ArticlePDF Available

Abstract

Probiotics are bacteria whose favorable physiological properties have mostly been demonstrated in regard to anxiety and stress. More recent experiments indicate potential use of single- and multi-strain probiotics on motor-related diseases. Initial studies in patients with Parkinson’s disease and Prader-Willi syndrome are concordant with this hypothesis. In experiments with rodents, probiotics improved motor coordination in normal animals and models of Parkinson’s disease, multiple sclerosis, and spinal cord injury as well as grip strength in hepatic encephalopathy. Further studies should delineate the bacterial profile of each condition and their experimental models.
Send Orders for Reprints to reprints@benthamscience.net
Current Neuropharmacology, XXXX, XX, XX-XX 1
REVIEW ARTICLE
1570-159X/XX $65.00+.00 © XXXX Bentham Science Publishers
Probiotic Influences on Motor Skills: A Review
Robert Lalonde1,* and Catherine Strazielle1,2
1Laboratory of Stress, Immunity, Pathogens (EA7300), Medical School, University of Lorraine, 54500, Vandœuvre-les-
Nancy, France; 2CHRU Nancy, Vandœuvre-les-Nancy, France
Abstract: The effects of probiotics have mostly been shown to be favorable on measures of anxiety
and stress. More recent experiments indicate single- and multi-strain probiotics in treating motor-
related diseases. Initial studies in patients with Parkinson’s disease and Prader-Willi syndrome are
concordant with this hypothesis. In addition, probiotics improved motor coordination in normal ani-
mals and models of Parkinson’s disease, multiple sclerosis, and spinal cord injury as well as grip
strength in hepatic encephalopathy. Further studies should delineate the most optimal bacterial profile
under each condition.
A R T I C L E H I S T O R Y
Received: March 21, 2022
Revised: June 23, 2022
Accepted: July 13, 2022
DOI:
00000000000000000000000000000000
Keywords: Lactobacilli, Bifidobacteria, Parkinson’s disease, multiple sclerosis, hepatic encephalopathy, motor coordination.
1. INTRODUCTION
Probiotics are defined as bacteria with favorable physio-
logical properties and are designated as psychobiotics when
they produce behavioral outcomes [1-3], the most important
of which include Lactobacillus (L.) Bifidobacterium (B.),
Streptococcus (S.), Escherichia (E.), Enterococcus (E.), and
Clostridium butyricum [4]. Most researchers in this field
have focused attention on anxiety, stress, and depression via
neurologic, endocrine, humoral, and immune systems [5-8].
The brain is affected in the absence of microbiota in germ-
free animals. Effects on the brain were likewise noted after
antibiotic administration. When bacterial strains, especially
those deemed to be beneficial, were given to animals, neuro-
biological effects occurred, some of which also occurred in
normal human subjects. Such studies were then extended to
treat specific diseases.
The effects of gut microbiota were first evaluated in gas-
trointestinal diseases, cancer, obesity, and infections and
then for neuropsychiatric symptoms [1-8]. In particular, the
treatment of liver disease and its neurological complications
with antibiotics led to an understanding of the role of micro-
biota in other neurological conditions. Since the gut is inner-
vated by the vagus nerve, several experimenters have aimed
at deciphering whether microbiotic actions are transmitted
via this pathway. At the present time, effects on animal and
human behavior have mostly been analyzed for cognition
*Address correspondence to this author at the Laboratory of Stress, Immuni-
ty, Pathogens (EA7300), Medical School, University of Lorraine, 54500,
Vandœuvre-les-Nancy, France; Tel: +33 (3) 83 15 42 56; Fax : +33 (3) 72
74 62 37; E-mail: lalondr54@gmail.com
and emotion, to a lesser extent, sensorimotor functions. In
the present review, we regard probiotic actions on sen-
sorimotor functions under various neurologic conditions.
Although most results are preliminary, this synthesis should
be of use to guide future research.
2. NORMAL SUBJECTS
2.1. Normal or Preterm Human Subjects
Probiotics undergo changes across the lifespan [9], so there
is interest in investigating their effects in children up to old
age. In a preliminary investigation of this kind, Bifidobacte-
rium, Lactobacillus, and Coprococcus strains were more
abundant in 18-month-old infants above the median in the
fine motor skill subscore of the Bayley-III battery of neuro-
development [10]. It remains to be determined whether the
administration of such strains increases motor skills in older
children and adults.
In a double-blind, placebo-controlled, randomized trial,
B. infantis BB-02 96579 + B. lactis BB-12 15954 + S. ther-
mophilus TH-4 15957 had no effect on the Gross Motor
Function Classification System score, the Bayley-III Motor
Composite Scale, or the Movement Assessment Battery for
Children in preterm children at 2-5 years of age [11]. In a
review of five papers, including the preceding one, Upadh-
yay [12] concluded that probiotics had no effect on motor
behaviors in preterm children.
2.2. Normal Animals
L. plantarum + L. longum increased latencies before fall-
ing from the rotorod in 1-month-old mice, either previously
Author Proofs
“For Personal Use Only”
2 Current Neuropharmacology, XXXX, Vol. XX, No. XX Lalonde and Strazielle
exposed to treadmill exercise or not [13]. The probiotic
mixture increased cerebellar concentrations of gamma-
aminobutyric acid (GABA), the cerebellum being particular-
ly relevant in performing this type of task [14]. Likewise, L.
fermentum JDFM216 increased latencies before falling from
the rotorod and decreased foot slips from a stationary beam
in 12-month-old mice [15]. These results lend credence to
the hypothesis that probiotics facilitate motor coordination
and thus may be of use in sporting events [16]. L. plantarum
SPA3 + L. rhamnosus B-8238 counteracted the decrease in
latencies before falling from the rotorod in antibiotic-induced
dysbiosis in mice [17], indicating that individuals with
suboptimal gut bacteria are more likely to improve with pro-
biotics.
3. PARKINSON’S DISEASE
3.1. Patients with Parkinson’s Disease
Most probiotic studies on motor control have been con-
ducted in the context of Parkinson’s disease. Based on gas-
trointestinal anomalies and alterations in the gut microbiota
of such patients [18-22], studies have appeared on probiotic-
induced improvements in patients’ constipation and neural
functions, possibly via anti-inflammatory and antioxidant
actions [23-30]. For example, L. salivarius LS01 and L. aci-
dophilus LA02 DSM 21717 reduced proinflammatory cyto-
kines as well as increased anti-inflammatory cytokines [31].
In an open-label, single-arm, baseline-controlled trial of
patients with Parkinson’s disease, L. plantarum PS128 im-
proved the total score of the Unified Parkinson’s Disease
Rating Scale (UPDRS) and its akinesia subscore as well as a
single index, mobility, and activities of daily living in the 39-
item Parkinson’s Disease Questionnaire (PDQ-39) [32].
Likewise, L. acidophilus + L. fermentum + L. reuteri + B.
bifidum improved the total score of the UPDRS in a double-
blind trial [33].
3.2. Animal Models of Parkinson’s Disease
L. acidophilus + L. plantarum + L. paracasei + L. del-
brueckii subsp. bulgaricus + L. brevis + B. longum + B. breve
+ B. infantis + S. thermophilus, designated as SLAB51, was
given before a unilateral 6-hydroxydopamine (6-OHDA)
injection in the mouse striatum and improved the use of the
contralateral forepaw in the cylinder test and postural asym-
metry in the elevated body swing test [34]. The supplement
also counteracted 6-OHDA-induced decreases in tyrosine
hydroxylase and dopamine transporter immunoreactivity in
the substantia nigra, indicating preservation of dopaminergic
neurons. Likewise, L. salivarius subsp. salicinius AP-32
increased locomotor speed and stride length while decreasing
stance time in gait analyses of rats unilaterally injected with
6-OHDA along the nigro-striatal tract [35]. The probiotic
also increased the number of tyrosine hydroxylase-positive
neurons in the substantia nigra and the striatum. In a similar
manner, L. plantarum PS128 improved step adjustment in
gait analyses of rats unilaterally injected with 6-OHDA
along the nigro-striatal tract and increased tyrosine hydrox-
ylase-positive neurons in the substantia nigra [36]. In addi-
tion, 6-OHDA unilaterally injected in the substantia nigra
followed by intraperitoneally administered apomorphine
augmented contralateral rotations compared with controls
and L. acidophilus + L. reuteri + L. fermentum + B. bifidum
prevented these and increased the number of substantia nigra
neurons destroyed by the neurotoxin [37]. L. fermentum U-
21 alone counteracted the paraquat-induced increase in la-
tencies before descending from a vertical pole and the de-
crease in the number of tyrosine hydroxylase-positive neu-
rons in the substantia nigra [38].
