; Abbeele, P.V.D.; Bruggeman, A.; Said, J.; Smith, B.; Barker, L.A.; Jordan, C.; Leta, V.; et al. Influence of probiotic bacteria on gut microbiota composition and gut wall function in an in-vitro model in patients with Parkinson’s disease. Int. J. Pharm. X 2021, 3, 100087. [Google Scholar] [CrossRef]
  93. Lu, C.-S.; Chang, H.-C.; Weng, Y.-H.; Chen, C.-C.; Kuo, Y.-S.; Tsai, Y.-C. The Add-On Effect of Lactobacillus plantarum PS128 in Patients With Parkinson’s Disease: A Pilot Study. Front. Nutr. 2021, 8, 650053. [Google Scholar] [CrossRef]
  94. Fei, Y.; Wang, R.; Lu, J.; Peng, S.; Yang, S.; Wang, Y.; Zheng, K.; Li, R.; Lin, L.; Li, M. Probiotic intervention benefits multiple neural behaviors in older adults with mild cognitive impairment. Geriatr. Nurs. 2023, 51, 167–175. [Google Scholar] [CrossRef]
  95. Bonfili, L.; Cecarini, V.; Gogoi, O.; Berardi, S.; Scarpona, S.; Angeletti, M.; Rossi, G.; Eleuteri, A.M. Gut microbiota manipulation through probiotics oral administration restores glucose homeostasis in a mouse model of Alzheimer’s disease. Neurobiol. Aging 2020, 87, 35–43. [Google Scholar] [CrossRef]
  96. Lee, D.-Y.; Shin, Y.-J.; Kim, J.-K.; Jang, H.-M.; Joo, M.-K.; Kim, D.-H. Alleviation of cognitive impairment by gut microbiota lipopolysaccharide production-suppressing Lactobacillus plantarum and Bifidobacterium longum in mice. Food Funct. 2021, 12, 10750–10763. [Google Scholar] [CrossRef] [PubMed]
  97. Mallikarjuna, N.; Praveen, K.; Yellamma, K. Role of Lactobacillus plantarum MTCC1325 in membrane-bound transport ATPases system in Alzheimer’s disease-induced rat brain. BioImpacts 2016, 6, 203–209. [Google Scholar] [CrossRef]
  98. Kaur, H.; Nookala, S.; Singh, S.; Mukundan, S.; Nagamoto-Combs, K.; Combs, C.K. Sex-Dependent Effects of Intestinal Microbiome Manipulation in a Mouse Model of Alzheimer’s Disease. Cells 2021, 10, 2370. [Google Scholar] [CrossRef]
  99. Liao, J.-F.; Cheng, Y.-F.; You, S.-T.; Kuo, W.-C.; Huang, C.-W.; Chiou, J.-J.; Hsu, C.-C.; Hsieh-Li, H.-M.; Wang, S.; Tsai, Y.-C. Lactobacillus plantarum PS128 alleviates neurodegenerative progression in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced mouse models of Parkinson’s disease. Brain Behav. Immun. 2020, 90, 26–46. [Google Scholar] [CrossRef]
  100. Lee, Y.Z.; Cheng, S.-H.; Chang, M.-Y.; Lin, Y.-F.; Wu, C.-C.; Tsai, Y.-C. Neuroprotective Effects of Lactobacillus plantarum PS128 in a Mouse Model of Parkinson’s Disease: The Role of Gut Microbiota and MicroRNAs. Int. J. Mol. Sci. 2023, 24, 6794. [Google Scholar] [CrossRef]
  101. Wang, L.; Li, S.; Jiang, Y.; Zhao, Z.; Shen, Y.; Zhang, J.; Zhao, L. Neuroprotective effect of Lactobacillus plantarum DP189 on MPTP-induced Parkinson’s disease model mice. J. Funct. Foods 2021, 85, 104635. [Google Scholar] [CrossRef]
