Download as pdf or txt
Download as pdf or txt
You are on page 1of 25

Neurochemical Research

https://doi.org/10.1007/s11064-023-04084-7

REVIEW

Gut‑Brain Axis Modulation of Metabolic Disorders: Exploring


the Intertwined Neurohumoral Pathways and Therapeutic Prospects
Diya Bhalla1 · Susha Dinesh2 · Sameer Sharma2 · Gonchigar Jayanna Sathisha3

Received: 29 August 2023 / Revised: 6 December 2023 / Accepted: 8 December 2023


© The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature 2024

Abstract
A significant rise in metabolic disorders, frequently brought on by lifestyle choices, is alarming. A wide range of preliminary
studies indicates the significance of the gut-brain axis, which regulates bidirectional signaling between the gastrointestinal
tract and the cognitive system, and is crucial for regulating host metabolism and cognition. Intimate connections between
the brain and the gastrointestinal tract provide a network of neurohumoral transmission that can transmit in both directions.
The gut-brain axis successfully establishes that the wellness of the brain is always correlated with the extent to which the
gut operates. Research on the gut-brain axis has historically concentrated on how psychological health affects how well the
gastrointestinal system works. The latest studies, however, revealed that the gut microbiota interacts with the brain via the
gut-brain axis to control phenotypic changes in the brain and in behavior. This study addresses the significance of the gut
microbiota, the role of the gut-brain axis in management of various metabolic disorders, the hormonal and neural signal-
ing pathways and the therapeutic treatments available. Its objective is to establish the significance of the gut-brain axis
in metabolic disorders accurately and examine the link between the two while evaluating the therapeutic strategies to be
incorporated in the future.

* Sameer Sharma
sameer@bionome.in
1
Faculty of Life and Allied Health Sciences, MS Ramaiah
University of Applied Science, Bangalore 560048, India
2
Department of Bioinformatics, BioNome, Bangalore 560043,
India
3
Department of Post Graduate Studies and Research
in Biochemistry, Jnanasahyadri, Kuvempu University,
Shankaraghatta, Shimoga 577451, India

Vol.:(0123456789)
Neurochemical Research

Graphical Abstract

Deciphering the intricate communication of the GBA reveals its pivotal role in regulating metabolic disorders and cogni-
tion. This exploration underscores the dynamic interplay between gut microbiota, neurohumoral signaling, and potential
therapeutic interventions, highlighting new avenues for holistic metabolic management.

Keywords Gut-brain axis · Metabolic disorders · Gut microbiota · Signaling pathways · GBA

Abbreviations GLP Glucagon-like peptide 1


ANS Autonomous nervous system GM Gut microbiota
BMI Body mass index HFD High-fat diet
CNS Central nervous system HM Humanized
ENS Enteric nervous system IBS Irritabe bowel syndrome
FMT Faecal microbiota transplantation MeS Metabolic syndrome
GBA Gut-brain axis PNS Peripheral nervous system
GF Germ-free PYY Peptide YY
GHS-R Growth hormone-secretagogue receptor SCFAs Short chain fatty acid
GI Gastrointestinal T2DM Type 2 diabetes
Neurochemical Research

Introduction biochemical processes, such as satiation, dietary intake,


regulation of glucose, fat and lipid metabolism and insu-
The 'gut-brain' or 'brain-gut axis', is a bidirectional commu- lin secretion [1]. Although the hypothesis of GBA initially
nication system consisting of humoral pathways, including focused pn the regulation of cholecystokinin secretion, it has
cytokines, hormones, and neuropeptides as signalling mol- subsequently been expanded to encompass all interactions
ecules, along with neural networks like the vagus, sympa- between the central nervous system (CNS) and the gastroin-
thetic, spinal nerves and the enteric nervous system (ENS) testinal (GI) tract. CNS functioning has been correlated with
[1]. The gut-brain axis (GBA), commonly referred to as the the healthy functioning of the gut. It has been determined
central nervous system-gut microbiota (GM) interaction that neurotransmitters, hormones, and immune-regulating
pathway, has recently drawn a lot of focus. The intricate substances secreted from the gut interact with the CNS via
community of GM contributes to preserving a dynamic met- direct contact or through the autonomous nervous system
abolic biological equilibrium. An adult's body has approx- (ANS) [1, 2]. The novel research conducted by Sudo and
imately 100 trillion bacteria, 80% of which reside in the colleagues who discovered the altered reaction to stress in
gut and are around ten times more prolific than all of the germ-free (GF) mice established evidence for the presence
body's cells [1]. More than 100 species of bacteria with over of the GBA [2]. Besides establishing its existence, further
150 times more genes than the human genome have been investigations involving GF mice further strengthened the
identified in the gut microbiome. The microbiota typically hypothesis that the GBA expands these two systems and
includes microorganisms that thrive in a particular habitat extends into the endocrine, neural, and immune systems [3].
whereas the microbiome is composed of genomes from the A recent comprehensive analysis by Mayer et al. examined
microbes found in the surroundings. The human microbi- the neurological components of GBA interactions as well as
ome has been reported to consist of about 5000 strains of the purpose of centrally-driven networks [3].
bacteria and over 1000 different types of microflora. Bac- Both the CNS and the peripheral nervous system (PNS)
teroidetes and Firmicutes are the two bacterial phylotypes are impacted by the interactions between these endocrine
that primarily constitute the microbiome; Proteobacteria, and neural networks. The hormones employ specialized
Actinomyces, Fusobacterium, and Verrucomicrobia are neural receptors for stimulation in order to converse with
present in relatively modest levels. Except for the recogni- the CNS about the functioning of the hormone's producing
tion that pathogenic microorganisms in the gut can cross organs. The ANS and ENS, the neuroendocrine system, the
the blood–brain barrier and impact the brain, the effect of metabolic system, and the immunological system are some
GM on the brain was barely recognized by people initially. of the acknowledged avenues of gut-brain interaction [3].
On the other hand, a public health emergency brought to In recent years, an increasing variety of scientific evidence
light the potential connection between the brain and GM. from research on the gut microbiome in animal models has
When a flood struck the Canadian municipality of Walker- led to a reexamination of how the CNS and PNS interact.
ton in 2000, E. coli and C. jejuni contaminated the drinking Considering abundant current research demonstrating that
water. Of the 4561 individuals who contracted the infection, the inhabitants of gut bacteria are essential for CNS func-
2451 underwent a follow-up evaluation eight years later, and tioning, neuroscientists are paying attention to such distinc-
1166 received an Irritable Bowel Syndrome (IBS) diagnosis tive findings which directly emphasize the "bottom-up"
[1, 2]. Both depression and anxiety were discovered to be impact of microorganisms [1].
separate risk factors for ongoing IBS in these IBS patients. Over the past few decades, metabolic disorders MDs)
The precise process by which the GM and the brain com- have grown significantly in prevalence across the globe
municate is yet to be thoroughly elucidated. In general, the (Table 1). According to statistics from the World Health
neurological system (the gut-brain neuroanatomical path- Organization, 300 million individuals globally are medically
way) is not the only way that GM affects the brain; other obese, which is characterized by possessing a body mass index
pathways include the endocrine, immunological, and meta- (BMI) of at least 30 kg/m2 [4]. Particularly worrisome is the
bolic systems. The gut microbiota‑gut‑brain axis (henceforth similarly frightening increase in childhood obesity. Alarmingly
referred to as the gut microbiota‑brain axis) is the interaction elevated incidences of type 2 diabetes (T2DM), atherosclero-
of GM with GBA. Emphasis is placed on the role of bac- sis, resistance to insulin, and other illnesses prevail. Therefore,
teria in the gut microbiota-brain axis with the capability to it is essential to evaluate all potential causes of these MDs.
actively alter GM content and so employ it as an independ- More and more data indicate the pivotal role of GM in regulat-
ent variable. ing the GBA. The link between GBA and MDs cannot be dis-
Both the brain and the gut inhabit the identical neural regarded as evidenced by previous research studies. Therefore,
crest during development, extensively coordinating and it is crucial to analyze the GBA and its role in MDs.
influencing one another. The GBA is involved in numerous This review highlights the translational study areas that
are essential for assessing the function of the GBA in MDs,
Neurochemical Research