In addition to 6-OHDA, probiotics appear effective in
counteracting anomalies caused by the peripheral injection of
another dopamine neurotoxin, methyl-phenyl-tetrahydro-
pyridine (MPTP). Indeed, L. plantarum CRL 2130 + S.
thermophilus CRL 807 and CRL 808 decreased latencies
before descending from the vertical pole and latencies before
traversing the horizontal stationary beam and increased tyro-
sine hydrolase-positive cell counts in the substantia nigra of
MPTP-injected mice [39]. The same effects were found with
Clostridium butyricum [40]. Moreover, a mixture of L. lactis
MG1363 + glucagon-like peptide-1 (GLP-1) reduced laten-
cies before descending from the vertical pole and increased
open-field activity in mice injected with MPTP as well as
counteracting declines in the number of tyrosine hydrox-
ylase-positive neurons in the substantia nigra [41, 42].
In yet another Parkinsonian model, L. rhamnosis GG + L.
rhamnosus + L. plantarum LP28 + L. lactis subsp. lactis + B.
bifidum + B. longum reduced latencies before traversing the
stationary beam and increased latencies before falling from
the rotorod as well as improving gait patterns in MitoPark
PD mice, characterized by inactivation of the Tfam (tran-
scription factor A) mitochondrial gene in dopamine neurons
[43]. The mixture also added protective actions on substantia
nigra dopamine neurons as determined by the number of
tyrosine hydroxylase-positive cells.
4. MULTIPLE SCLEROSIS
4.1. Patients with Multiple Sclerosis
Motor dysfunctions, sometimes leading to paresis, figure
as important features in the symptomatology of multiple scle-
rosis [44]. Although there is doubt as to whether gut dysbiosis
marks the cause or consequence of the disease, it has been
considered a contributing factor [45-49]. Meta-analyses re-
vealed the beneficial effects of probiotic supplementation on
general health and depression in patients with multiple scle-
rosis [50-52]. In a double-blind trial, L. reuteri + L. casei +
L. plantarum + L. fermentum + B. infantis + B. lactis im-
proved the global score of the Expanded Disability Status
Scale (EDSS) and the General Health Questionnaire-28
(GHQ-28) in patients with multiple sclerosis, the former
featuring the ability to walk [53]. Further experiments must
delineate whether this or other mixtures specifically improve
motor scores.
4.2. Animal Models of Multiple Sclerosis
Antibiotics increased latencies before falling from the
rotorod and improved axon damage in mice exposed to intra-
cranial infection with Theiler’s murine encephalomyelitis
virus (TMEV), a model of multiple sclerosis [54], lending
credence to the hypothesis that gut microbiota influences
demyelinating diseases. In further support of this hypothesis,
L. acidophilus DSM 24735 + L. paracasei DSM 24734 +L.
Author Proofs
“For Personal Use Only”
Probiotic Influences on Motor Skills Current Neuropharmacology, XXXX, Vol. XX, No. XX 3
plantarum DSM 24730 + L. + L. bulgaricus DSM 24734 +
B. infantis DSM 24737 + B. longum DSM 24736 + B. breve
DSM 24732 + S. thermophiles DSM 24732, designated Vi-
vomixx, increased latencies before falling from the rotorod
and exhibited anti-inflammatory actions in TMEV-treated
mice [55]. Both the Vivomixx mixture and a 3-strain mixture
of L. acidophilus LA 201 + L. salivarius LA 304 + B. lactis
LA 304, designated Lactibiane iki, were examined in another
murine model of multiple sclerosis: pertussis toxin-induced
experimental autoimmune encephalomyelitis [56]. As in the
previous study, both probiotic mixtures increased latencies
before falling from the rotorod. On the contrary, neither L.
plantarum nor Bifidobacterium B94 had any effect on
swimming speed in rats exposed to ethidium bromide-induced
demyelination [57]. In a third model of demyelination, the
murine cuprizone-induced model, L. casei T2 augmented
spontaneous alternation rates in a Y-maze [58], a measure of
mental flexibility [59], so it remains to be determined whether
these results can be generalized to motor coordination. More
experiments are needed to determine whether motor signs
should be included in the conclusions of a general review [60]
and a meta-analysis [61] indicating probiotic-induced im-
provements on overall signs, body weight gain, and survival
in experimental autoimmune encephalomyelitis.
5. SPINAL CORD INJURY
5.1. Patients with Spinal Cord Injury
Gut microbiota may be an important factor in mediating
neural repair following spinal cord trauma [62]. In particular,
spinal cord trauma may cause gut dysbiosis [63], so treating
this condition becomes all the more relevant to the patholog-
ical process. Even with minimal actions on sensorimotor
functions, psychobiotics may be of use in mitigating anxiety
and depression [5-7].
5.2. Animal Models of Spinal Cord Injury
A mixture of broad-spectrum antibiotics worsened spinal
cord pathology and slowed down recovery of locomotion in
mice exposed to spinal cord trauma [64]. Like control mice,
antibiotic-treated mice eventually exhibited consistent plantar
stepping but without forelimb/hindlimb coordination and were
more prone to paw anomalies and trunk instability. Converse-
ly, L. casei + L. plantarum + L. acidophilus + L. delbrueckii
subsp. bulgaricus + B. longum + B. breve + B. infantis + S.
salivarius subsp. thermophilus, designated VSL#3, promoted
spinal cord recovery relative to the vehicle-treated group by
increasing the frequency of plantar stepping and fore-
limb/hindlimb coordination along with improving paw posi-
tion and trunk stability. The product also reduced lesion vol-
ume as well as axon and myelin pathology at the injury site.
These data should encourage further analyses in the patterns
of dysbiosis seen after spinal cord injury so that optimal pro-
biotics are proposed.
6. PRADER-WILLI SYNDROME
6.1. Patients with Prader-Willi Syndrome
Prader-Willi syndrome is a disorder caused by the dele-
tion of paternally expressed genes at the Chr. 15q11.2-q13
locus, resulting in hypotonia, developmental delay, hyper-
phagia, obesity, and such neuropsychiatric signs as psychosis
and compulsive behaviors [65]. Gut microbiotas have been
linked to Prader-Willi syndrome [66], so a randomized, dou-
ble-blind, placebo-controlled trial of L. reuteri LR-99 was
conducted [67]. The probiotic improved the total score and
fine motor skill subscore of the ASQ-3 test as well as the
social communication subscore of the Gilliam Autism Rating
Scale (GARS-3) while reducing body mass index.
6.2. Animal Models of Prader-Willi Syndrome
To our knowledge, a probiotic has yet to be examined in
rodent models of Prader-Willi syndrome [68, 69].
7. HEPATIC ENCEPHALOPATHY
7.1. Patients with Hepatic Encephalopathy
Motor performance is impaired even in minimal hepatic
encephalopathy [70]. Neurocognitive scores in patients with
hepatic encephalopathy were correlated with the presence of
bacterial DNA [71] in line with the hypothesis that favorable
bacteria are of use in treating this condition. Indeed, probiot-
ics are useful in treating biochemical anomalies of hepatic
encephalopathy [72-75]. However, their use on human motor
control has been left unexamined.