  102. Perez Visñuk, D.; Savoy de Giori, G.; LeBlanc, J.G.; de Moreno de LeBlanc, A. Neuroprotective effects associated with immune modulation by selected lactic acid bacteria in a Parkinson’s disease model. Nutrition 2020, 79–80, 110995. [Google Scholar] [CrossRef]
  103. Perez Visñuk, D.; del Milagro Teran, M.; Savoy de Giori, G.; LeBlanc, J.G.; de Moreno de LeBlanc, A. Neuroprotective Effect of Riboflavin Producing Lactic Acid Bacteria in Parkinsonian Models. Neurochem. Res. 2022, 47, 1269–1279. [Google Scholar] [CrossRef]
  104. Xu, Y.; Xie, M.; Xue, J.; Xiang, L.; Li, Y.; Xiao, J.; Xiao, G.; Wang, H. EGCG ameliorates neuronal and behavioral defects by remodeling gut microbiota and TotM expression in Drosophila models of Parkinson’s disease. FASEB J. 2020, 34, 5931–5950. [Google Scholar] [CrossRef]
  105. Ilie, O.-D.; Duta, R.; Balmus, I.-M.; Savuca, A.; Petrovici, A.; Nita, I.-B.; Antoci, L.-M.; Jijie, R.; Mihai, C.-T.; Ciobica, A.; et al. Assessing the Neurotoxicity of a Sub-Optimal Dose of Rotenone in Zebrafish (Danio rerio) and the Possible Neuroactive Potential of Valproic Acid, Combination of Levodopa and Carbidopa, and Lactic Acid Bacteria Strains. Antioxidants 2022, 11, 2040. [Google Scholar] [CrossRef]
  106. Mestre, L.; Carrillo-Salinas, F.J.; Feliú, A.; Mecha, M.; Alonso, G.; Espejo, C.; Calvo-Barreiro, L.; Luque-García, J.L.; Estevez, H.; Villar, L.M.; et al. How oral probiotics affect the severity of an experimental model of progressive multiple sclerosis? Bringing commensal bacteria into the neurodegenerative process. Gut Microbes 2020, 12, 1813532. [Google Scholar] [CrossRef]
  107. Shang, X.; Geng, L.; Zhao, Z.; Luo, L.; Shi, X.; Zhang, Q.; Du, R.; Cong, Y.; Xu, W. Transcriptomics reveals the mechanism of selenium-enriched Lactobacillus plantarum alleviating brain oxidative stress under cadmium stress in Luciobarbus capito. Ecotoxicol. Environ. Saf. 2022, 242, 113890. [Google Scholar] [CrossRef]
  108. Xu, Z.; Zhang, J.; Wu, J.; Yang, S.; Li, Y.; Wu, Y.; Li, S.; Zhang, X.; Zuo, W.; Lian, X.; et al. Lactobacillus plantarum ST-III culture supernatant ameliorates alcohol-induced cognitive dysfunction by reducing endoplasmic reticulum stress and oxidative stress. Front. Neurosci. 2022, 16, 976358. [Google Scholar] [CrossRef] [PubMed]
  109. Westfall, S.; Lomis, N.; Prakash, S. Longevity extension in Drosophila through gut-brain communication. Sci. Rep. 2018, 8, 8362. [Google Scholar] [CrossRef]
  110. Zaydi, A.I.; Lew, L.C.; Hor, Y.Y.; Jaafar, M.H.; Chuah, L.O.; Yap, K.P.; Azlan, A.; Azzam, G.; Liong, M.T. Lactobacillus plantarum DR7 improved brain health in aging rats via the serotonin, inflammatory and apoptosis pathways. Benef. Microbes 2020, 11, 753–766. [Google Scholar] [CrossRef] [PubMed]