strategically assessing existing preclinical literature and (Fig. 1). Animal model investigations served as a basis for
investigating currently available data from such studies. the initial studies on the effects of GM diversity and behav-
iour on metabolic diseases, particularly obesity. Despite
consuming 29% more food than conventional mice, it was
Gut Microbiota and Metabolic Disorders found that germ-free mice—especially those who were bred
without any microbes—have a lower quantity of fat [19,
Gut Microbiota's Influence on Metabolic Health 20]. Furthermore, Bäckhed et al. discovered that adult GF
and Disease mice with lower food intake experienced a 60% spike rise
in body fat composition and insulin resistance over 14 days
The functioning of GM and its impact on human health have of being colonized with GM from conventionally grown
attracted more attention in recent years, especially in relation animals. It has been observed that mice fed a high-fat diet
to metabolic diseases which boost the risk of cardiovascular (HFD) with the obesity-resistance trait consume fewer calo-
diseases and, as a result, the death rate. Hippocrates report- ries and excrete more lipids than normal mice fed an HFD
edly stated that "all diseases originate in the gut [18]." There [20]. Investigation into the basis of these events revealed
have been abundant studies corroborating this hypothesis that the GM makeup of obese and lean mice is distinct. The
over the last few decades. Although bacteria and fungi from microbiota of obese mice differs from lean mice due to a
the phyla Firmicutes and Bacteriodetes predominate, the higher concentration of Firmicutes and a lower prevalence of
human colon contains trillions of diverse microorganisms Bacterioidetes. As reported by Riva et al. obesity was linked
[18]. Lately, studies have been centred on how the immune to higher concentrations of Firmicutes, including Rumino-
system, brain development, and behaviour are impacted by coccaceae, and lower concentrations of Bacteroidetes, like
GM. It is plausible that intestinal microbes interact with the Bacteroidaceae and Bacteroides [21]. As per a Danish study
immune system and gut epithelial cells and have a role in the consisting of 169 obese and 123 non-obese subjects, people
emergence of an array of metabolic diseases. with lower microbial gene counts (LCG) have poorer inflam-
According to Zimmer, a normal human harbours 100 matory states, obesity, diabetes, insulin resistance, adiposity,
trillion microorganisms (referred to as microbiota or micro- and dyslipidemia [22].
flora) in their body [18]. The word "microbiota" refers to
the population of microbes that inhabit the human body's Dysbiosis and Its Association with Metabolic
multiple biological systems, such as the GM and the skin. Disorders
The gut contains 1­ 014 bacteria, which is ten times more than
the entirety of human cells [18]. Additionally, the DNA An alteration in the ratio of advantageous to harmful micro-
sequences of these microbes have 150 times more genes organisms, a shift in the distribution of intestinal bacteria,
than that of humans. The cellular makeup of the intesti- and a surge in the number of small bowel microbes can all
nal mucosa has been guarded and fostered by commensal be indicators of dysbiosis, a microbial imbalance in the gut.
gut microbes since the evolution of human existence. As Essentially, a number of factors, such as microbial metabo-
a result, GM is widely acknowledged as a crucial control- lites, microbial interactions, the immune response of the
ler of human physiology and pathophysiology and indis- host, host physiology, nutrition, and host environment, con-
putably performs an active role in both health and illness. tribute to the development of the disease. The human GI
MDs including obesity, diabetes, and eating disorders, in tract reportedly comprises more than fifty distinct bacterial
addition to stress-associated neuropsychological disorders phyla, and the GM composition and prevalence rates vary
such as depression and anxiety, are typically identified by among the diverse microbial habitats of the GI tract. In gen-
fluctuations in eating behaviour, all including variations in eral, distinct microbial comunities dwell in distinct GI tract
the human gut flora. Obesity, described as a chronic buildup segments [23]. Strict anaerobes constitute about 95% of the
of lipids in fatty tissues, is one of the current primary health microbial species that live in the colon; this is influenced
issues [19]. There are numerous causes that lead to obesity, by the nutrients present there. Intrinsic and extrinsic factors
but the most common one is an imbalance between energy considerably influence the development of bacterial growth
intake from food and beverages and energy expenditure. in the small intestine. The most prominent intrinsic factors
There is mounting data that indicates the dysregulated GM hindering this proliferation of bacteria are secretion of gas-
may be the cause of the imbalance. The GM serves a sub- tric juice and bile acid, peristaltic movements, typical gut
stantial role in several mechanisms that are associated with defence mechanisms, mucin fabrication, gut peptides with
the development of obesity, involving nutrient digestion and antimicrobial properties, and mitigation of pathogenic bacte-
utilization, energetic homeostasis, retainment of the rigidity ria retrograde translocation from the lower parts to the upper
of the epithelium of the intestine, carbohydrate, lipid, and parts of the intestine through the ileocecal valve. Diet, bac-
bile acid metabolism, and regulation of intestinal processes terial and viral infections, prokinetic pharmaceuticals that
Neurochemical Research

Table 1  Metabolic diseases: enzyme deficiency, symptoms and treatment


Metabolic disorders Enzyme deficiency Symptoms Treatments References

Carbohydrate metabolic disorders


T2DM Intestinal Alkaline Phosphatase Abdominal obesity Metformin tablets [5, 6]
Nocturia
Blurry Vision
Galactosemia Galactose 1 P uridyl transferase Jaundice Agalsidase injection [7]
Diarrhoea
Liver failure
Acid Mucopolysaccharides Glycosaminoglycans Hepatomegaly Enzyme replacement therapy and [8]
Mental Retardation Hematopoietic stem cell transplanta-
tion
Glycogen storage disorders Glycogen metabolism enzymes Liver failure Protein Supplementation and Corn- [9]
Heart failure starch
Schindler Disease Alpha-N-acetylgalactosaminidase Cardiomyopathy No specific therapy [10]
Hepatomegaly
Lipid metabolic disorders
Tay- Sachs Hexosaminidase-A Mental Retardation No specific treatment [11]
Muscle Stiffness
Gangliosidoses Beta-galactosidase-1 Visual Impairment Gene Therapy [11, 12]
Hepatosplenomegaly Enzyme Replacement Therapy
Gaucher Disease Glucocerebrosidase Anaemia Blood Transfusion [12]
Eye movement disorders Enzyme Replacement Therapy
Niemann—Pick Acid sphingomyelinase deficiency Neural degeneration No cure available [13]
Recurrent Pneumonia
Amino acid metabolic disorders
Hyperglycemia Glycine Cleavage enzyme Ochronosis Insulin therapy [14]
Frequent urination Fluid Replacement
Tyrosinemia Fumarylacetoacetate Hydrolase Liver failure Nitisinone tablets [14]
Photophobia
Maple syrup Urine Disease Alpha-keto acid dehydrogenase Delayed Development Low protein diet [15]
Abnormal movements
Urea cycle disorders
Citrullinemia Arginosuccinate Lyase Hyperactivity Liver Transplantation [16]
Arginosuccinate synthase
Ornithine Carbomyl transferase
Hyperammonemia Arginase Ataxia Dialysis and administration of Neo- [16, 17]
Behaviour Changes mycin and Metronidazole tablets

affect motility, and medications that affect the GM, such as of Firmicutes and Clostridia members than control partici-
H2 blockers, pre-and probiotics, proton pump inhibitors, and pants) [22, 23]. Although earlier research has linked obe-
antibiotics, are examples of extrinsic variables. Commensal sity and metabolic disorders to alterations in the makeup
and pathogenic GM colonize disproportionately and result and variety of the GM, no reliable indicators of these det-
in dysbiosis if one or more external events upset the delicate rimental health conditions are currently established in the
equilibrium that numerous protective mechanisms have been gut microbial communities. This could also be because the
designed to protect [23]. It is well established that dysbiosis implications sizes are smaller than in other conditions that
is linked to both obesity and reduced glucose level toler- are more clearly linked to the GM, like bowel inflammation
ance observed in individuals with diabetes and pre-diabetes. [23]. However, in order to direct us towards more effective
According to certain studies, the microbiota of obese and methods of disease diagnosis, treatment, and prevention, a
overweight people differs from that of lean people in terms better knowledge of the role played by GM in the onset and
of the proportions of Bacteroidetes members and Firmicutes progression of obesity and metabolic disorders is of para-
[23]. However, there have also been contradictory or inde- mount importance [23]. The gut microbiome shifts as people
terminate results [23]. Among other characteristics, blood age from adolescence to elderly years, becoming less diverse
glucose levels were positively linked with the Bacteroidetes and more variable among individuals.
to Firmicutes ratio, and people with T2DM had lower ratios
Neurochemical Research

Fig. 1  Metabolic disorders; causes, symptoms and treatment methods

Influence of Gut Microbiota on Energy Metabolism food than GF mice, they still had roughly 40% lower total
and Nutrient Absorption body fat [24]. The GM could directly affect the CNS by
altering the GBA's endocrine signaling pathways, such as
GM can obtain energy from food and regulate fat accumu- glucagon-like peptide-1 (GLP-1) and peptide YY signaling,
lation in the body. The GM has a significant role in mul- or by triggering reward pathways [24, 25]. An optimally
tiple physiological processes within the gut, including the functioning metabolism and homoeostasis are dependent
body's pathogen defence, intestinal microvilli growth, and upon a healthy GM, which considerably assists the host in
non-digestible polysaccharide breakdown. As a result, the recompense for dwelling and thriving in the gut ecosystem.
GM uses food components that the host swallows but can- The association between GM and human physiology is not
not digest to generate energy for the host. Several metabolic solely commensal; but actually, a mutualistic relationship,
disorders linked to the regulation of glucose levels and the as evidenced by statistical analysis of the alterations in the
emergence of cardiovascular diseases are associated with genetic makeup of GM and accompanying clinical investiga-
obesity. In spite of substantial immediate shifts in energy tions. It is now apparent that the host's GM notably impacts
expenditure, body mass and adiposity remain relatively sta- the host's physiology and phenotypes such as intake of nutri-
ble, while the level of quality and quantity content of dietary ents and well-being, obesity, diabetes, metabolic syndrome
intake varies greatly from every meal every day. Most people (MeS), behavior, and numerous neurological conditions
can accurately calculate their cumulative calorie consump- [26].
tion and expenditure when tracking their dietary habits and
exercise over a period of time comprising at least several
meals. Energy homeostasis is a constantly evolving mech- Gut‑Derived Signaling Molecules
anism that maintains an equilibrium of metabolic energy
accumulated as fat [24]. Therefore, every system regulating Short Chain Fatty Acids (SCFAs) and Effects
the intake of calories and their following harvesting ought on Metabolism and Appetite Regulation
to be involved in maintaining an equilibrium of body mass.
Numerous recently published studies conducted by the J. The three most extensively studied SCFAs are butyrate,
Gordon group have demonstrated the role that the genetic propionate, and acetate. They are the primary fermenta-
makeup of the intestinal microbes serves in the administra- tion metabolites of the gut microbial breakdown of dietary
tion of energy equilibrium [24]. Backhed et al. discovered fibre, contributing approximately 10% of the host's over-
that while mice having a normal GM consumed 30% less all energy needs. Additionally, the GM produces butyrate,
Neurochemical Research