7.2. Animal Models of Hepatic Encephalopathy
The E. coli Nissle 1917 bacterium was genetically modi-
fied to consume and convert excessive ammonia to arginine
and further modified to synthesize butyrate in rats with ligat-
ed bile ducts [76]. When the engineered product synthesized
arginine + butyrate, forelimb and hind-limb grip strength
improved, but not latencies before falling from the rotorod.
The combination of the antibiotic rifaximin, along with the
Vivomixx probiotic mentioned above, was given in rats with
ligated bile ducts, a model of type C hepatic encephalopathy
[77]. On magnetic resonance spectroscopy scans, duct liga-
tion increased glutamine levels in the hippocampus and cer-
ebellum, effects counteracted by rifaximin + Vivomixx,
whereas the antibiotic alone was without effect. Duct ligation
also decreased creatine levels in the cerebellum, an effect
also counteracted by rifaximin + Vivomixx administration,
while yet again, the antibiotic alone was without effect. De-
spite these favorable biochemical actions, there was no effect
on bile duct ligation-induced hypoactivity in the open field.
Further experimentation is required to determine whether
probiotics specifically improve the sensorimotor impairment
underlying hepatic encephalopathy.
CONCLUSION
Positive results have been obtained on the UPDRS in
patients with Parkinson’s disease given two Lactobacillus
mixtures, data supported by those on three animal models
measuring gait and motor coordination in vertical poles, sta-
tionary beams, and rotorod tests. One positive finding has
been reported with L. reuteri on fine motor skills of the
GARS-3 in Prader-Willi syndrome. But only experiments in
mice or rats exist in regard to multiple sclerosis, spinal cord
injury, and hepatic encephalopathy. In two models of multi-
ple sclerosis, Lactobacillus combined with Bifidobacterium
improved rotorod performance. Another mixture containing
Author Proofs
“For Personal Use Only”
4 Current Neuropharmacology, XXXX, Vol. XX, No. XX Lalonde and Strazielle
Lactobacillus and Bifidobacterium facilitated gait patterns in
spinal cord injury. In hepatic encephalopathy, an improve-
ment in grip strength was noted with E. coli. There is interest
in extending such protocols with other motor-related diseas-
es, such as spinocerebellar atrophy and amyotrophic lateral
sclerosis.
LIST OF ABBREVIATIONS
GLP-1 = Glucagon-like Peptide-1
MPTP = Methyl-phenyl-tetrahydro-pyridine
PDQ-39 = Parkinson’s Disease Questionnaire
TMEV = Theiler’s Murine Encephalomyelitis Virus
6-OHDA = 6-Hydroxydopamine
UPDRS = Unified Parkinson’s Disease Rating Scale
CONSENT FOR PUBLICATION
Not applicable.
FUNDING
Supported by EA7300.
CONFLICT OF INTEREST
The authors declare no conflict of interest, financial or
otherwise.
ACKNOWLEDGEMENTS
Declared none.
REFERENCES
[1] Bermúdez-Humarán, L.G.; Salinas, E.; Ortiz, G.G.; Ramirez-
Jirano, L.J.; Morales, J.A.; Bitzer-Quintero, O.K. From probiotics
to psychobiotics: live beneficial bacteria which act on the brain-gut
axis. Nutrients, 2019, 11(4), 890.
http://dx.doi.org/10.3390/nu11040890 PMID: 31010014
[2] Cryan, J.F.; O’Riordan, K.J.; Cowan, C.S.M.; Sandhu, K.V.; Bas-
tiaanssen, T.F.S.; Boehme, M.; Codagnone, M.G.; Cussotto, S.;
Fulling, C.; Golubeva, A.V.; Guzzetta, K.E.; Jaggar, M.; Long-
Smith, C.M.; Lyte, J.M.; Martin, J.A.; Molinero-Perez, A.; Molo-
ney, G.; Morelli, E.; Morillas, E.; O’Connor, R.; Cruz-Pereira, J.S.;
Peterson, V.L.; Rea, K.; Ritz, N.L.; Sherwin, E.; Spichak, S.;
Teichman, E.M.; van de Wouw, M.; Ventura-Silva, A.P.; Wallace-
Fitzsimons, S.E.; Hyland, N.; Clarke, G.; Dinan, T.G. The microbi-
ota-gut-brain axis. Physiol. Rev., 2019, 99(4), 1877-2013.
http://dx.doi.org/10.1152/physrev.00018.2018 PMID: 31460832
[3] Zagórska, A.; Marcinkowska, M.; Jamrozik, M.; Wiśniowska, B.;
Paśko, P. From probiotics to psychobiotics the gut-brain axis in
psychiatric disorders. Benef. Microbes, 2020, 11(8), 717-732.
http://dx.doi.org/10.3920/BM2020.0063 PMID: 33191776
[4] Sharma, R.; Gupta, D.; Mehrotra, R.; Mago, P. Psychobiotics: the
next-generation probiotics for the brain. Curr. Microbiol., 2021,
78(2), 449-463.
http://dx.doi.org/10.1007/s00284-020-02289-5 PMID: 33394083
[5] Appleton, J. The gut-brain axis: influence of microbiota on mood
and mental health. Integr. Med. (Encinitas), 2018, 17(4), 28-32.
PMID: 31043907
[6] El Dib, R; Periyasamy, AG; de Barros, JL; França, CG; Senefonte,
FL; Vesentini, G Probiotics for the treatment of depression and
anxiety: a systematic review and meta-analysis of randomized con-
trolled trials. Clin Nutr., 2021, 45, 75-90.
http://dx.doi.org/10.1016/j.clnesp.2021.07.027
[7] Long-Smith, C.; O’Riordan, K.J.; Clarke, G.; Stanton, C.; Dinan,
T.G.; Cryan, J.F. Microbiota-gut-brain axis: new therapeutic oppor-
tunities. Annu. Rev. Pharmacol. Toxicol., 2020, 60(1), 477-502.
http://dx.doi.org/10.1146/annurev-pharmtox-010919-023628
PMID: 31506009
[8] Wang, H.; Lee, I.S.; Braun, C.; Enck, P. Effect of probiotics on
central nervous system functions in animals and humans: a system-
atic review. J. Neurogastroenterol. Motil., 2016, 22(4), 589-605.
http://dx.doi.org/10.5056/jnm16018 PMID: 27413138
[9] Borre, Y.E.; O’Keeffe, G.W.; Clarke, G.; Stanton, C.; Dinan, T.G.;
Cryan, J.F. Microbiota and neurodevelopmental windows: implica-
tions for brain disorders. Trends Mol. Med., 2014, 20(9), 509-518.
http://dx.doi.org/10.1016/j.molmed.2014.05.002 PMID: 24956966
[10] Acuña, I.; Cerdó, T.; Ruiz, A.; Torres-Espínola, F.J.; López-
Moreno, A.; Aguilera, M.; Suárez, A.; Campoy, C. Infant gut mi-
crobiota associated with fine motor skills. Nutrients, 2021, 13(5),
1673.
http://dx.doi.org/10.3390/nu13051673 PMID: 34069166
[11] Jacobs, S.E.; Hickey, L.; Donath, S.; Opie, G.F.; Anderson, P.J.;
Garland, S.M.; Cheong, J.L.Y. Probiotics, prematurity and neuro-
development: follow-up of a randomised trial. BMJ Paediatr.