  111. Xia, Y.; Gong, Y.; Lin, X.; Yang, Y.; Song, X.; Wang, G.; Xiong, Z.; Qian, Y.; Liao, Z.; Zhang, H.; et al. Lactobacillus plantarum AR113 attenuates liver injury in D-galactose-induced aging mice via the inhibition of oxidative stress and endoplasmic reticulum stress. Food Sci. Hum. Wellness 2024, 13, 885–897. [Google Scholar] [CrossRef]
  112. Mirzaei, H.; Sedighi, S.; Kouchaki, E.; Barati, E.; Dadgostar, E.; Aschner, M.; Tamtaji, O.R. Probiotics and the Treatment of Parkinson’s Disease: An Update. Cell. Mol. Neurobiol. 2022, 42, 2449–2457. [Google Scholar] [CrossRef]
  113. Tan, A.H.; Hor, J.W.; Chong, C.W.; Lim, S. Probiotics for Parkinson’s disease: Current evidence and future directions. JGH Open 2021, 5, 414–419. [Google Scholar] [CrossRef]
  114. Metta, V.; Leta, V.; Mrudula, K.R.; Prashanth, L.K.; Goyal, V.; Borgohain, R.; Chung-Faye, G.; Chaudhuri, K.R. Gastrointestinal dysfunction in Parkinson’s disease: Molecular pathology and implications of gut microbiome, probiotics, and fecal microbiota transplantation. J. Neurol. 2022, 269, 1154–1163. [Google Scholar] [CrossRef]
  115. Felice, V.D.; Quigley, E.M.; Sullivan, A.M.; O’Keeffe, G.W.; O’Mahony, S.M. Microbiota-gut-brain signalling in Parkinson’s disease: Implications for non-motor symptoms. Park. Relat. Disord. 2016, 27, 1–8. [Google Scholar] [CrossRef]
  116. Dutta, S.K.; Verma, S.; Jain, V.; Surapaneni, B.K.; Vinayek, R.; Phillips, L.; Nair, P.P. Parkinson’s Disease: The Emerging Role of Gut Dysbiosis, Antibiotics, Probiotics, and Fecal Microbiota Transplantation. J. Neurogastroenterol. Motil. 2019, 25, 363–376. [Google Scholar] [CrossRef]
  117. Chu, C.; Yu, L.; Li, Y.; Guo, H.; Zhai, Q.; Chen, W.; Tian, F. Meta-analysis of randomized controlled trials of the effects of probiotics in Parkinson’s disease. Food Funct. 2023, 14, 3406–3422. [Google Scholar] [CrossRef]
  118. Park, J.M.; Lee, S.C.; Ham, C.; Kim, Y.W. Effect of probiotic supplementation on gastrointestinal motility, inflammation, motor, non-motor symptoms and mental health in Parkinson’s disease: A meta-analysis of randomized controlled trials. Gut Pathog. 2023, 15, 9. [Google Scholar] [CrossRef]
  119. van Wamelen, D.J.; Martinez-Martin, P.; Weintraub, D.; Schrag, A.; Antonini, A.; Falup-Pecurariu, C.; Odin, P.; Ray Chaudhuri, K. The Non-Motor Symptoms Scale in Parkinson’s disease: Validation and use. Acta Neurol. Scand. 2021, 143, 3–12. [Google Scholar] [CrossRef]
  120. Hermanowicz, N.; Jones, S.A.; Hauser, R.A. Impact of non-motor symptoms in Parkinson’s disease: A PMDAlliance survey. Neuropsychiatr. Dis. Treat. 2019, 15, 2205–2212. [Google Scholar] [CrossRef]
  121. Goya, M.E.; Xue, F.; Sampedro-Torres-Quevedo, C.; Arnaouteli, S.; Riquelme-Dominguez, L.; Romanowski, A.; Brydon, J.; Ball, K.L.; Stanley-Wall, N.R.; Doitsidou, M. Probiotic Bacillus subtilis Protects against α-Synuclein Aggregation in C. elegans. Cell Rep. 2020, 30, 367–380.e7. [Google Scholar] [CrossRef]