propionate, and acetate, capable of binding to G-protein- regulation of metabolism. In mice and human patients,
coupled receptors such as GPR43 and GPR41, also referred T-MCA has been demonstrated to lower intestinal FXR
to as free fatty acid receptors 2 and 3 (FFAR2 and FFAR3, activity and increase weight gain. Mice with intestinal FXR
respectively) [27]. When SCFAs bind to FFAR3, enteroen- inhibition have diminished ceramide production and altered
docrine L-cells produce the hormone peptide YY, which hepatic gluconeogenesis. The intestine-restricted FXR ago-
is demonstrated to stabilize gastrointestinal motility and nist fexaramine, on the other hand, lowers body weight
boost the amount of accessible energy obtained from meals and insulin resistance, which may be driven by FGF15-
in mice. The secretion of GLP-1 by L-cells is triggered by mediated adipocyte browning [31]. Enhanced insulin sen-
the interactions of SCFAs to FFAR2 and FFAR3 in the sitivity is triggered by intestinal FXR and TGR5 crosstalk
lining cells of the small intestinal tract and colon. This has being stimulated in intestinal L cells. It was found that
a profound impact on insulin release, overall pancreatic GW4064's activation of FXR in intestinal L cells affects
function, and hormonal effects which regulate the sensa- the expression of proglucagon via interfering with the
tion of hunger. Propionate and butyrate possess the ten- glucose-responsive carbohydrate-response element bind-
dency to trigger the synthesis of intestinal gluconeogenic ing protein [32]. It is widely recognized that bile acids
enzymes, and propionate is capable of acting indepen- increase GLP-1 release due to glucose. The observed con-
dently as a precursor for intestinal gluconeogenesis [27]. tradictory effects on glucose tolerance and insulin sensitiv-
SCFAs autonomously perform a wide range of metabolic ity could be addressed by the mice's housing conditions,
activities. In mouse white adipose tissue, all 3 primary genetic makeup, and the varied effects of FXR agonists on
SCFAs have the potential to stimulate FFAR2, which sup- their gut flora [33]. Through regulation of the GM, host
presses insulin signaling, decreases the retention of fat, and metabolism, and diseases, bile acid signaling controls
further increases the expenditure of energy in hepatocytes hepatic metabolism by way of FXR signaling in the intes-
and myocytes. In accordance with a study by Reigstad et al. tinal tract and liver, as well as TGR5 in the intestine. Drug
the stimulation of EC cells by SCFAs in the colon induces therapy for liver diseases, NASH, diabetes, and obesity has
the expression of the tryptophan hydroxylase 1 (Tph1) been designed as a result of recent advances in bile acid
gene and the synthesis of 5-HT [28]. In order to examine metabolism and homeostasis studies. Although mice have
the relationship between intestinal bacteria, gut contrac- a substantially distinct bile acid composition and pool size
tions, and serotonergic gene expression, GF or humanized than humans, the majority of bile acid research is carried
(HM) mice were employed. Results indicated that when out on them[34]. Bariatric procedures are successful in
contrasted to GF mice, the microbiota from traditionally helping obese individuals lose weight and improve their
bred mice and HM mice markedly elevated the colonies in insulin sensitivity, and FXR and TGR5 signaling has been
mRNA levels of TPH1[28]. The findings indicate that GI linked to an increase in serum bile acids and FGF19 after
and metabolic equilibrium depend on SCFA synthesis by bariatric procedures. To develop therapeutic approaches to
the GM, which is essential for controlling GI motility and treat diabetes and NAFL, the fundamental mechanism of
metabolic activities. bile acid signaling’s improvement of diabetes complying
with bariatric surgery must be elucidated [34].
Bile Acids and Their Role in Metabolic Homeostasis
Gut Peptides and Their Impact on Energy Imbalance
The primary role of bile acids is in the equilibrium of and Satiety
cholesterol and dietary lipid absorption. However, current
results from bile acid studies suggest bile acids as essential Peptide YY(PYY)
signaling molecules involved in the regulation of glucose.
Multiple studies have revealed an improvement in the gly- Peptide YY (PYY) is a gut hormone linked to neuropeptide
cemic index of type 2 diabetes patients medicated with Y(NPY) [35]. Both peptides operate through members of
numerous bile acid-binding resins [29]. Bile acids influ- the Y family of receptors and possess the structural motif of
ence the GM having a profound effect on the host metabo- the PP fold. Similar levels of affinity are exhibited by all Y
lism and the development of metabolic disorders. Specific receptors and full-length PYY. PYY3-36, which primarily
agonists that activate the FXR inhibit the production of bile adheres to the Y2 receptor (Y2R), is the amino-terminally
acids and fatty acids while boosting insulin and glucose abbreviated form of PYY that demonstrates the majority of
sensitivity in obese and diabetic rat models[29]. In gen- circulating PYY immunoreactivity.
eral, liver FXR signalling activation delays the advance- Although it is more abundant in distal regions of the gut,
ment of fibrosis and the development of primary biliary PYY exists in L cells along its length. Upon eating, PYY
cirrhosis and non-alcoholic steatohepatitis (NASH) [30]. is released into the bloodstream and is suppressed by fast-
It is uncertain how intestinal FXR signaling affects the ing [35, 36]. In both rodents and people, acute peripheral
Neurochemical Research

administration of PYY3-36 cuts down food intake. These Gut‑Brain Signaling Pathways
results were initially debatable because several different labs
were unable to replicate certain components of the investi- Vagus Nerve Signaling and Its Involvement
gation [36]. Stress can lower normal calorie intake, which in Gut‑Brain Axis
makes it more challenging for anorectic drugs to further
decrease hunger. In mice that have not been accustomed to The ENS, both sympathetic and parasympathetic nerves,
experimental methods or in rats that have been exposed to a along with other ANS components are involved in the gut-
new environment, peripheral PYY3-36 administration does brain neural interaction networks. The sympathetic nerv-
not lower food intake [37]. Subsequent studies have veri- ous system, which is linked to GBA, regulates the lamina
fied that PYY3-36 acutely inhibits feeding in rodents and propria and Peyer's patches additionally, particularly in
primates. The anorectic effects of PYY3-36 are thought to T lymphocyte-rich areas. It also regulates the blood ves-
be mediated by Y2R, as they're attenuated by Y2R antago- sels of the GI tract and the ENS. The sympathetic nervous
nists and eliminated in Y2R-knockout mice. Because of their system regulates immune response stimulation, circula-
modified energy homeostasis, Pyy-knockout mice could tion, the integrity of barriers, and movement within the
offer insight into how the PYY system regulates it. [38]. GBA [45]. A widely recognized conduit for gut-brain
signaling by the vagus nerve, a part of the PNS, is now
Glucagon‑Like Peptide 1(GLP‑1) recognized as a vital microbiota for brain interaction path-
ways. The ENS is often described as "the second brain"
Preproglucagon is a big precursor protein that is synthe- and is made up of intrinsic fundamental afferent neurons
sized by the same type of gut endocrine cell that produces and motor neurons. Recent study using germ-free (GF)
PYY [39]. This is then processed to yield a wide range of mice indicates that GM and ENS are interconnected [46]
biologically relevant peptides, such as glucagon, oxynto- (Fig. 2). The body's immune system is underdeveloped in
modulin, GLP-1 and GLP-2 [40]. There are various distinct GF mice as they lack the commensal gut flora. Studies on
variants of GLP-1, but glucagon-like peptide-1(GLP-1) is the GF animal models have made considerable strides in the
most extensively circulated one. GLP-1 is a powerful incretin previous years by advancing our knowledge of the way
that penetrates the bloodstream after a meal and substantially the microbiota affect gut-brain function [47]. Postnatal
stimulates the release of insulin when administered centrally morphological and physiological disorders were detected
or peripherally. Rodent food intake is substantially decreased in GF mice by contrast with specific pathogen-free (SPF)
when GLP-1 is administered intracerebroventricularly, while mice and mice inoculated with modified Schaedler flora,
peripheral GLP-1 treatment minimises hunger in humans as which normalizes the immune response. In both the jeju-
well as animals[39, 40]. A GLP-1 receptor agonist known as num and ileum of GF mice, the plexus of myenteric nerves
exendin-4 is found in the saliva of the Gila monster lizard, had fewer ganglia and shorter fibres of the nerve [48].
Heloderma suspectum [41]. Exendin, a condensed version of When contrasted with SPF and mice with modified Schae-
this peptide, serves as an antagonist that competes with the dler flora colonization, intestinal movement in GF mice
same receptor [41, 42]. Exendin enhances food intake when declined both in the duration and frequency of spontane-
administered acutely into the brain, and body weight when ous muscular contractions. Numerous studies have inves-
administered continuously. Thus, it appears that endogenous tigated GI nerve impulses and myoelectric contractions in
peripheral GLP-1 may be a component of the physiological mature GF mice. During these investigations, it was deter-
mechanism that reduces appetite and food intake. However, mined that GF mice had impaired gut operation, which
GLP-1 receptor knockout mice had regular dietary habits and was restored by standardization with typical microbiota.
body mass index. Exendin-4, sometimes referred to as exena- Myenteric afterhyperpolarization (AH) nerves in adult
tide and sold under the brand name Byetta, has been shown in GF mice have been found to exhibit decreased excitability
clinical studies to be effective in regulating glucose homeo- of cells and a shorter duration of the suppressive slow
stasis among type 2 diabetic patients [43]. It's crucial to note AH upon just one action response produced by a brief
that exenatide dramatically decreased body weight in treated electrical pulse [49]. The input conductance and relax-
diabetics during phase III 30-week clinical trials. Similar to ing membrane potential also proved different in GF mice
PYY3-36, nausea is a relatively frequent adverse reaction to when contrasted with SPF animals. The colonization of
therapy, but it does not appear to be indissociably associated GF animals with SPF bacteria restored unresponsive mem-
with changes in appetite [44]. These findings also imply that, brane functions and normalized excitability, indicating
even though peripheral GLP-1 doesn't physiologically con- the existence of GM mediates the neuronal activity of the
trol appetite, it could still be possible to employ a medication aforementioned ENS neurons. Although the ENS is able
supplied peripherally to lower body weight by targeting the to operate autonomously of the CNS, the microbiota-ENS
GLP-1 system.
Neurochemical Research