Open, 2017, 1(1), e000176.
http://dx.doi.org/10.1136/bmjpo-2017-000176 PMID: 29637171
[12] Upadhyay, R.P.; Taneja, S.; Chowdhury, R.; Strand, T.A.;
Bhandari, N. Effect of prebiotic and probiotic supplementation on
neurodevelopment in preterm very low birth weight infants: find-
ings from a meta-analysis. Pediatr. Res., 2020, 87(5), 811-822.
http://dx.doi.org/10.1038/s41390-018-0211-9 PMID: 30353041
[13] Kim, J.; Yoon, B.E.; Jeon, Y.K. Effect of treadmill exercise and
probiotic ingestion on motor coordination and brain activity in ado-
lescent mice. Healthcare (Basel), 2020, 9(1), 7.
http://dx.doi.org/10.3390/healthcare9010007 PMID: 33374692
[14] Lalonde, R.; Strazielle, C. Motor performances of spontaneous and
genetically modified mutants with cerebellar atrophy. Cerebellum,
2019, 18(3), 615-634.
http://dx.doi.org/10.1007/s12311-019-01017-5 PMID: 30820866
[15] Park, M.R.; Shin, M.; Mun, D.; Jeong, S.Y.; Jeong, D.Y.; Song,
M.; Ko, G.; Unno, T.; Kim, Y.; Oh, S. Probiotic Lactobacillus fer-
mentum strain JDFM216 improves cognitive behavior and modu-
lates immune response with gut microbiota. Sci. Rep., 2020, 10(1),
21701.
http://dx.doi.org/10.1038/s41598-020-77587-w PMID: 33303803
[16] Leite, G.S.F.; Resende Master Student, A.S.; West, N.P.; Lancha,
A.H., Jr Probiotics and sports: A new magic bullet? Nutrition,
2019, 60, 152-160.
http://dx.doi.org/10.1016/j.nut.2018.09.023 PMID: 30590242
[17] Arslanova, A.; Tarasova, A.; Alexandrova, A.; Novoselova, V.;
Shaidullov, I.; Khusnutdinova, D.; Grigoryeva, T.; Yarullina, D.;
Yakovleva, O.; Sitdikova, G. Protective effects of probiotics on
cognitive and motor functions, anxiety level, visceral sensitivity,
oxidative stress and microbiota in mice with antibiotic-induced
dysbiosis. Life (Basel), 2021, 11(8), 764.
http://dx.doi.org/10.3390/life11080764 PMID: 34440509
[18] Barichella, M.; Pacchetti, C.; Bolliri, C.; Cassani, E.; Iorio, L.;
Pusani, C.; Pinelli, G.; Privitera, G.; Cesari, I.; Faierman, S.A.;
Caccialanza, R.; Pezzoli, G.; Cereda, E. Probiotics and prebiotic fi-
ber for constipation associated with Parkinson disease. Neurology,
2016, 87(12), 1274-1280.
http://dx.doi.org/10.1212/WNL.0000000000003127 PMID:
27543643
[19] Lubomski, M.; Tan, A.H.; Lim, S.Y.; Holmes, A.J.; Davis, R.L.;
Sue, C.M. Parkinson’s disease and the gastrointestinal microbiome.
J. Neurol., 2020, 267(9), 2507-2523.
http://dx.doi.org/10.1007/s00415-019-09320-1 PMID: 31041582
[20] 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 impli-
cations of gut microbiome, probiotics, and fecal microbiota trans-
plantation. J. Neurol., 2022, 269(3), 1154-1163.
http://dx.doi.org/10.1007/s00415-021-10567-w PMID: 33881598
[21] Ramprasad, C.; Douglas, J.Y.; Moshiree, B. Parkinson’s disease
and current treatments for its gastrointestinal neurogastromotility
effects. Curr. Treat. Options Gastroenterol., 2018, 16(4), 489-510.
http://dx.doi.org/10.1007/s11938-018-0201-3 PMID: 30361854
[22] Wallen, Z.D.; Appah, M.; Dean, M.N.; Sesler, C.L.; Factor, S.A.;
Molho, E.; Zabetian, C.P.; Standaert, D.G.; Payami, H. Character-
izing dysbiosis of gut microbiome in PD: evidence for overabun-
Author Proofs
“For Personal Use Only”
Probiotic Influences on Motor Skills Current Neuropharmacology, XXXX, Vol. XX, No. XX 5
dance of opportunistic pathogens. NPJ Parkinsons Dis., 2020, 6(1),
11.
http://dx.doi.org/10.1038/s41531-020-0112-6 PMID: 32566740
[23] Chen, Z.; Ruan, J.; Li, D.; Wang, M.; Han, Z.; Qiu, W.; Wu, G.
The role of intestinal bacteria and gut-brain axis in hepatic enceph-
alopathy. Front. Cell. Infect. Microbiol., 2021, 10, 595759.
http://dx.doi.org/10.3389/fcimb.2020.595759 PMID: 33553004
[24] Gazerani, P. Probiotics for Parkinson’s disease. Int. J. Mol. Sci.,
2019, 20(17), 4121.
http://dx.doi.org/10.3390/ijms20174121 PMID: 31450864
[25] Leta, V.; Ray Chaudhuri, K.; 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.
http://dx.doi.org/10.1016/j.bbi.2021.07.026 PMID: 34364965
[26] Mirzaei, H.; Sedighi, S.; Kouchaki, E.; Barati, E.; Dadgostar, E.;
Aschner, M. Probiotics and the treatment of Parkinson’s disease: an
update. Cell. Mol. Neurobiol., 2021, 2021, 2449-2457.
http://dx.doi.org/10.1007/s10571-021-01128-w PMID: 34283340
[27] Tan, A.H.; Hor, J.W.; Chong, C.W.; Lim, S.Y. Probiotics for Par-
kinson’s disease: Current evidence and future directions. JGH
Open, 2021, 5(4), 414-419.
http://dx.doi.org/10.1002/jgh3.12450 PMID: 33860090
[28] Uyar, G.Ö.; Yildiran, H. A nutritional approach to microbiota in
Parkinson’s disease. Biosci. Microbiota Food Health, 2019, 38(4),
115-127.
http://dx.doi.org/10.12938/bmfh.19-002 PMID: 31763115
[29] Van Laar, T.; Boertien, J.M.; Herranz, A.H. Faecal transplantation,
pro- and prebiotics in Parkinson’s disease; hope or hype? J.
Parkinsons Dis., 2019, 9(s2), S371-S379.
http://dx.doi.org/10.3233/JPD-191802 PMID: 31609702
[30] Wang, Q.; Luo, Y.; Ray Chaudhuri, K.; Reynolds, R.; Tan, E.K.;
Pettersson, S. The role of gut dysbiosis in Parkinson’s disease:
mechanistic insights and therapeutic options. Brain, 2021, 144(9),
2571-2593.
http://dx.doi.org/10.1093/brain/awab156 PMID: 33856024
[31] Magistrelli, L.; Amoruso, A.; Mogna, L.; Graziano, T.; Cantello,
R.; Pane, M.; Comi, C. Probiotics may have beneficial effects in
Parkinson’s disease: in vitro evidence. Front. Immunol., 2019, 10,
969.
http://dx.doi.org/10.3389/fimmu.2019.00969 PMID: 31134068
[32] 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 pa-
tients with Parkinson’s disease: a pilot study. Front. Nutr., 2021, 8,
650053.
http://dx.doi.org/10.3389/fnut.2021.650053 PMID: 34277679
[33] Tamtaji, O.R.; Taghizadeh, M.; Daneshvar Kakhaki, R.; Kouchaki,
E.; Bahmani, F.; Borzabadi, S.; Oryan, S.; Mafi, A.; Asemi, Z.
Clinical and metabolic response to probiotic administration in peo-
ple with Parkinson’s disease: A randomized, double-blind, placebo-
controlled trial. Clin. Nutr., 2019, 38(3), 1031-1035.
http://dx.doi.org/10.1016/j.clnu.2018.05.018 PMID: 29891223
[34] Castelli, V.; d’Angelo, M.; Lombardi, F.; Alfonsetti, M.; Antono-
sante, A.; Catanesi, M.; Benedetti, E.; Palumbo, P.; Cifone, M.G.;
Giordano, A.; Desideri, G.; Cimini, A. Effects of the probiotic for-
mulation SLAB51 in in vitro and in vivo Parkinson’s disease mod-
els. Aging, 2020, 12(5), 4641-4659.
http://dx.doi.org/10.18632/aging.102927 PMID: 32155131
[35] Nurrahma, B.A.; Tsao, S.P.; Wu, C.H.; Yeh, T.H.; Hsieh, P.S.;
Panunggal, B.; Huang, H.Y. Probiotic supplementation facilitates
recovery of 6-OHDA-induced motor deficit via improving mito-
chondrial function and energy metabolism. Front. Aging Neurosci.,
2021, 13, 668775.
http://dx.doi.org/10.3389/fnagi.2021.668775 PMID: 34025392
[36] Ma, Y.F.; Lin, Y.A.; Huang, C.L.; Hsu, C.C.; Wang, S.; Yeh, S.R.