  122. Liu, X.; Du, Z.R.; Wang, X.; Sun, X.R.; Zhao, Q.; Zhao, F.; Wong, W.T.; Wong, K.H.; Dong, X.-L. Polymannuronic acid prebiotic plus Lacticaseibacillus rhamnosus GG probiotic as a novel synbiotic promoted their separate neuroprotection against Parkinson’s disease. Food Res. Int. 2022, 155, 111067. [Google Scholar] [CrossRef]
  123. Chen, Z.; Zhong, C. Oxidative stress in Alzheimer’s disease. Neurosci. Bull. 2014, 30, 271–281. [Google Scholar] [CrossRef]
  124. Nimgampalle, M.; Kuna, Y. Anti-Alzheimer Properties of Probiotic, Lactobacillus plantarum MTCC 1325 in Alzheimer’s Disease induced Albino Rats. J. Clin. Diagn. Res. JCDR 2017, 11, KC01. [Google Scholar] [CrossRef]
  125. Cekanaviciute, E.; Yoo, B.B.; Runia, T.F.; Debelius, J.W.; Singh, S.; Nelson, C.A.; Kanner, R.; Bencosme, Y.; Lee, Y.K.; Hauser, S.L.; et al. Gut bacteria from multiple sclerosis patients modulate human T cells and exacerbate symptoms in mouse models. Proc. Natl. Acad. Sci. USA 2017, 114, 10713–10718. [Google Scholar] [CrossRef]
  126. Amini, M.E.; Shomali, N.; Bakhshi, A.; Rezaei, S.; Hemmatzadeh, M.; Hosseinzadeh, R.; Eslami, S.; Babaie, F.; Aslani, S.; Torkamandi, S.; et al. Gut microbiome and multiple sclerosis: New insights and perspective. Int. Immunopharmacol. 2020, 88, 107024. [Google Scholar] [CrossRef]
  127. Brown, J.; Quattrochi, B.; Everett, C.; Hong, B.-Y.; Cervantes, J. Gut commensals, dysbiosis, and immune response imbalance in the pathogenesis of multiple sclerosis. Mult. Scler. J. 2021, 27, 807–811. [Google Scholar] [CrossRef]
  128. Pilotto, S.; Zoledziewska, M.; Fenu, G.; Cocco, E.; Lorefice, L. Disease-modifying therapy for multiple sclerosis: Implications for gut microbiota. Mult. Scler. Relat. Disord. 2023, 73, 104671. [Google Scholar] [CrossRef]
  129. Zhang, L.; Liu, C.; Li, D.; Zhao, Y.; Zhang, X.; Zeng, X.; Yang, Z.; Li, S. Antioxidant activity of an exopolysaccharide isolated from Lactobacillus plantarum C88. Int. J. Biol. Macromol. 2013, 54, 270–275. [Google Scholar] [CrossRef]
  130. Gao, D.; Gao, Z.; Zhu, G. Antioxidant effects of Lactobacillus plantarum via activation of transcription factor Nrf2. Food Funct. 2013, 4, 982–989. [Google Scholar] [CrossRef] [PubMed]
  131. Tang, W.; Xing, Z.; Li, C.; Wang, J.; Wang, Y. Molecular mechanisms and in vitro antioxidant effects of Lactobacillus plantarum MA2. Food Chem. 2017, 221, 1642–1649. [Google Scholar] [CrossRef] [PubMed]
  132. Izuddin, W.I.; Humam, A.M.; Loh, T.C.; Foo, H.L.; Samsudin, A.A. Dietary Postbiotic Lactobacillus plantarum Improves Serum and Ruminal Antioxidant Activity and Upregulates Hepatic Antioxidant Enzymes and Ruminal Barrier Function in Post-Weaning Lambs. Antioxidants 2020, 9, 250. [Google Scholar] [CrossRef] [PubMed]
  133. Ge, Q.; Yang, B.; Liu, R.; Jiang, D.; Yu, H.; Wu, M.; Zhang, W. Antioxidant activity of Lactobacillus plantarum NJAU-01 in an animal model of aging. BMC Microbiol. 2021, 21, 182. [Google Scholar] [CrossRef] [PubMed]