Fig. 2  Gut-brain signaling: vagus nerve signaling

conjunction additionally influences CNS signaling mech- pathways by which gut distension is perceived by these
anisms. ENS neurons' neural functions eventually come afferent terminals are yet unknown. Numerous physiologi-
to a stop in the gut's epithelial lining, so they can either cal mechanosensory functions, such as pulmonary stretch,
immediately react to the luminal surroundings or they light touch, and blood pressure variation, are mediated by
may indirectly react to neurological chemicals made by mechanosensitive PIEZO channels [53]. Whether PIEZO
luminal microbes or enteroendocrine cells [50]. The vagus channels possess a part in gut mechanosensation is uncer-
nerve is a potential route suggested for microbiota-ENS- tain. However, IGLEs' mechano-sensitivity is Gd3 + insen-
brain connection. It has been hypothesized recently that sitive while PIEZO channels are Gd3 + sensitive, point-
ENS neurons could regulate the functioning of CNS via ing to the possibility of PIEZO-independent processes. A
stimulating the vagal afferents. Primary vagal afferents can thin layer of epithelial cells makes up the gastrointestinal
interact with the CNS concurrently in response to changes mucosal barrier that keeps luminal entities out of the GI
in GI stimulants and regulatory peptides. Animal research tissue underlying it. The principal sensors for intestinal
using subdiaphragmatic vagotomy surgery conveys con- nutrients are enteroendocrine cells, which serve as a link
firmation regarding the significance of the vagus nerve between afferent nerves and luminal contents. Cholecys-
mediating microbiota-brain communication [51]. tokinin (CCK), glucagon-like peptide-1 (GLP-1), peptide
Specialized sensory terminal endings can more effec- YY (PYY), and serotonin are just some of the various
tively recognize a wide range of sensory signals. Along GI neurohormones that are derived from enteroendocrine
the GI tract, vagal sensory neurons form various kinds of cells to modulate digestion, nutrient absorption, and food
mechano- and chemo-sensory terminals, such as intragan- intake [54]. This finding suggests that the programming
glionic laminar endings (IGLEs), which link to myenteric logic for intestinal nutrients in enteroendocrine cells is
neuron clusters located between the longitudinal and circu- possibly only partially understood. Traditionally, enter-
lar muscle layers, intramuscular array endings (IMAs) are oendocrine cells were alphabetically categorized into 8
branches of parallel telodendria running parallel to smooth subpopulations based on the hormones they produce. The
muscle fibres more strategically situated near the sphinc- intestinal villus shaft is the path taken by vagal afferents as
ters, and mucosal endings arbour [52]. Additionally, intes- they arborize along the apical pole and develop varicosi-
tinal glands, antral glands, and the extreme posterior taste ties and swellings close to the enteroendocrine cells [55].
buds in the throat and upper oesophagus are all innervated Afferent nerves and enteroendocrine cells were anticipated
by vagal afferents. Although it is believed that IGLEs and to not physically interact for a prolonged period. Instead,
IMAs sense mechanical changes, the underlying molecular they interact paracrinely by diffusing hormones that have
Neurochemical Research

been produced to signal neighboring afferent terminals Hormonal Signaling Mechanisms Between Gut
[56]. Vagal sensory neurons express an array of gut hor- and the Brain
mone receptors, such as CCKAR, GLP1R, NPY2R, and
HTR3A. The significance of this paracrine connection in The homeostatic system is the equilibrium of energy.
the GBA has been largely characterized and is generally Although problems with this system can lead to obesity, a
acknowledged, despite some gut peptide/receptor pairings, relatively recent spike in the prevalence of the disease is
such as GLP-1/GLP1R [57]. Utilizing immunostaining of believed to be a consequence of a regulatory system that
synaptic markers, monosynaptic rabies virus tracing, and cannot function in a certain environment of accessible,
electron microscopy, recent studies have revealed that energizing food increased transportation[62]. The greatest
about two-thirds of endocrine cells are capable of form- opportunity for us to fight obesity could originate from
ing synapses with adjacent neurons as a supplement to commanding aspects of this regulatory system.
this classic model. According to recent studies employing The hypothalamus and brainstem are the primary neural
electron microscopy, monosynaptic rabies virus tracing, regions in charge of regulating energy homeostasis [63].
and immunostaining of synaptic indicators, about two- Numerous modern medications for obesity target central
thirds of enteroendocrine cells have the potential of form- neurotransmitters to assist patients lose weight. Rimon-
ing synapses with proximal nerves. A novel link between abant accomplishes a similar feat by opposing central
vagal afferents and enteroendocrine cells including glu- type-1 cannabinoid receptors, similar to how sibutramine
tamatergic synapses has been identified. When combined suppresses appetite by boosting noradrenaline and sero-
with paracrine communication, these connections facilitate tonin transmission. In clinical trials, rimonabant and
ultrafast millisecond transmission from enteroendocrine sibutramine both led to a minor drop in BMI. However,
cells to vagal sensory neurons, providing gut-brain signal- there is an elevated risk of adverse effects because the
ing a continuous spectrum from fast to slow [58]. the endocannabinoidsystems that these medicines affect
The vagus nerve is the neural pathway for the inter- do not just control appetite [64]. For instance, sibutramine
action between adipose tissue and the liver. Peroxisome has been linked to hypertension, while rimonabant has
proliferative-activated receptor (PPAR)-c2 expression in been linked to potential reproductive and psychological
the liver of experimental mice induced acute hepatic stea- challenges.
tosis while drastically lowering peripheral adiposity that The PNS transmits neurological and hormonal signals
is accompanied by a spike in energy expenditure and a to the hypothalamus and brainstem that are encoded with
surge in systemic insulin sensitivity [59]. In addition to information about the acute nutritional situation and obe-
experiencing greater hepatic PPAR-c2 expression, these sity [59]. Leptin, an adipose hormone that alerts the nerv-
animals also showed significantly better insulin sensitivity ous system to the amount of fat that accumulates in the
as indicated by significantly lower fasting plasma glucose, body, isn't particularly effective in combating obesity [60].
insulin, leptin, and TNF-a levels, decreased glucose out- The majority of obese people have elevated leptin levels in
put from the liver, and lower inflammatory response as their blood and are immune to their physiological effects.
indicated by decreased TNF-a levels [60]. These animals There are currently no established techniques to overcome
displayed high sympathetic nervous system tonus based this barrier.
on the enhanced activity of UCP-1, PGC1a, and blood- Although the hunger experienced prior to a meal is con-
free fatty acid levels elevated. The afferent vagus nerve sidered to be mainly regulated by leptin, shorter-term sig-
regulates the implications of hepatic PPARc2 activity, nals may continue to influence the food intake. These neural
demonstrated by the complete inhibition of an upsurge signals are subsequently coupled with hormones generated
in serum fatty acid levels, and resting intake of oxygen, from the gastrointestinal system and other structures to relay
with destruction of the Vagus Nerve. [61]. Therefore, it information [65]. The vagal pathways that ascend from the
seems that the electrical stimulation of the afferent vagal gut to the CNS are subsequently triggered by these gut hor-
nerve generated by the hepatocytes governs the far-off mones, or these hormones directly affect the neurons in the
ramifications of hepatic PPARc2 activation on peripheral brain.
regions. [62]. Through humoral and neuronal pathways, The primary regions of the CNS that regulate the equi-
the CNS receives feedback from an array of tissues and librium of energy, the hypothalamus and brainstem, can be
organs. These signals are subsequently integrated by the activated by gut hormones [66]. The specific mechanisms of
CNS to generate appropriate feedback, such as parasympa- central action for several gut hormones are obscure or debat-
thetic regulation or stimulation of the sympathetic nervous able. It can be challenging to isolate the endocrine effects
system to ensure energy equilibrium. of certain gut hormones such as ghrelin, PYY and GLP-1
because they also work as neurotransmitters in the brain,
where their roles might vary from those in the periphery.
Neurochemical Research