Lactiplantibacillus plantarum PS128 alleviates exaggerated corti-
cal beta oscillations and motor deficits in the 6-hydroxydopamine
rat model of Parkinson’s disease. Probiotics Antimicrob. Proteins,
2021, 2021, 312-325.
http://dx.doi.org/10.1007/s12602-021-09828-x PMID: 34449056
[37] Alipour Nosrani, E.; Tamtaji, O.R.; Alibolandi, Z.; Sarkar, P.;
Ghazanfari, M.; Azami Tameh, A.; Taghizadeh, M.; Banikazemi,
Z.; Hadavi, R.; Naderi Taheri, M. Neuroprotective effects of probi-
otics bacteria on animal model of Parkinson’s disease induced by
6-hydroxydopamine: A behavioral, biochemical, and histological
study. J. Immunoassay Immunochem., 2021, 42(2), 106-120.
http://dx.doi.org/10.1080/15321819.2020.1833917 PMID:
33078659
[38] Marsova, M.; Poluektova, E.; Odorskaya, M.; Ambaryan, A.;
Revishchin, A.; Pavlova, G.; Danilenko, V. Protective effects of
Lactobacillus fermentum U-21 against paraquat-induced oxidative
stress in Caenorhabditis elegans and mouse models. World J. Mi-
crobiol. Biotechnol., 2020, 36(7), 104.
http://dx.doi.org/10.1007/s11274-020-02879-2 PMID: 32632560
[39] Perez Visñuk, D.; Savoy de Giori, G.; LeBlanc, J.G.; de Moreno de
LeBlanc, A. Neuroprotective effects associated with immune mod-
ulation by selected lactic acid bacteria in a Parkinson’s disease
model. Nutrition, 2020, 79-80, 110995.
http://dx.doi.org/10.1016/j.nut.2020.110995 PMID: 32977125
[40] Sun, J.; Li, H.; Jin, Y.; Yu, J.; Mao, S.; Su, K.P.; Ling, Z.; Liu, J.
Probiotic Clostridium butyricum ameliorated motor deficits in a
mouse model of Parkinson’s disease via gut microbiota-GLP-1
pathway. Brain Behav. Immun., 2021, 91, 703-715.
http://dx.doi.org/10.1016/j.bbi.2020.10.014 PMID: 33148438
[41] Fang, X.; Tian, P.; Zhao, X.; Jiang, C.; Chen, T. Neuroprotective
effects of an engineered commensal bacterium in the 1methyl4
phenyl1, 2, 3, 6tetrahydropyridine Parkinson disease mouse mod-
el via producing glucagonlike peptide1. J. Neurochem., 2019,
150(4), 441-452.
http://dx.doi.org/10.1111/jnc.14694 PMID: 30851189
[42] Fang, X.; Zhou, X.; Miao, Y.; Han, Y.; Wei, J.; Chen, T. Therapeu-
tic effect of GLP-1 engineered strain on mice model of Alzheimer’s
disease and Parkinson’s disease. AMB Express, 2020, 10(1), 80.
http://dx.doi.org/10.1186/s13568-020-01014-6 PMID: 32333225
[43] Hsieh, T.H.; Kuo, C.W.; Hsieh, K.H.; Shieh, M.J.; Peng, C.W.;
Chen, Y.C.; Chang, Y.L.; Huang, Y.Z.; Chen, C.C.; Chang, P.K.;
Chen, K.Y.; Chen, H.Y. Probiotics alleviate the progressive deteri-
oration of motor functions in a mouse model of Parkinson’s dis-
ease. Brain Sci., 2020, 10(4), 206.
http://dx.doi.org/10.3390/brainsci10040206 PMID: 32244769
[44] Dutta, R.; Trapp, B.D. Relapsing and progressive forms of multiple
sclerosis. Curr. Opin. Neurol., 2014, 27(3), 271-278.
http://dx.doi.org/10.1097/WCO.0000000000000094 PMID:
24722325
[45] Esmaeil Amini, M.; Shomali, N.; Bakhshi, A.; Rezaei, S.; Hem-
matzadeh, M.; Hosseinzadeh, R.; Eslami, S.; Babaie, F.; Aslani, S.;
Torkamandi, S.; Mohammadi, H. Gut microbiome and multiple
sclerosis: New insights and perspective. Int. Immunopharmacol.,
2020, 88, 107024.
http://dx.doi.org/10.1016/j.intimp.2020.107024 PMID: 33182024
[46] Ghezzi, L.; Cantoni, C.; Pinget, G.V.; Zhou, Y.; Piccio, L. Target-
ing the gut to treat multiple sclerosis. J. Clin. Invest., 2021,
131(13), e143774.
http://dx.doi.org/10.1172/JCI143774 PMID: 34196310
[47] Kohl, H.M.; Castillo, A.R.; Ochoa-Repáraz, J. The microbiome as
a therapeutic target for multiple sclerosis: can genetically engi-
neered probiotics treat the disease? Diseases, 2020, 8(3), 33.
http://dx.doi.org/10.3390/diseases8030033 PMID: 32872621
[48] Parodi, B.; Kerlero de Rosbo, N. The gut-brain axis in multiple
sclerosis: is its dysfunction a pathological trigger or a consequence
of the disease? Front. Immunol., 2021, 12, 718220.
http://dx.doi.org/10.3389/fimmu.2021.718220 PMID: 34621267
[49] Pellizoni, F.P.; Leite, A.Z.; Rodrigues, N.C.; Ubaiz, M.J.; Gonzaga,
M.I.; Takaoka, N.N.C.; Mariano, V.S.; Omori, W.P.; Pinheiro,
D.G.; Matheucci Junior, E.; Gomes, E.; de Oliveira, G.L.V. Detec-
tion of dysbiosis and increased intestinal permeability in Brazilian
patients with relapsing-remitting multiple sclerosis. Int. J. Environ.
Res. Public Health, 2021, 18(9), 4621.
http://dx.doi.org/10.3390/ijerph18094621 PMID: 33925359
[50] Jiang, J.; Chu, C.; Wu, C.; Wang, C.; Zhang, C.; Li, T.; Zhai, Q.;
Yu, L.; Tian, F.; Chen, W. Efficacy of probiotics in multiple scle-
rosis: a systematic review of preclinical trials and meta-analysis of
randomized controlled trials. Food Funct., 2021, 12(6), 2354-2377.
http://dx.doi.org/10.1039/D0FO03203D PMID: 33629669
[51] Mirashrafi, S.; Hejazi Taghanaki, S.Z.; Sarlak, F.; Moravejolahka-
mi, A.R.; Hojjati Kermani, M.A.; Haratian, M. Effect of probiotics
supplementation on disease progression, depression, general health,
Author Proofs
“For Personal Use Only”
6 Current Neuropharmacology, XXXX, Vol. XX, No. XX Lalonde and Strazielle
and anthropometric measurements in relapsingremitting multiple
sclerosis patients: A systematic review and metaanalysis of clini-
cal trials. Int. J. Clin. Pract., 2021, 75(11), e14724.
http://dx.doi.org/10.1111/ijcp.14724 PMID: 34379879
[52] Ullah, H.; Tovchiga, O.; Daglia, M.; Khan, H. Modulating gut
microbiota: an emerging approach in the prevention and treatment
of multiple sclerosis. Curr. Neuropharmacol., 2021, 19(11), 1966-
1983.
http://dx.doi.org/10.2174/18756190MTE02Mjcrz PMID: 33596808
[53] Salami, M.; Kouchaki, E.; Asemi, Z.; Tamtaji, O.R. How probiotic
bacteria influence the motor and mental behaviors as well as im-
munological and oxidative biomarkers in multiple sclerosis? A
double blind clinical trial. J. Funct. Foods, 2019, 52, 8-13.
http://dx.doi.org/10.1016/j.jff.2018.10.023
[54] Mestre, L.; Carrillo-Salinas, F.J.; Mecha, M.; Feliú, A.; Espejo, C.;
Álvarez-Cermeño, J.C.; Villar, L.M.; Guaza, C. Manipulation of
gut microbiota influences immune responses, axon preservation,
and motor disability in a model of progressive multiple sclerosis.