  134. Huang, H.J.; Chen, J.L.; Liao, J.F.; Chen, Y.H.; Chieu, M.W.; Ke, Y.Y.; Hsu, C.C.; Tsai, Y.C.; Hsieh-Li, H.M. Lactobacillus plantarum PS128 prevents cognitive dysfunction in Alzheimer’s disease mice by modulating propionic acid levels, glycogen synthase kinase 3 beta activity, and gliosis. BMC Complement. Med Ther. 2021, 21, 259. [Google Scholar] [CrossRef]
  135. Lin, S.-W.; Tsai, Y.-S.; Chen, Y.-L.; Wang, M.-F.; Chen, C.-C.; Lin, W.-H.; Fang, T.J. Lactobacillus plantarum GKM3 Promotes Longevity, Memory Retention, and Reduces Brain Oxidation Stress in SAMP8 Mice. Nutrients 2021, 13, 2860. [Google Scholar] [CrossRef]
  136. Al, K.F.; Craven, L.J.; Gibbons, S.; Parvathy, S.N.; Wing, A.C.; Graf, C.; A Parham, K.; Kerfoot, S.M.; Wilcox, H.; Burton, J.P.; et al. Fecal microbiota transplantation is safe and tolerable in patients with multiple sclerosis: A pilot randomized controlled trial. Mult. Scler. J. Exp. Transl. Clin. 2022, 8, 20552173221086662. [Google Scholar] [CrossRef]
  137. Rosa, F.; Michelotti, T.C.; St-Pierre, B.; Trevisi, E.; Osorio, J.S. Early Life Fecal Microbiota Transplantation in Neonatal Dairy Calves Promotes Growth Performance and Alleviates Inflammation and Oxidative Stress during Weaning. Animals 2021, 11, 2704. [Google Scholar] [CrossRef] [PubMed]
  138. Zhao, Z.; Ning, J.; Bao, X.-Q.; Shang, M.; Ma, J.; Li, G.; Zhang, D. Fecal microbiota transplantation protects rotenone-induced Parkinson’s disease mice via suppressing inflammation mediated by the lipopolysaccharide-TLR4 signaling pathway through the microbiota-gut-brain axis. Microbiome 2021, 9, 226. [Google Scholar] [CrossRef] [PubMed]
  139. Xiang, W.; Xiang, H.; Wang, J.; Jiang, Y.; Pan, C.; Ji, B.; Zhang, A. Fecal microbiota transplantation: A novel strategy for treating Alzheimer’s disease. Front. Microbiol. 2023, 14, 1281233. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Flow diagram of the search and selection process. Twenty (n = 20) studies met the eligibility criteria and investigated the effects of Lactiplantibacillus plantarum or Lactobacillus plantarum alone or in combination on neurodegenerative processes (3 studies in humans and 17 studies in animal models).
Figure 1. Flow diagram of the search and selection process. Twenty (n = 20) studies met the eligibility criteria and investigated the effects of Lactiplantibacillus plantarum or Lactobacillus plantarum alone or in combination on neurodegenerative processes (3 studies in humans and 17 studies in animal models).
Ijms 25 03010 g001
Table 1. Effects of Lactiplantibacillus plantarum, alone or in combination with neurodegenerative diseases (in human and animal model).
Table 1. Effects of Lactiplantibacillus plantarum, alone or in combination with neurodegenerative diseases (in human and animal model).