Leptin homo-oligomerization upon ligand engagement and


binds to JAK, especially JAK2. While weak connections
Since its discovery in 1994, leptin has garnered a lot of inter- between Ob-Rb/JAK1 and Ob-Ra/JAK2 and leptin therapy
est as one of the essential central and peripheral signals for were documented in the context of transient transfection-
the preservation of energy homeostasis [67]. Adipose mass induced JAK overexpression, STAT-binding is specific
and plasma leptin are often inversely correlated. Leptin's to Ob-Rb. However, The STAT-binding exists solely in
main physiological function is to alert the CNS of the num- Ob-Rb [73].
ber of energy reserves that are readily accessible, to limit Research conducted in vivo has revealed that STAT3
dietary intake, and boost energy expenditure [68]. As a is primarily involved in signaling [74]. Tyr1138 is phos-
result, leptin deficiency causes morbid obesity by increas- phorylated, which binds STAT proteins to the Ob-Rb/
ing appetite and food intake. Notably, leptin insufficiency JAK2 complex. To stimulate the transcription of targeted
has only sometimes been linked to severe early childhood genes, notably the one responsible for the suppression of
obesity. Leptin levels among the obese population are usu- the cytokine signalling family (SOCS3), tyrosine-phos-
ally high. Leptin resistance is hypothesized to be the cause phorylated STAT3 molecules split and are delivered to
of the inability of leptin to promote weight reduction in such the nucleus [75]. The Ob-Rb/JAK2 complex's Tyr985
instances [68]. and other binding sites on SOCS3 serve as a conduit for
A disruption of the hormone's characteristic behavior, negative feedback related to leptin signaling. Tyr985 phos-
presumably at diverse locations in the circulation and/or phorylation by JAK2 additionally activates the extracel-
in the signaling cascade, is identified in conjunction with lular signal-regulated kinase (ERK) signalling pathway by
hyperleptinemia, which is generally detected in people with phosphorylating the src homology 2 (SH2) domain of the
obesity and in animal models [69]. Obesity is caused by tyrosine phosphatase SHP-2 (src homology 2-containing
the disturbance of leptin signaling in the hypothalamus, tyrosine phosphatase. Overexpression of SHP-2 has been
demonstrating the critical role of this hormone in maintain- demonstrated to lower SOCS3-mediated inhibition, most
ing energy equilibrium. In the CNS, leptin resistance may likely through interacting with Tyr985 competitively[75].
be specific, as evidenced by the disruption of its impact on Numerous additional consequences result from JAK2
central metabolic processes but the retention of its various autophosphorylation that is not reliant on tyrosine phos-
other effects like sympathetic stimulation of blood pressure phorylation sites on Ob-Rb [76]. One of these results is the
[69]. Leptin receptors (Ob-Rs) reside in several peripheral phosphorylation of receptors for insulin substrate proteins,
tissues and regulate a range of systemic processes including a prerequisite for the activation of downstream signaling
immunity, reproduction and cardiovascular operations, along via the phosphatidylinositol 3′-kinase (PI3K). The leptin-
with its activities in the CNS. [69]. associated PI3K pathway in the heart appears to be essen-
In lean individuals, leptin distributes at a level of 5–15 ng/ tial to cardiomyocyte proliferation and shielding cardiac
mL and is predominantly released by adipocytes [70]. Fast- cells from ischemia/reperfusion damage, along with ERK
ing, testosterone, thyroid stimulating hormone, surplus glu- cascades [77].
cocorticoids and prolonged contact with cold temperatures In the CNS, the hypothalamus is the region where the
lower its expression while overeating, insulin, glucocorti- majority of Ob-Rs reside. Leptin induces the production
coids, endotoxins, and cytokines boost it. Elevated leptin of proopiomelanocortin, which then undergoes process-
expression is observed in the heart upon reperfusion after ing to generate -melanocyte-stimulating hormone and
ischemia, and endothelin (ET)-1 and angiotensin (Ang) II stimulate downstream melanocortin-3 and -4 receptors to
therapy increases leptin levels in cardiomyocyte culture reduce appetite.. In the arcuate nucleus, leptin effectively
serum, implicating synthesis of leptin in the heart [71]. suppresses the hormone neuropeptide Y, which increases
The class I cytokine receptors comprise six isoforms of appetite, as well as agouti-related peptide, inhibiting the
the Ob-R (a to f) reported in a mouse model. The primary signaling of the melanocortin-3 and -4 receptors [78].
isoforms in the heart are Ob-Ra and Ob-Rb, whereas the JAK2/STAT3 signaling induced by ob-Rb is essential
expression of the remaining forms is at modest levels and is for leptin regulation of dietary intake and expenditure of
poorly preserved across species [72]. Ob-Re is the released energy. Although JAK, an insulin receptor substrate, and
variant of leptin that adheres to leptin in the blood and mod- MAPK proteins may be triggered by short versions of the
ulates the level of free leptin. leptin receptor, only Ob-Rb may activate STAT3 [79]. It
The Janus-activated kinase (JAK), signaling sensors has been demonstrated that STAT3 is necessary for the
and transcription activators (STAT), and mitogen-acti- homologous replacement of Ob-Rb by a receptor mutant
vated protein kinase (MAPK) systems have all been dem- for Tyr1138, which regulates the expression of proopi-
onstrated to be induced by ob-Rs [73]. Ob-R undergoes omelanocortin and NPY in the hypothalamus.
Neurochemical Research

Ghrelin essentially irreversible approach there. Ghrelin agonists


could be beneficial in managing certain patient groups
Ghrelin is a peptide hormone that enters the bloodstream experiencing anorexia. Acute ghrelin therapy has been
from the stomach and was first indicated to be an endog- proven to promote digestion in diabetic individuals with
enous receptor for the growth hormone-secretagogue recep- gastroparesis and appetite loss in cancer patients. It may
tor (GHS-R). The addition of an octanoyl ring to the serine additionally boost appetite in dialysis patients who are
residue in the third position is the single distinctive altera- severely malnourished [86]. Thus, persistent ghrelin admin-
tion to the 28 amino acid molecule that makes up ghrelin. istration's orexigenic effects on anorexic individuals could
[80]. For ghrelin to bind to the GHS-R and pass across be advantageous, however, this has yet to be established.
the blood–brain barrier, this acylation is necessary. Stud- Before ghrelin could be utilized as a therapeutic, the issue
ies of increased appetite due to ghrelin were received with of potential adverse effects must be thoroughly addressed.
enormous enthusiasm. Although the other circulating com- It's essential to note that new research suggests peripheral
pounds, such as the adipose hormone leptin and a number of ghrelin treatment can alter the development of hippocam-
gut hormones, are linked to lower dietary intake, ghrelin is pal neurons and the flexibility of the hypothalamus[86].
the sole identified substance that stimulates appetite through Obestatin, an entirely distinct peptide, has been identi-
circulation. [81]. Fasting elevates circulating ghrelin levels, fied to be encoded by the same gene as ghrelin [86].
causing them to reduce after eating. Acute stimulation of When delivered peripherally or intracerebroventricularly,
acylated ghrelin, either centrally or peripherally, stimulates obestatin has been observed to lowe food intake and, when
dietary intake as well as growth hormone secretion in rats; administered peripherally, to decrease weight gain. It has
prolonged use leads to increased body weight. Humans who been proposed that the orphan G-protein-coupled recep-
acquire ghrelin intravenously or subcutaneously observe a tor GPR39 is responsible for these effects [86]. The reason
surge in both appetite and consumption of food. The term why the same gene could generate both an orexigenic and
"hunger hormone" is frequently utilized to describe ghrelin an anorectic signal was the subject of much debate. The
[82]. initial findings, however, have not been corroborated by
According to studies, persistent central ghrelin infusion subsequent investigations, which suggest obestatin may not
enhances the production of enzymes which foster fat accu- signal through GPR39 or have a function in the regulation
mulation in adipose tissue while peripheral ghrelin admin- of calorie consumption [86].
istration limits fat usage [82, 83]. Thus, ghrelin's effects on The overview of the hormones and their effect on recep-
metabolism may not be solely dependent on increased food tors is summarized in Table 2.
consumption.
Initial research revealed that ghrelin- and ghs-r-knockout
mice failed to show significant changes in their eating habits Role of Gut‑Brain Axis in Metabolic
or levels of fat in their bodies [84]. However, it has been Disorders
recently determined that ghrelin signaling disruption mod-
els can exhibit intricate phenotypes associated with energy Obesity and Its Relationship with Gut‑Brain Axis
homeostasis. When administered a high-fat meal plan early
in life, ghs-r-knockout mice and male ghrelin-deficient mice Several facets of physiology, especially glucose homeosta-
exhibit resilience to diet-induced obesity. When fed a high- sis, gut motility, and appetite, are greatly impacted by the
fat diet, ghrelin-deficient mice modified their metabolic GBA. Therapeutics for several diseases like T2DM and obe-
fuel preference, opting for fat as an energy source rather sity, have been designed by studying the role of GBA.
than wild-type controls. Additional study has revealed that Obesity is a severe health condition that is linked to
reduction of ghrelin suppresses diabetes in the ob/ob obese higher mortality and morbidity in addition to increased
mouse while diabetic ghrelin-knockout mice exhibit reduced personal, social, and economic repercussions. The brain
hyperphagia [85]. and gastrointestinal system interact together in a vast and
It is enticing to contemplate employing GHS-R antago- intricate system of neurons and hormones, and their recep-
nists to regulate ghrelin signaling as a strategy for prevent- tors influence appetite, dietary intake, and obesity. The
ing obesity. A recent study has demonstrated that immu- vagus nerve relays the complex hormonal and neurologi-
nizing rats against ghrelin can limit weight gain [85]. It cal signaling caused by the existence of nutrients in the
remains unclear whether inhibiting ghrelin signaling is GI Tract to the brain. Effector fibres carry data from the
an effective long-term treatment for obesity in people, gut to the NTS and the smooth muscle cells of the gut. It
and it might not be an appropriate decision to attempt an obtains data from the NTS and employs it for regulating
Neurochemical Research