Front. Immunol., 2019, 10, 1374.
http://dx.doi.org/10.3389/fimmu.2019.01374 PMID: 31258540
[55] 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.; Guaza, C. How oral probiotics affect the severity of
an experimental model of progressive multiple sclerosis? Bringing
commensal bacteria into the neurodegenerative process. Gut Mi-
crobes, 2020, 12(1), 1813532.
http://dx.doi.org/10.1080/19490976.2020.1813532 PMID:
32900255
[56] Calvo-Barreiro, L.; Eixarch, H.; Ponce-Alonso, M.; Castillo, M.;
Lebrón-Galán, R.; Mestre, L.; Guaza, C.; Clemente, D.; del Campo,
R.; Montalban, X.; Espejo, C. A commercial probiotic induces
tolerogenic and reduces pathogenic responses in experimental auto-
immune encephalomyelitis. Cells, 2020, 9(4), 906.
http://dx.doi.org/10.3390/cells9040906 PMID: 32272791
[57] Goudarzvand, M.; Rasouli Koohi, S.; Khodaii, Z.; Soleymanzadeh
Moghadam, S. Probiotics Lactobacillus plantarum and bifidobacte-
rium B94: cognitive function in demyelinated model. Med. J. Is-
lam. Repub. Iran, 2016, 30, 391.
PMID: 27579282
[58] Gharehkhani Digehsara, S.; Name, N.; Esfandiari, B.; Karim, E.;
Taheri, S.; Tajabadi-Ebrahimi, M.; Arasteh, J. Effects of Lactoba-
cillus casei strain T2 (IBRC-M10783) on the modulation of
Th17/Treg and evaluation of miR-155, miR-25, and IDO-1 expres-
sion in a cuprizone-induced C57BL/6 mouse model of demye-
lination. Inflammation, 2021, 44(1), 334-343.
http://dx.doi.org/10.1007/s10753-020-01339-1 PMID: 32914363
[59] Lalonde, R. The neurobiological basis of spontaneous alternation.
Neurosci. Biobehav. Rev., 2002, 26(1), 91-104.
http://dx.doi.org/10.1016/S0149-7634(01)00041-0 PMID:
11835987
[60] Blais, L.L.; Montgomery, T.L.; Amiel, E.; Deming, P.B.; Kremen-
tsov, D.N. Probiotic and commensal gut microbial therapies in mul-
tiple sclerosis and its animal models: a comprehensive review. Gut
Microbes, 2021, 13(1), 1943289.
http://dx.doi.org/10.1080/19490976.2021.1943289 PMID:
34264791
[61] Valizadeh, S.; Majdi Seghinsara, A.; Maleki Chollou, K.; Bahadori,
A.; Abbaszadeh, S.; Taghdir, M.; Behniafar, H.; Riahi, S.M. The
efficacy of probiotics in experimental autoimmune encephalomye-
litis (an animal model for MS): a systematic review and meta
analysis. Lett. Appl. Microbiol., 2021, 73(4), 408-417.
http://dx.doi.org/10.1111/lam.13543 PMID: 34310737
[62] Jing, Y.; Bai, F.; Yu, Y. Spinal cord injury and gut microbiota: A
review. Life Sci., 2021, 266, 118865.
http://dx.doi.org/10.1016/j.lfs.2020.118865 PMID: 33301807
[63] Kigerl, K.A.; Mostacada, K.; Popovich, P.G. Gut microbiota are
disease-modifying factors after traumatic spinal cord injury. Neuro-
therapeutics, 2018, 15(1), 60-67.
http://dx.doi.org/10.1007/s13311-017-0583-2 PMID: 29101668
[64] Kigerl, K.A.; Hall, J.C.E.; Wang, L.; Mo, X.; Yu, Z.; Popovich,
P.G. Gut dysbiosis impairs recovery after spinal cord injury. J. Exp.
Med., 2016, 213(12), 2603-2620.
http://dx.doi.org/10.1084/jem.20151345 PMID: 27810921
[65] Cassidy, S.B.; Schwartz, S.; Miller, J.L.; Driscoll, D.J. Prader-Willi
syndrome. Genet. Med., 2012, 14(1), 10-26.
http://dx.doi.org/10.1038/gim.0b013e31822bead0 PMID: 22237428
[66] Olsson, L.M.; Poitou, C.; Tremaroli, V.; Coupaye, M.; Aron-
Wisnewsky, J.; Bäckhed, F.; Clément, K.; Caesar, R. Gut microbio-
ta of obese subjects with Prader-Willi syndrome is linked to meta-
bolic health. Gut, 2020, 69(7), 1229-1238.
http://dx.doi.org/10.1136/gutjnl-2019-319322 PMID: 31611297
[67] Kong, X.J.; Liu, K.; Zhuang, P.; Tian, R.; Liu, S.; Clairmont, C.;
Lin, X.; Sherman, H.; Zhu, J.; Wang, Y.; Fong, M.; Li, A.; Wang,
B.K.; Wang, J.; Yu, Z.; Shen, C.; Cui, X.; Cao, H.; Du, T.; Wan,
G.; Cao, X. The effects of Limosilactobacillus reuteri LR-99 sup-
plementation on body mass index, social communication, fine motor
function, and gut microbiome composition in individuals with Prader-
Willi syndrome: a randomized double-blinded placebo-controlled tri-
al. Probiotics Antimicrob. Proteins, 2021, 13(6), 1508-1520.
http://dx.doi.org/10.1007/s12602-021-09800-9 PMID: 34115318
[68] Bervini, S.; Herzog, H. Mouse models of Prader-Willi Syndrome: A
systematic review. Front. Neuroendocrinol., 2013, 34(2), 107-119.
http://dx.doi.org/10.1016/j.yfrne.2013.01.002 PMID: 23391702
[69] Garfield, A.S.; Davies, J.R.; Burke, L.K.; Furby, H.V.; Wilkinson,
L.S.; Heisler, L.K.; Isles, A.R. Increased alternate splicing of Htr2c
in a mouse model for Prader-Willi syndrome leads disruption of
5HT2C receptor mediated appetite. Mol. Brain, 2016, 9(1), 95.
http://dx.doi.org/10.1186/s13041-016-0277-4 PMID: 27931246
[70] Sharma, P. Minimal hepatic encephalopathy. J. Assoc. Physicians
India, 2009, 57, 760-763.