ProbioticsPopulationMethodologyInterventionResultsReferences
Parkinson’s Disease
HUMAN MODELLactobacillus acidophilus NCIMB 30175, Lactobacillus plantarum NCIMB 30173, Lactobacillus rhamnosus NCIMB 30174 and Enterococcus faecium NCIMB 30176Fecal samples from six donors (three healthy control subjects, three diagnosed with PD)In vitro dynamic, multi-compartment gastrointestinal modelFermentation with probiotic for 48 hIncreased levels of Firmicutes, Actinobacteria and Bacteroidetes in PD patients. Modulation of the intestinal microbiota that delays pathology progression10.1016/j.ijpx.2021.100087[92]
Lactobacillus plantarum PS12825 patientsOpen-label, single-arm, baseline-controlled trialPS128 (2 capsules containing 30 billion colony-forming units per capsule) daily for 12 weeksImproved motor scores and quality of life10.3389/fnut.2021.650053[93]
MCI (Mild Cognitive Impairment)
Lactobacillus plantarum BioF-228, Lactococcus lactis BioF-224, Bifidobacterium lactis CP-9, Lactobacillus rhamnosus Bv-77, Lactobacillus johnsonii MH-68, Lactobacillus paracasei MP137, Lactobacillus salivarius AP-32, Lactobacillus acidophilus TYCA06, Lactococcus lactis LY-66, Bifidobacterium lactis HNO19, Lactobacillus rhamnosus HNO01, Lactobacillus paracasei GL-156, Bifidobacterium animalis BB-115, Lactobacillus casei CS-773, Lactobacillus reuteri TSR332, Lactobacillus fermentum TSF331, Bifidobacterium infantis BLI-02, and Lactobacillus plantarum CN201842 patientsPilot randomized controlled trial (RCT)2 g (>2 × 1010 CFU/g) probiotics daily for 12 weeksCognitive function and sleep quality were improved10.1016/j.gerinurse.2023.03.006[94]
Alzheimer’s Disease
ANIMAL MODELSLAB51: Streptococcus thermophilus DSM 32245, Bifidobacterium lactis DSM 32246, B. lactis DSM 32247, Lactobacillus acidophilus DSM 32241, Lactobacillus helveticus DSM 32242, Lactobacillus paracasei DSM 32243, Lactobacillus plantarum DSM 32244 and Lactobacillus brevis DSM 2796148 AD male mice triple-transgenic, B6; 129-Psen1tm1Mpm Tg (amyloid precursor protein [APP] Swe, tauP301L) 1Lfa/J (named 3xTg-AD) and the wt B6129SF2 miceTreatment with probiotic vs. ShamSLAB51 (200 billion bacteria/kg/d) daily for 16 and 48 weeksIt improved impaired brain glucose metabolism implicated in AD pathogenesis, delaying disease progression10.1016/j.neurobiolaging.2019.11.004[95]
NK151: Lactobacillus plantarum; NK173: Bifidobacterium longum35 specific pathogen-free C57BL/6 mice Treatment with probiotic vs. ShamEscherichiacoli K1 (EC, 1 × 109 CFU per mouse per day) were orally gavage daily for 5 daysReduced memory impairment and modulated gut microbial profile10.1039/d1fo02167b[96]
Lactobacillus plantarum MTCC132548 healthy adult male Wistar ratsThe hippocampal (HP) and cerebral cortex (CC) brain regions of each animal were isolatedLactobacillus plantarum MTCC1325 daily for 60 daysNeurodegeneration was reduced by the protective effect on the ATPase system in the brain10.15171/bi.2016.27[97]
VSL#3 ®: Lactobacillus plantarum, Lactobacillus delbrueckii subsp. Bulgaricus, Lactobacillus paracasei, Lactobacillus acidophilus, Bifidobacterium breve, Bifidobacterium longum, Bifidobacterium infantis and Streptococcus salivarius subsp. Thermophilus140 mice: 70 male and female AppNL-G-F (AD) and 70 male and female C57BL/6J (wild type, WT)Treatment with probiotic vs. Sham vs. ABX (antibiotic) vs. ABX+probioticVSL#3 ® (4 × 109 UFC/día/mice) for 8 weeksAll groups of female mice with intervention improved mnesic ability but none affected memory in male mice; reduced brain levels of TNF-α in female AppNL-G-F mice10.3390/cells10092370[98]
Parkinson’s Disease