Table 2  Major hormones: site Hormones Site of release Receptor Effect on food References
of release, receptors and effects consumption
on food consumption
Peptide YY L cells of the GI tract Y2R ­(PYY3-36) Decreases [81]
Y1R, Y2R, Y4R
GLP-1 Distal L cells GLP-1R [82, 83]
Pancreatic Polypeptide F cells of the Pancreas PPY2 [84]
PPY4
Cholecystokinin L cells of the GI tract CCK1R [85]
CCK2R
Leptin S cells of the GI tract Ob-Rb [85, 86]
Leptin Receptor
Ghrelin Stomach GHSR Increases [86]
Oxyntomodulin L cells of the GI tract GLP-1R Decreases [87]
GCGR​
Amylin β cells of the Pancreas AMY1a [87]
AMY2a
AMY3a
Motilin Mo cells of the Small Intestine GPCR38 [88]

appetite, dietary intake, and energy levels in the neurons indigestible carbs and proteins by increasing the synthe-
of the arcuate nucleus (ARC). Cocaine and amphetamine- sis of several primary fermentation enzymes and nutrient
regulated mRNA, agouti-related protein, orexigenic neu- carriers.
ropeptide Y, anorexigenic peptides (LEP), and pro-opi-
omelanocortin neurons comprise the ARC [86]. Research Non‑Alcoholic Fatty Liver Disease (NAFLD)
has shown that vagotomy reduces the signaling of the ano- and the Influence of the Gut‑Brain Axis
rexigenic hormone, which causes animal models to con-
sume more food and gain more weight. [86]. The release NAFLD is a prevalent chronic liver condition that leads
of several gut hormones, such as GLP-1, ghrelin, PYY, and to various kinds of damage to the liver, from steatosis to
LEP, can be altered by certain gut bacteria. As a result, steatohepatitis [89]. Hepatocellular carcinoma and other
hypothalamic neuroendocrine pathways influence satiety problems, such as cirrhosis, may originate from fibrosis. In
and hunger. SCFAs from microbiota may bind to receptors terms of histology, NAFLD may split into non-alcoholic ste-
on EECs and modify how enteric hormones are released atohepatitis (NASH) and non-alcoholic fatty liver (NAFL).
into the bloodstream. Furthermore, ghrelin, GLP-1, and Obesity, hypertension, hyperlipidemia, T2M, and metabolic
cholecystokinin are secreted by EECs when different syndrome are all strongly associated with the development
taste receptors—such as those for bitter, fat, umami, and of NAFLD. NAFLD is typically thought of to be meta-
sweet—are activated. The primary SCFA released by gut bolic syndrome's hepatic manifestation [90]. Currently, the
bacteria, acetate, reduces hunger processes in the central cause of NAFLD remains to be unknown. The resistance to
hypothalamic processes [87]. insulin, oxidative stress, lipid metabolism shift, release of
Notably, altered microbiota induces a spike in acetate inflammatory cytokines, endoplasmic reticulum stress, gut
concentration, which upregulates the activity of the para- dysbiosis or activation of the gut-liver axis, and genetic and
sympathetic nervous system and elevates levels of ghrelin, epigenetic factors have been suggested to be contributing
stimulating the release of insulin in response to glucose, as factors. Additionally, research has revealed links between
well as obesity [88]. The gut bacteria generate neuroactive hyperammonemia, gut dysbiosis, metabolism, and functional
metabolites that have an effect on the central regulation issues with particular brain regions and the neurobehavioral
of hunger, such as serotonin and GABA. Serotonin-regu- condition known as NAFLD [91]. The key players in the
lated common melanocortin neurons suppress appetite via pathophysiology of NAFL/NASH are GM, its products of
regulating body weight homoeostasis. The right control bacteria, and the intestinal barrier. As a result, the microbi-
of energy balance requires GABA, which is the primary ota-gut-liver-brain axis serves a crucial regulatory function.
inhibitory neurotransmitter in the CNS. The colonization of the GM of NASH patients can worsen
The efficacy of calorie intake from consumed foods is hepatic steatosis and inflammation in sterile mice served a
boosted by obesity-associated bacteria. Therefore, com- high-fat diet (HFD), indicating that GM is crucial in the
pared to a lean-associated GM, an obesity-associated emergence of NAFLD. Furthermore, studies conducted by
microbiota offers the host more energy from additional Boursier et al. revealed an association between gut dysbiosis
Neurochemical Research

and modifications in GM's metabolic activity and the sever- Alzheimer’s Disease
ity of NAFLD [92]. Intestinal permeability, low-grade
inflammation, dietary choline metabolism, bile acid metabo- Alzheimer's disease (AD) is a prevalent neurological con-
lism, and endogenous ethanol production are various ways dition characterized by cognitive decline in elderly people.
that modify GM, promoting hepatic fat deposition. Further- Dementia is a major symptom with this condition, which
more, it has been suggested by Ke Hu et al. that GM assists affects activity, recall, cognition, language, and mental
in the neuroendocrine control of lipid metabolism [93]. functioning [98]. The excessive production and deposition
Additional research is required in order to fully comprehend of amyloid-peptide (A) and the translocation of microbes
the fundamental cause of functional modifications driven by and their byproducts into the brain, where they can initiate
cognitive impairment in NASH, however, it is possible that neurological inflammation and neurodegenerative alterna-
GM metabolites and ammonia can induce neurotoxic dam- tion in AD, are the primary causes of these events. As a
age, which contributes to the impairment of cognition in result, AD is frequently linked to an increasing cerebral
NASH. According to the latest study by Mouries et al. the A-peptide accumulation. The amyloid precursor protein
breakdown of the gut vascular barrier (GVB) and intestinal (APP) is cleaved by proteases to produce the 40–42 amino
epithelial barrier (IEB) are preliminary occurrences in the acid peptide known as “A.” By starting the neuroinflam-
development of NASH, and HFD-mice will display dam- matory response in AD, it plays a significant part in the
aged GVB and transmit bacteria shortly after one week [94]. pathogenesis of AD. In contrast to wild-type mice without
Therefore, the intestinal barrier is just as critical to NAFLD the APP gene, the APP transgenic mice exhibited an enor-
as the GM and its end products are. mous shift in the variety of their gut flora. Additionally,
Comprehensive studies on the microbiota-gut-liver- GF APP transgenic (TG) mice showed a marked reduction
brain axis have rendered the GM an effective player in in the degree of cerebral A-amyloid pathology as com-
NAFLD in recent times. Alteration in lifestyle, diet and pared to control animals with GM [99]. It’s intriguing to
physical activity are first-line therapies for NAFLD. Studies consider that TG mice colonized with microbiota from
have demonstrated an important link between GM and the wild-type mice demonstrated a lower impact on elevat-
management of NAFLD through diet and exercise [95]. The ing the level of cerebral A, as opposed to TG mice that
makeup and variability of the GM are significantly influ- got GM from standard APP transgenic mice [100]. Bauerl
enced by the mediterranean diet, which is more concentrated et al. who stated that alterations to the GM's makeup were
in monounsaturated fatty acids. The high dietary fibre con- noticed in the transgenic APP/PS1 mice model of AD,
tent and polyphenols that are present enhance bifidobacteria provided similar outcomes [101]. When TG animals were
while reducing firmicutes and elevating Bacteroides. Addi- contrasted to wild-type control mice, it was observed that
tionally, spinach consumption also has similar effects by the Erysipelotrichaceae family was more prevalent in TG
regulating the gut flora, which can alleviate the liver dys- mice. Additionally, transgenic APP/PS1 mice displayed
function associated with NAFLD[95]. It's also intriguing less A pathology than regular mice, suggesting that the
that exercise successfully reverses the gut dysbiosis caused GM may be associated with both the aetiology of A and
by the HFD, minimizing the disruption of the gut-liver axis AD. In an additional in vivo investigation, TG APPswe/
and enhancing bile acid homeostasis, thereby helping to pre- PS1dE9 mice exhibited a decline in the expression of post-
vent the development of NAFLD [96]. synaptic density protein 95 (PSD-95) and synapsin I and
a boost in the brain deposition of the A-peptide, Tau pro-
tein, COX-2, and CD11b. In comparison to WT mice, TG
Gut‑Brain Axis and Neurological Disorders mice that received faecal microbiota transplants from WT
mice demonstrated superior amyloid peptide, p-tau protein
Gut-brain interaction not only keeps the body in a state of level, synaptic plasticity, and variation in GM composi-
wellness, but it has also been associated with the emergence tion[102]. Further evidence that AD patients' intestines
of neurological and psychological disorders in the context could mimic the nervous system and trigger inflamma-
of gut-brain axis disruption. The host immune system, host tory and immunological changes related to APP and A
behavior, and neurological growth are all significantly influ- pathology with a higher intestinal A burden, observation
enced by the gut flora. Elevated intestinal permeability, how- of APP, CD68, and p-Tau immunoreactivity has been
ever, results in the translocation of gut microorganisms and reported in both AD patients and APP/PS1 mice [103].
associated neuroactive metabolites and components, which In addition, fructooligosaccharides alleviated cognitive
triggers a neuroinflammatory reaction in the brain [97]. impairments and neurodegeneration in TG APP/PS1 mice
by stimulating the expression levels of PSD-95, synapsin
I, and GLP-1 and suppressing the levels of p-JNK and
GLP1R, based on research on the modulatory properties
Neurochemical Research