PMID: 20329443
[71] Moratalla, A.; Ampuero, J.; Bellot, P.; Gallego-Durán, R.; Zapater,
P.; Roger, M.; Figueruela, B.; Martínez-Moreno, B.; González-
Navajas, J.M.; Such, J.; Romero-Gómez, M.; Francés, R. Lactulose
reduces bacterial DNA translocation, which worsens neurocogni-
tive shape in cirrhotic patients with minimal hepatic encephalopa-
thy. Liver Int., 2017, 37(2), 212-223.
http://dx.doi.org/10.1111/liv.13200 PMID: 27388776
[72] Bloom, PP; Tapper, EB; Young, VB; Lok, AS Microbiome therapeu-
tics for hepatic encephalopathy. J Hepatol., 2021, 21, S0168-8278.
http://dx.doi.org/10.1016/j.jhep.2021.08.004
[73] Chen, Z.J.; Liang, C.Y.; Yang, L.Q.; Ren, S.M.; Xia, Y.M.; Cui, L.;
Li, X.F.; Gao, B.L. Association of Parkinson’s disease with mi-
crobes and microbiological therapy. Front. Cell. Infect. Microbiol.,
2021, 11, 619354.
http://dx.doi.org/10.3389/fcimb.2021.619354 PMID: 33763383
[74] Lunia, M.K.; Sharma, B.C.; Sharma, P.; Sachdeva, S.; Srivastava,
S. Probiotics prevent hepatic encephalopathy in patients with cir-
rhosis: a randomized controlled trial. Clin. Gastroenterol. Hepatol.,
2014, 12(6), 1003-1008.e1.
http://dx.doi.org/10.1016/j.cgh.2013.11.006 PMID: 24246768
[75] Maslennikov, R.; Ivashkin, V.; Efremova, I.; Poluektova, E.; Shi-
rokova, E. Probiotics in hepatology: An update. World J. Hepatol.,
2021, 13(9), 1154-1166.
http://dx.doi.org/10.4254/wjh.v13.i9.1154 PMID: 34630882
[76] Ochoa-Sanchez, R.; Oliveira, M.M.; Tremblay, M.; Petrazzo, G.;
Pant, A.; Bosoi, C.R.; Perreault, M.; Querbes, W.; Kurtz, C.B.;
Rose, C.F. Genetically engineered E. coli Nissle attenuates hyper-
ammonemia and prevents memory impairment in bileduct ligated
rats. Liver Int., 2021, 41(5), 1020-1032.
http://dx.doi.org/10.1111/liv.14815 PMID: 33548108
[77] Flatt, E.; McLin, V.A.; Braissant, O.; Pierzchala, K.; Mastro-
marino, P.; Mitrea, S.O.; Sessa, D.; Gruetter, R.; Cudalbu, C. Pro-
biotics combined with rifaximin influence the neurometabolic
changes in a rat model of type C HE. Sci. Rep., 2021, 11(1), 17988.
http://dx.doi.org/10.1038/s41598-021-97018-8 PMID: 34504135
DISCLAIMER: The above article has been published, as is, ahead-of-print, to provide early visibility but is not the final version. Major pub-
lication processes like copyediting, proofing, typesetting and further review are still to be done and may lead to changes in the final published
version, if it is eventually published. All legal disclaimers that apply to the final published article also apply to this ahead-of-print version.
Author Proofs
“For Personal Use Only”
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
The gut-liver axis plays an important role in the pathogenesis of various liver diseases. Probiotics are living bacteria that may be used to correct disorders of this axis. Notable progress has been made in the study of probiotic drugs for the treatment of various liver diseases in the last decade. It has been proven that probiotics are useful for hepatic encephalopathy, but their effects on other symptoms and syndromes of cirrhosis are poorly studied. Their effectiveness in the treatment of metabolic associated fatty liver disease has been shown both in experimental models and in clinical trials, but their effect on the prognosis of this disease has not been described. The beneficial effects of probiotics in alcoholic liver disease have been shown in many experimental studies, but there are very few clinical trials to support these findings. The effects of probiotics on the course of other liver diseases are either poorly studied (such as primary sclerosing cholangitis, chronic hepatitis B and C, and autoimmune hepatitis) or not studied at all (such as primary biliary cholangitis, hepatitis A and E, Wilson's disease, hemochromatosis, storage diseases, and vascular liver diseases). Thus, despite the progress in the study of probiotics in hepatology over the past decade, there are many unexplored and unclear questions surrounding this topic.
Article
Full-text available
A large and expending body of evidence indicates that the gut-brain axis likely plays a crucial role in neurological diseases, including multiple sclerosis (MS). As a whole, the gut-brain axis can be considered as a bi-directional multi-crosstalk pathway that governs the interaction between the gut microbiota and the organism. Perturbation in the commensal microbial population, referred to as dysbiosis, is frequently associated with an increased intestinal permeability, or “leaky gut”, which allows the entrance of exogeneous molecules, in particular bacterial products and metabolites, that can disrupt tissue homeostasis and induce inflammation, promoting both local and systemic immune responses. An altered gut microbiota could therefore have significant repercussions not only on immune responses in the gut but also in distal effector immune sites such as the CNS. Indeed, the dysregulation of this bi-directional communication as a consequence of dysbiosis has been implicated as playing a possible role in the pathogenesis of neurological diseases. In multiple sclerosis (MS), the gut-brain axis is increasingly being considered as playing a crucial role in its pathogenesis, with a major focus on specific gut microbiota alterations associated with the disease. In both MS and its purported murine model, experimental autoimmune encephalomyelitis (EAE), gastrointestinal symptoms and/or an altered gut microbiota have been reported together with increased intestinal permeability. In both EAE and MS, specific components of the microbiota have been shown to modulate both effector and regulatory T-cell responses and therefore disease progression, and EAE experiments with germ-free and specific pathogen-free mice transferred with microbiota associated or not with disease have clearly demonstrated the possible role of the microbiota in disease pathogenesis and/or progression. Here, we review the evidence that can point to two possible consequences of the gut-brain axis dysfunction in MS and EAE: 1. A pro-inflammatory intestinal environment and “leaky” gut induced by dysbiosis could lead to an altered communication with the CNS through the cholinergic afferent fibers, thereby contributing to CNS inflammation and disease pathogenesis; and 2. Neuroinflammation affecting efferent cholinergic transmission could result in intestinal inflammation as disease progresses.
Article
Full-text available
Type C hepatic encephalopathy (HE) is a neuropsychiatric disease caused by chronic liver disease. Management of type C HE remains an important challenge because treatment options are limited. Both the antibiotic rifaximin and probiotics have been reported to reduce the symptoms of HE, but longitudinal studies assessing their effects on brain metabolism are lacking and the molecular mechanisms underpinning their effects are not fully understood. Therefore, we evaluated in detail the effects of these different treatments on the neurometabolic changes associated with type C HE using a multimodal approach including ultra-high field in vivo ¹ H MRS. We analyzed longitudinally the effect of rifaximin alone or in combination with the probiotic Vivomixx on the brain metabolic profile in the hippocampus and cerebellum of bile duct ligated (BDL) rats, an established model of type C HE. Overall, while rifaximin alone appeared to induce no significant effect on the neurometabolic profile of BDL rats, its association with the probiotic resulted in more attenuated neurometabolic alterations in BDL rats followed longitudinally (i.e. a smaller increase in Gln and milder decrease in Glu and Cr levels). Given that both rifaximin and some probiotics are used in the treatment of HE, the implications of these findings may be clinically relevant.
Article
Full-text available
Parkinson’s disease (PD) is a neurodegenerative disorder characterized by midbrain dopaminergic neuronal loss and subsequent physical impairments. Levodopa manages symptoms best, while deep brain stimulation (DBS) is effective for advanced PD patients; however, side effects occur with the diminishing therapeutic window. Recently, Lactiplantibacillus plantarum PS128 (PS128) was found to elevate dopamine levels in rodent brains, suggesting its potential to prevent PD. Here, the therapeutic efficacy of PS128 was examined in the 6-hydroxydopamine rat PD model. Suppression of the power spectral density of beta oscillations (beta PSD) in the primary motor cortex (M1) was recorded as the indicator of disease progression. We found that 6 weeks of daily PS128 supplementation suppressed M1 beta PSD as well as did levodopa and DBS. Long-term normalization of M1 beta PSD was found in PS128-fed rats, whereas levodopa and DBS showed only temporal effects. PS128 + levodopa and PS128 + DBS exhibited better therapeutic effects than did levodopa + DBS or either alone. Significantly improved motor functions in PS128-fed rats were correlated with normalization of M1 beta PSD. Brain tissue analyses further demonstrated the role of PS128 in dopaminergic neuroprotection and the enhanced availability of neurotransmitters. These findings suggest that psychobiotic PS128 might be used alongside conventional therapies to treat PD patients.