Lactobacillus plantarum PS12890 six-week-old male C57BL/6J miceTreatment with probiotic vs. ShamPS128 (109 CFU in 200 μL saline) daily for 28 daysMotor deficits were improved and dopaminergic neuronal death was alleviated. Glial reactivity is reduced and striatal neurotrophic factors are increased10.1016/j.bbi.2020.07.036[99]
Lactobacillus plantarum PS12840 eight-week-old male C57BL/6J miceTreatment with probiotic vs. ShamPS128 (109 CFU/d) (week 1 to 6) and rotenoneModified the microbial profile and maintained neuroprotective effects by increasing the expression of the suppressor of cytokine signaling10.3390/ijms24076794[100]
Lactobacillus plantarum DP18990 male C57BL/6 miceTreatment with probiotic vs. Sham DP189 (0.2 mL) daily for 14 days Improved motor symptoms and increased levels of monoamines in the nervous system. In addition, it promoted neuronal survival10.1016/j.jff.2021.104635[101]
Lactobacillus plantarum CRL 2130, Streptococcus thermophilus CRL 808, and Streptococcus thermophilus CRL 80756 eight-week-old male C57BL/6 mice Treatment with probiotic vs. ShamL. plantarum CRL 2130, S. thermophilus CRL 808, S. thermophilus CRL 807 or the bacterial mixture (100 µL that contain 8 ± 2 × 108 CFU/mL of each strain, individually or as a mixture daily) for 4 daysBoth alone or in combination, the strains produced significantly improved motor skills, with possible involvement of vitamin B interaction, and protective immune response to organ inflammation10.1016/j.nut.2020.110995[102]
Lactobacillus plantarum CRL2130, Lactobacillus plantarum CRL72535 neuro-2a (N2a) neuroblastoma cell line (ATCC CCL131) miceIn vitro: Half of each brain was removed for the determination of cytokines; The other half of the brain was stored in 10% paraformaldehyde/PBS during 24 h and then embedded in paraffin; In vivo: Treatment with probiotic vs. ShamIn vitro: N2a cells were incubated with intracellular extract from L. plantarum CRL2130. In vivo: Lactobacillus plantarum CRL2130, Lactobacillus plantarum CRL725 (100 μL) onceIn vitro: cells-maintained viability and significantly decreased IL-6 release and reactive oxygen species (ROS) formation. In vivo: attenuated motor deficits and prevented dopaminergic neuronal death10.1007/s11064-021-03520-w[103]
Lactobacillus plantarum KJ01, Acetobacter pomorum KJ02, Lactobacillus brevis KJ03, and Acetobacter pasteurianus KJ0430 to 40 adult DrosophilaTreatment with probiotic vs. EGCGLactobacillus plantarum o Acetobacter pomorum (5 g food/1 mL of bacterial suspension) for 20 days (OD600 = 1, approximately 5 × 107 viable cells/mL)LP KJ01 exacerbated PD symptoms and eliminated EGCG (eigallocatechin-3-gallate)-mediated symptom enhancement. In addition, it aggravated neuronal loss and hindered EGCG-mediated enhancement.10.1096/fj.201903125RR[104]
PROBIO: Lactobacillus casei W56, Lactobacillus acidophilus W22, Lactobacillus paracasei W20, Lactobacillus salivarius W24, Lactobacillus lactis W19, Lactobacillus plantarum W62, Bifidobacterium lactis W51, Bifidobacterium lactis W52, and Bifidobacterium bifidum W23120 adult (6–8 months) wild-type AB (WT, genetic line) zebrafishTreatment with probiotic vs. ShamPROBIO (3 g × 28 envelopes) were dissolved directly into 100 mL distilled water as a unique dose per dayROT triggers apoptosis and increases PARKIN and PINK1 expression. LEV/CARB and PROBIO maintain neurogenesis and angiogenesis, exerting neuroprotective functions and decreasing the impact of ROT10.3390/antiox11102040[105]
Multiple Sclerosis