of prebiotics against AD[103]. Bacteria are not the only of PD and lowered microbial dysbiosis, faecal SCFAs, and
factor in how the gut microbiome affects AD development; physical disability. The authors further claimed that FMT
viruses have additionally been linked to the disease [104]. suppressed TLR4/TNF- signalling in the gut and brain, as
However, further research is required to fully comprehend well as the proliferation of microglial cells and astrocytes, in
the impact of the microbiome on the GBA in AD. PD mice [110, 111]. In accordance with a recent study, PD
patients had higher colonic expressions of TLR4, CD3 + T
Parkinson’s Disease cells, and cytokines and lower concentrations of bacteria-
producing SCFAs [111]. Researchers additionally demon-
Approximately seven to ten million people globally are strated that rotenone treatment may repair or lessen similar
thought to be affected by Parkinson's disease (PD), the sec- gut and neurological conditions in TLR4-KO mice, indicat-
ond most prevalent neurodegenerative ailment [105]. The ing that TLR4-induced inflammatory signaling serves an
primary characteristics of PD involve both motor and non- essential part in gut and neural inflammation in Parkinson's
motor abnormalities. The most apparent signs on the motor disease. The LPS-induced PD animals exhibited a rise in
side are inflexibility, delay in motion, and resting tremors. microglial cell activation, proinflammatory cytokines, heme
Cognitive difficulties, depressive disorders, mood swings, oxygenase-1 (HO-1) activity, and a drop in ferroportin (Fpn)
sensory disturbances, behavioral changes, and autonomic [111].
dysfunctions all lead to serious handicaps on the non-motor A role for fungi in PD should also be considered [112].
side [106]. Such a broad range of medical symptoms is the Epp and Mravic (2006) pointed to the Candida fungus
result of alpha-synuclein buildup in the brain's central and accompanying some cases of PD, whereas Pisa et al. (2020)
peripheral nervous systems. The vast majority of patients reported several fungal families, including Candida, in the
with PD report GI problems. Individuals with PD have CNS of PD patients [113].
been documented to suffer from a number of GI symptoms,
including hypersalivation, constipation, dysphagia, nausea, Gut‑Brain Axis and Mood Disorders
modified bowel movements, and defecatory dysfunction
[107]. In the early stages of PD, higher intestinal permeation The brain and the gut are linked by the ANS, the HPA axis,
has been reported. Increased intestinal permeability causes and nerves in the GI tract. This allows the cognitive sys-
the bacteria and microorganisms in the gut to migrate, caus- tem to affect intestinal operations such as the functioning
ing oxidative stress and inflammation in the ENS, leading of immune effector cells, and the gut to impact mental well-
to enteric-synucleinopathy in PD [108]. PD has been clini- being, mood, and cognition. Findings from clinical, scien-
cally linked to gastric ulcers, caused by a Helicobacter pylori tific, and epidemiological studies indicate that the gut-brain
(HP) infection, since the 1960s. Drug-induced HP infection relationship—that is, psychological state, behavioral control,
eradication reduces PD symptoms. Evidence gathered from neuromuscular functioning, and HPA regulation—is greatly
numerous studies indicates an association between PD and and substantially influenced by GM [114].
the microbiota which inhabits the gut. The two most sig- Studies have revealed that variations in the microbiota
nificant co-factors in PD are inflammatory state and shifts are connected to modifications in these neural networks, and
in GM [109]. Evaluation of faeces revealed that the cohort they are now striving to clarify the underlying mechanisms
of PD patients had a substantial change in the gut bacte- of action that govern the physiological effects of bacteria on
rial composition, with lower levels of Prevotellaceae and the brain's emotional and cognitive processes, both directly
greater levels of Enterobacteriaceae. These alterations dem- and indirectly. For instance, effective GI disruptions have
onstrated a positive association with greater postural insta- been linked to a number of mood disorders, including anxi-
bility and a unique gait. The levels of alpha-synuclein, a ety and depression. Furthermore, studies have shown that
presynaptic neuronal protein related to motor impairment fetal and neonatal neural growth may be influenced by the
and a neuroinflammatory condition in mice, elevated in GF overall composition of the GM. It should come as no sur-
mice given faecal microbiota from PD patients [109]. In an prise that nutrition has also been linked to the effects of
experiment employing a mouse model of chemically induced GM on cognition [114].
Parkinson's disease, it was additionally found that the overall
makeup of the GM had altered. According to a recent study, Anxiety
PD patients and a human-synuclein overexpressing mice
model of PD (Thy-Syn) have larger relative abundances of According to reports, the global incidence of anxiety dis-
mucin-degrading Verrucomicrobiae and LPS-producing orders was estimated to be 7.3% worldwide, with a range
Gammaproteobacteria than healthy and wild-type mice [108, of 5.3% in African cultures and 10.4% within Euro/Anglo
109]. It was discovered that faecal microbiota transplantation cultures [115]. The complexity and multifaceted nature
(FMT) improved striatal DA and 5-HT in a rodent model of anxiety and trauma-related diseases make it difficult to
Neurochemical Research

distinguish between them and to treat them due to pheno- patients experience a lacklustre response [117]. While keta-
typic variation. These diseases are hypothesized to occur as mine's strong and immediate antidepressant effects have
a result of complex interactions between the environment, altered the course of antidepressant advancements, its long-
the human genome, and the epigenome [115]. Animal mod- term efficiency and side effects must additionally be con-
els serving as practical model systems with greater influ- sidered [118, 119]. Therefore, it is important to investigate
ence on environmental and genetic variables affecting the the pathophysiology of depression extensively to determine
microbiome were the subject of most previous investigations possible therapeutic targets. While the exact mechanism of
on the microbiome. The GF animal model is a potent instru- the microbiota-GBA’s effect on depression is unknown, an
ment for studying the impact of specific bacteria or dietary expanding body of research shows that neuronal, endocrine,
interventions on the GBA axis, as well as for examining immunological, and metabolic pathways play a significant
the impact of GM on behaviour in an effort to demonstrate role in the interaction between intestinal microbes and the
causation. brain.
The operation of the body's immune system and GI tract The influence of gut bacteria on mood and depression
may have substantial effects on behavioral and cognitive disorders can be attributed to their bidirectional contact with
functioning [116]. It should be highlighted, nonetheless, that the CNS through interacting pathways involving immuno-
additional parameters, all of which are still unknown, such logical, endocrine, and neurological system signaling sys-
as time, stress, sex, and species, are also part of such interc- tems. The liver’s anatomical location and circulatory supply
tions. The acceptory animal demonstrated the donor's behav- enable it to play a role in and coordinate crucial immune
ioral profile after FMT. The way that GF animals behave is functions [119]. Within the chemical signaling system, intes-
very different from how control animals behave. GF mice tinal flora controls the production of monoamine neurotrans-
showed an amplification of the HPA axis reaction when mitters, specifically 5-HT, DA, and GABA, which in turn
exposed to stress, which was counteracted once a specific controls the emergence and resilience of neural networks
type of Bifidobacterium was monocolonized [116]. Simi- related to behavior and mood [119]. It is possible to avoid
larly, GF rats displayed greater neuroendocrine responses or normalize the hyperactive state of the HPA axis, which
and anxiety-like behavior in contrast to SPF animals when is demonstrated by elevated cortisol, adrenocorticotropin
compared to stress-sensitive F344 rats that were free of (ACTH), and corticotropin-releasing hormone (CRH), by
particular pathogens (SPF) [116]. However, the connection modifying the GM's role in the microbiota-GBA [120]. A
between behavior and the microbiome is not one-way; stress member of the neurotrophin family, the brain-derived neu-
and emotions may alter the genetic makeup of the gut micro- rotrophic factor (BDNF) has been linked to a number of
bial population by releasing sympathetic neurotransmitters activities, including cell division, neuronal survival, the con-
or stress hormones, which may modify the microbiota's struction of synapses, and the development of neuroplasti-
habitat and affect gut physiology [116, 117]. city [119, 120]. A deficiency in BDNF increases the likeli-
Human probiotic interventions suggest the gut micro- hood of both decreased neuroplasticity and the emergence
biome may be directed to improve results associated with of depression symptoms. Several studies have revealed that
stress, anxiety, and trauma [117]. Nevertheless, a number when compared to healthy controls, depressive patients have
of restrictions make it challenging to properly analyze these comparatively lower serum BDNF levels. Antidepressant
results, such as small numbers of samples and confounding medications like ketamine may regulate cognitive and emo-
factors like gender bias, demographic and ethnic differences, tional functioning in connection to depression by increasing
and variations in probiotic strains and dosages as well as BDNF activation through mTOR signalling [120]. The onset
treatment durations. More coordinated attempts to enlist of depressive-like behaviour may be influenced by altera-
substantial cohorts and use standardized methodologies may tions in the GM since they can elevate BDNF expression
provide further understanding regarding how to focus on the levels.
GM to mitigate the effects of stress and trauma or anxiety. Likewise, by altering the GM and BDNF expression, pro-
biotic therapy has a demonstrated potential as a therapeutic
Depression or preventive strategy for reversing behavioral deficiencies.
According to Bercik et al. oral antimicrobial-treated SPF
Over 350 million people worldwide suffer from depression, mice exhibit a momentarily altered gut microbiome, that
a common neuropsychiatric condition with a high recurrence raises hippocampus BDNF concentrations and minimizes
rate that has a major detrimental effect on both public health depression. Additionally, it was demonstrated that giving
and economic growth [117]. Unfortunately, the majority stressed mice a two-week pretreatment with probiotics
of antidepressants that are presently available on the mar- (a blend of L. helveticus R0052 and B. longum R0175)
ket, work by boosting monoamine neurotransmitter levels, increased the hippocampal expression of BDNF, reversing
with incomplete therapeutic success; in fact, about 30% of synaptic imbalances and eventually improving efficiency
Neurochemical Research