Article
Full-text available
Accumulating clinical and preclinical data indicate a prominent role of gut microbiota in regulation of physiological functions. The gut-brain axis imbalance due to gut dysbiosis is associated with a range of neurodegenerative diseases. Probiotics were suggested not only to restore intestinal dysbiosis but also modulate stress response and improve mood and anxiety symptoms. In this study, we assessed the effects of probiotic lactobacilli on behavioral reactions, the level of oxidative stress and microbiota content in mice administered to broad-spectrum antibiotics. Our study demonstrates that antibiotic treatment of adolescent mice for two weeks resulted in higher mortality and lower weight gain and induced significant changes in behavior including lower locomotor and exploratory activity, reduced muscle strength, visceral hypersensitivity, higher level of anxiety and impaired cognitive functions compared to the control group. These changes were accompanied by decreased diversity and total amount of bacteria, abundance of Proteobacteria and Verrucomicrobia phyla, and reduced Firmicutes/Bacteroides ratio in the gut microbiota. Moreover, a higher level of oxidative stress was found in brain and skeletal muscle tissues of mice treated with antibiotics. Oral administration of two Lactobacillus strains prevented the observed changes and improved not only microbiota content but also the behavioral alterations, suggesting a neuroprotective and antioxidant role of probiotics.
Article
Full-text available
There is increasing evidence highlighting the potential role of the gut-brain axis in the pathogenesis of Parkinson’s disease (PD) and on the use of probiotics as a therapeutic strategy for this neurodegenerative disorder. While several studies have been published on the topic in recent years, there is still a lack of a comprehensive understanding of the effects of probiotics in PD and their possible underlying mechanisms. Through this systematic review, we collected a total of 17 articles, consisting of preclinical and clinical models of PD investigating the effect of probiotics on (1) energy metabolism, (2) inflammation and oxidative stress, (3) neurodegeneration, as well as (4) motor and (5) non-motor function. Articles were obtained from PubMed/Medline, Scopus, Web of Science and EMBASE databases. Findings from preclinical studies suggest that treatment with probiotics increased glucose metabolism (increased secretion of glucagon-like peptide-1), reduced peripheral and central inflammation (reduced interleukin-6 and tumor necrosis factor-α (TNF-α)), reduced peripheral and central oxidative stress (reduced peripheral superoxide anion levels and increased central antioxidant glutathione levels), decreased neurodegeneration (increased numbers of tyrosine hydroxylase dopaminergic neurons and levels of brain-derived neurotrophic factor), increased motor function (increased motor agility) and non-motor function (decreased memory deficits). Similarly, findings from clinical studies suggest that probiotics increased glucose metabolism (reduced insulin resistance), reduced peripheral inflammation (reduced peripheral TNF-α expression and C-reactive protein levels), and increased motor and non-motor function (decreased overall PD symptomatology and constipation); however, findings on oxidative stress were inconclusive across studies. Overall, this review is the first one to systematically report evidence for the putative beneficial effects of probiotics on molecular and cellular mechanisms, as well as behavioural phenotypes, in either preclinical or clinical studies in PD. However, additional and more robust studies are still needed to confirm these outcomes, and should aim to focus more on bench-to-bedside approaches, in order to address the existing gaps between preclinical and clinical findings in this field.
Article
Hepatic encephalopathy (HE) is a complication of cirrhosis characterized by neuropsychiatric and motor dysfunction. Microbiota-host interactions have an important role in HE pathogenesis. Therapies targeting microbial community composition and function have been explored for the treatment of HE. Prebiotics, probiotics and fecal microbiota transplant (FMT) have aimed to increase the abundance of potentially beneficial taxa, while antibiotics have aimed to decrease the abundance of potentially harmful taxa. Other microbiome therapeutics, including postbiotics and absorbents, have been used to target microbial products. Microbiome-targeted therapies for HE have had some success, notably lactulose and rifaximin, with early promise for probiotics and FMT. Microbiome therapeutics face several challenges in HE, including the resilience of the microbiome to sustainable change and unpredictable clinical outcomes from microbiota alterations. Future work in this space should focus on rigorous trial design, microbiome therapy selection, and a personalized approach to HE.
Article
Background Probiotics may have a promising role in chronic autoinflammatory diseases. The current systematic review and meta-analysis investigated the effects of probiotics on disease progression, depression, general health and anthropometric measurements in Relapsing-Remitting Multiple Sclerosis (RRMS) patients. Methods The English literature search was performed using PubMed, Scopus, Web of Science, and the Central Cochrane Library through January 2021. Random effect models were used to synthesize quantitative data by STATA14. Results From a total of 152 identified entries, four trials were included in quantitative synthesis (n=213; 106 as intervention, 107 as control). An additional six studies with the same structure and different markers were also systematically reviewed. The pooled effect size showed that Expanded Disability Status Scale (EDSS) (WMD=-0.43; 95% CI=-0.65, -0.20; P<0.001), Beck Depression Inventory-Ⅱ (BDI-Ⅱ) (WMD=-3.22; 95% CI=-4.38, -2.06; P<0.001) and General Health Questionnaire (GHQ) (WMD=-4.37; 95% CI=-6.43, -2.31; P<0.001) were improved following probiotics supplementation. However, body weight and body mass index did not statistically change. Conclusion Our findings revealed that probiotics supplementation can improve disease progression, suppress depression, and general health in MS patients; although, further investigations may be needed.
Article
Background & aim Probiotics in the gut have been suggested to have a beneficial effect on anxiety response and depressive disorder. Hence we conducted a systematic review and meta-analysis to summarize the effects of probiotics associated with or without pharmacological or psychological therapies in patients with depressive and/or anxiety symptoms or disorders. Methods We conducted searches of MEDLINE, EMBASE, CENTRAL, PsycINFO, CINAHL, ProQuest, LILACS, and Web of Science up to February 2020 to identify randomized controlled trials (RCTs) investigating the efficacy of probiotics associated with or without pharmacological or psychological therapies for patient-important outcomes including relief of depressive, anxiety and stress symptoms, cognitive functions, adverse events and quality of life. We used the GRADE approach to rate the overall certainty of the evidence by outcome. The protocol of the systematic review was registered with PROPSERO and published under the number CRD4202016329. Results 16 RCTs including 1,125 patients proved eligible. Results suggested a significant improvement in using Beck Depression Index (MD, −3.20 [95% CI, −5.91 to −0.49], p = 0.02; I² = 21%, p = 0.28) for depression symptoms and State-Trait Anxiety Inventory (STAI) (MD, −6.88 [95% CI, −12.35 to −1.41], p = 0.01; I² = 24%, p = 0.25) for anxiety with overall certainty in evidence rated as moderate and low, respectively. However, Depression Scale (DASS-Depression) (MD, 2.01 [95% CI, −0.80 to 4.82], p = 0.16; I² = 0%, p = 0.62), Montgomery-Asberg Depression Rating Scale (MADRAS) (MD, −2,41 [95% CI, −10,55 to 5,72], p = 0,56; I² = 87%, p = 0,006), Anxiety scale (DASS-Anxiety) (MD, 0.49 [95% CI, −4.05 to 5.02], p = 0.83; I² = 74%, p = 0.05), and Stress Scale (DASS-Stress) (MD, 0.84 [95% CI, −2.64 to 4.33], p = 0.64; I² = 34%, p = 0.22) showed no significant decrease in the relief of depression, anxiety and stress symptoms of probiotics compared to placebo with overall certainty in evidence rated as very low for all outcomes. We also found no differences in the Beck Anxiety Index (BAI) (MD, −3.21 [95% CI, −6.50 to 0.08], p = 0.06; I² = 0%, p = 0.88) with overall certainty in evidence rated as low. Results suggested a non-statistically significantly effect of probiotics in the adverse events outcomes. Conclusions The current review suggests that probiotics may improve symptoms of depression and anxiety in clinical patients. However, given the limitations in the included studies, RCTs with long-term follow-up and large sample sizes are needed.