Lactobacillus paracasei DSM 24734, Lactobacillus plantarum DSM 24730, Lactobacillus acidophilus DSM 24735, Lactobacillus delbruckeii subspecies bulgaricus DSM 24734, Bifidobacterium longum DSM 24736, Bifidobacterium infantis DSM 24737, Bifidobacterium breve 24732 and Streptococcus thermophilus DSM 24731Fecal samples freshly obtained from each mouse on day 70 and 85Treatment with probiotic vs. Sham2 times a week: 3 × 108 CFU (100 μL) of Vivomixx for 70 to 85 days Motor function was improved. Increased anti-inflammatory activation in the microglia10.1080/19490976.2020.1813532[106]
Oxidative Stress
Se-enriched Lactobacillus plantarum CCFM8610 270 Luciobarbus capitoTreatment with probiotic vs. ShamDiet of 1–2% body weight twice a day for 30 daysReversed Cd toxicity in the blood and brain, improving antioxidant capacity and reducing memory loss10.1016/j.ecoenv.2022.113890[107]
Lactobacillus plantarum ST-III AB161 (CGMCC 22782)24 male ICR miceTreatment with probiotic vs. ShamFree access for 28 daysSignificantly improved cognitive dysfunction (memory and learning). Modifications in hippocampal morphology and synaptic dysfunction were found, reducing the impact of oxidative stress10.3389/fnins.2022.976358[108]
Lactobacillus plantarum NCIMB 8826 (Lp8826), Lactobacillus fermentum NCIMB 5221 (Lf5221) and Bifidobacteria longum spp. infantis NCIMB 702255 (Bi702255) Wildtype Drosophila melanogaster (Oregon R)Treatment with probiotic vs. synbioticProbiotic formulation: 3.0 × 109 CFU/mL of probiotics with equal distribution between Lp8826 (1.0 × 109 CFU/mL), Lf5221 (1.0 × 109 CFU/mL) and Bi702255 (1.0 × 109 CFU/mL). Synbiotic formulation: probiotic formulation + 0.5% of TFLA powderThe probiotic and synbiotic reduced metabolic stress levels and inflammation, improved oxidative stress and loss of mitochondrial integrity. Synbiotic formulation maintains results better than separate formulation10.1038/s41598-018-25382-z[109]
Cognitive decline and oxidative stress
Lactobacillus plantarum DR76 Male Sprague Dawley ratsTreatment with probiotic vs. ShamDR7 (109 CFU/day) for 12 weeksMemory capacity improved. Anxiety also improved in the DR7-treated group. In addition, protective effects on serotonergic pathways were demonstrated10.3920/BM2019.0200[110]
Neurodegenerative process
Lactobacillus plantarum AR11332 male C57BL/6J miceTreatment with probiotic vs. Sham AR113 (1 × 109 CFU/mL) daily It significantly reduced oxidative stress injury induced by D-galactose, decreasing its cytotoxicity. In addition, cell membrane integrity was maintained and antioxidant enzyme activity was increased10.26599/fshw.2022.9250076[111]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Beltrán-Velasco, A.I.; Reiriz, M.; Uceda, S.; Echeverry-Alzate, V. Lactiplantibacillus (Lactobacillus) plantarum as a Complementary Treatment to Improve Symptomatology in Neurodegenerative Disease: A Systematic Review of Open Access Literature. Int. J. Mol. Sci. 2024, 25, 3010. https://doi.org/10.3390/ijms25053010

AMA Style

Beltrán-Velasco AI, Reiriz M, Uceda S, Echeverry-Alzate V. Lactiplantibacillus (Lactobacillus) plantarum as a Complementary Treatment to Improve Symptomatology in Neurodegenerative Disease: A Systematic Review of Open Access Literature. International Journal of Molecular Sciences. 2024; 25(5):3010. https://doi.org/10.3390/ijms25053010

Chicago/Turabian Style

Beltrán-Velasco, Ana Isabel, Manuel Reiriz, Sara Uceda, and Víctor Echeverry-Alzate. 2024. "Lactiplantibacillus (Lactobacillus) plantarum as a Complementary Treatment to Improve Symptomatology in Neurodegenerative Disease: A Systematic Review of Open Access Literature" International Journal of Molecular Sciences 25, no. 5: 3010. https://doi.org/10.3390/ijms25053010

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

Article Metrics

Back to TopTop