on memory and cognition tasks [120]. Furthermore, in the microbial organisms, including essential microbially-derived
rat model of persistent stress-induced depression, treatment compounds, are shown to display diurnal oscillations in their
with L. helveticus NS8 could enhance hippocampus BDNF relative abundances over a 24-h duration. Microbial oscil-
mRNA expression as well as behaviour performance [120]. lations are observed throughout all studies, even in spite
Thus, the onset of depressive-like behaviour may be influ- of potential covariates including variations in the initial
enced by the GM through modulating the quantity of BDNF. microbial communities of mice cohorts, variations in ani-
mal facilities, and distinctive research designs focused on
Gut Microbiota and Circadian Rhythms manipulating the CR. Additionally, GM is associated with
host cellular rhythms; in a dearth of gut microbes, GF mice
Circadian rhythms (CRs) are 24-h cycles governing an display completely distinctive suprachiasmatic nuclei (SCN)
organism's habits, organs, and cells. In order to maximize and hepatic transcriptional patterns, especially in fundamen-
efficiency and well-being, these rhythms match biological tal CR. In recent times, studies indicate that the timing of
processes with periodic and systematic environmental pat- a person's meal consumption is correlated with their oral
terns [121]. A disturbance in these cycles may have nega- microbiome's diurnal dynamics. It's evident that GM and
tive consequences, such as MeS, tumours, or cardiovascular CRs, particularly peripheral clocks, react to identical stimuli
diseases. More and more evidence is directed to the notion such as feeding habits and nutrition[123].
that the host organism and intrinsic circadian clocks both There are still several obstructions and challenges to
control the GM in response to time changes. Microbiota the study of CR: determining human CR accurately neces-
rhythms are dictated by food and eating timing, which may sitate labor-intensive monitored conditions that are primarily
alter the overall makeup of the microbe communities and attainable in laboratories; participant-to-participant variation
their metabolic activity [121, 122]. These alterations can is significant; and assessing the human GI tract's CR is tough.
have a substantial effect on the immunological and metabolic Although evaluating human CR may seem relatively easy
systems of the host. (for example, using melatonin and temperature regulation),
The gut microbial "organ" is completely integrated into study must be carried out in extremely monitored settings
numerous fundamental life processes and its chemical that limits light exposure while regulating meal timing and
makeup and functioning are highly sensitive to dietary and portion sizes—factors that have been associated with CR
environmental variables [122]. Several studies indicate that [124]. Furthermore, humans vary greatly in terms of their
the GM is crucial to modulating host metabolism, especially genetic makeup, gender, age, and food, all of which effect
when it pertains to responding to and influencing host CRs. CR making it difficult to identify strong signals in data. The
For instance, research on rodents has demonstrated that alter- most significant challenge, though, is that assessing CR in the
ation of the biological circadian clock by genetic changes GI system is not feasible under the experimental conditions.
in animals (i.e., disruptions in Bmal1, Clock, or Rev-erb) While it is viable in rodents, continuous tissue sampling isn't
is linked to characteristics associated with MeS, such as feasible in humans. Clinically targeting metabolic well-being
hepatic steatosis, hyperglycemia, hypoinsulinemia and and nutritional condition has proven to difficult to date, but
abnormal lipid profiles [123]Host metabolism is eventu- this is mostly due to the synchronization of these mecha-
ally influenced by the interactive relationship between the nisms to one another and to external stimuli. Studying certain
GM and CRs[124]. This intricate, bidirectional network con- mechanisms employed by the two "organs" for controlling
tains numerous bioactive chemicals from the host, for which metabolism will enable researchers to utilize that knowledge
scientists have only recently begun to recognize and define and ultimately modify both of these systems for medicinal
their modes of action. Anti-microbial peptides and glu- interventions in metabolic disorders. Within this system, the
cocorticoid hormones are host-derived regulators that are ex vivo assessment of CR using intestine organoids i.e. self-
demonstrated to modulate GM in a circadian setting. Sev- organised three-dimensional structures that can partially rep-
eral antioxidants, tryptophan derivatives, and hydrogen sul- licate the original tissue's characteristics, cell heterogeneity,
phide are examples of microbially derived regulators that and behaviour in vitro, ay show some possibilities (Table 3).
are additionally believed to serve as circadian modulators.
There is presently minimal information available about
the underlying mechanisms that regulate these mediators,
despite the fact that they are probably significant parts of Therapeutic Implications and Interventions
the circadian-microbiome connection and require further
study. In mice, host CRs and GM have been demonstrated The intricate interplay between GBA and metabolic disor-
to share extensive interactions in recent studies. Despite not ders offers a promising avenue for therapeutic advancements.
being subjected to sunlight, diurnal host indications cause This overview delves into the multifaceted therapeutic impli-
oscillations in the amount and functionality of GM. Several cations and interventions that harness the GBA's intricate
Neurochemical Research

network to mitigate metabolic distrurbances. Among the These tests, which included a wide spectrum of human
forefront strategies are probiotics, postbiotics, prebiotics, individuals, used distinct probiotic strains. Probiotic ther-
fecal microbiota transplantation (FMT), and metabolic apy proved to be inefficient for lowering weight and BMI
therapy (Fig. 3). in a recent meta-analysis evaluating the impact of probiotic
Probiotics, live beneficial microorganisms, exhibit supplements on body weight and BMI [128]. The authors
potential in ameliorating metabolic disorders by modulat- emphasized that the potential of the study to reach firm
ing the gut microbiota composition (see sections below for conclusions was constrained by the overall number of rand-
all details). Their administration can promote a balanced omized controlled trials incorporated in the meta-analysis,
microbial profile, fostering favorable metabolic outcomes. the relatively small sample size, and the heterogeneity of the
Postbiotics, comprising bioactive compounds derived from research. To establish which strains may be the most efficient
probiotics, exert direct effects on host physiology and and most suitable prospects for probiotic therapy, additional
metabolism, demonstrating a novel avenue for intervention. studies are required.
Prebiotics, on the other hand, provide substrate for beneficial Postbiotics are an array of biologically active compo-
microorganisms, supporting their growth and activity. These nents comprising inactivated microbes, cell substances and
non-digestible compounds confer metabolic benefits by any easily soluble substances or metabolites produced by
influencing gut microbiota composition and function. FMT, microorganisms [128, 129]. When consumed by people,
a pioneering approach, involves transferring fecal material postbiotics can directly or indirectly regulate supportive bio-
from a healthy donor to a recipient, effectively reestablish- logical reactions. Postbiotics are consequently suitable for
ing a healthier gut microbiome and potentially rectifying underdeveloped countries as they have been demonstrated
metabolic imbalances. Metabolic therapy, encompassing to imitate the behavior and properties of probiotics while
pharmacological and lifestyle interventions, capitalizes on not necessitating strict manufacture or preservation require-
GBA-mediated pathways to regulate metabolic disorders. ments. Several kinds of postbiotic substances manufac-
This comprehensive overview accentuates the integration tured by extrinsic and intrinsic probiotic bacteria are being
of these interventions, rooted in the GBA, as a progressive researched by experts. Therefore, postbiotics are believed
strategy for addressing metabolic disorders holistically. By to provide therapeutic benefits by demonstrating local-
elucidating the intricate relationships between the GBA and ized impacts on particular regions of the intestinal epithe-
metabolic health, this exploration underscores the therapeu- lium. This is owing to highly predefined molecular makeup
tic promise of harnessing this axis for innovative and effec- and extended shelf life[128]. Besides being more resilient
tive interventions (Fig. 4). and possessing extended storage stability, postbiotics seek
to imitate the curative effects of probiotics, minimizing
Probiotics, Prebiotics and Postbiotics for Metabolic the potential hazards of administering live microbes with
Disorders modified gastrointestinal walls or weakened immunological
defences [130].The microbes can be through inactivation,
Probiotics have been scientifically demonstrated to improve fermentation or extraction to produce microbes. The inacti-
lactose tolerance, preventing diarrhoea, strengthening immu- vation can involve methods like heat treatment or chemical
nity, lessening inflammation, and healing dysbiosis in the preservation [130]. Postbiotics can also be produced during
gut-associated with obesity [125]. They are dietary supple- the fermentation process where carbohydrates are broken by
ments containing living microbes that, which when ingested microbes to produce various metabolic products like organic
in sufficient quantities, boost the host's health. Prebiotics are acids, alcohols and carbon dioxide [128]. Postbiotics can
food components that are fermentable but indigestible and also be extracted from certain microorganisms, such as Bifi-
encourage the growth of intestinal microorganisms [126]. dobacterium and Lactobacillus, which produce compounds
To improve the effectiveness of probiotics, prebiotics are like bacteriocins, exopolysaccharides, and conjugated lin-
frequently combined with them. Through immediate altera- oleic acid [128, 130]. These processes allow the generation
tions of the GM and control of gastrointestinal inflamma- of non-living beneficial compounds that can be used to pro-
tion and permeability, probiotics demonstrate their potential mote gut health and minimize potential hazards associated
therapeutic influence on the body. with live microbes.. They could possess a broad spectrum
According to recent human trials on the influence of of prospective effects including anti-allergic and antimicro-
probiotics on metabolic diseases, implementing a probiotic bial characteristics and anti-ageing actions. They may also
supplement for 8–12 weeks could have beneficial impacts bolster the gastrointestinal barrier’s ability to operate while
on blood pressure, weight, BMI, total and low-density lipo- exerting antibacterial responses. To investigate the wide
protein cholesterol, both systolic and diastolic blood pres- range of such postbiotic constituents’ potential biological
sure A1c, and high-sensitivity levels of C-reactivity as well mechanisms and their exact therapeutic effects addressing
as prohibit insulin resistance and body weight gain [127].
Neurochemical Research

Table 3  Types of disorders with Types of diseases Implicated microbiota References


the linked Gut Microbiota
Metabolic disorders
Obesity Firmicutes/Bacteriodetes ratio [19–21]
Eubacterium
Faecalibacterium
Proteobacteria
NAFLD Bacteroides [92, 94]
Veillonella
Ruminococcus
T2D Clostridium Hathewayi [21, 22]
Clostridium Symbiosum
Escherichia Coli
Neurological disorders
Alzheimer disease Collinsella [98, 99]
Bacteroides
Lachnospira
Veillonella
Parkinson disease Desulfovibrio [102, 108, 109]
Bifidobacterium
Lactobacillus
Mood disorders
Anxiety Bifidobacterium [115, 116]
Lactobacillus
Lactococcus
Depression Subdoligranulum [117–119]
Coprococcus
Eggerthella
Ruminococcaceae
Circadian rhythm disorders Selenomoadales [121–124]
Negativicutes
Odoribacter

specific illnesses, it’s essential to thoroughly purify them cell 17 and Treg17/effector Th 17 imbalanceand disrupts
first. cytokines, subsequently contributing to encephalomyelitis
[133]. Further studies should focus on identifying donors
with favorable particular microbial profiles and a typical
Fecal Microbiota Transplantation (FMT) dysbiosis index, as well as accurately identifying the most
appropriate treatment dosage for microbiota transplanta-
In contrast with a healthy lifestyle, fecal microbiota trans- tion. Lately, an intriguing study demonstrated that FMT
plantation (FMT) has gained significant attention in the can primarily exercise its therapeutic ability by reverting
past few years. Randomized regulated studies indicate that the major miRNA alterations associated with the disease
FMT works successfully in managing metabolic disorders [134]. Additionally, FMT regulates the CNS by interacting
like obesity and irritable intestinal disorders such as ulcera- not merely with the gut but also with the Microbiota Gut
tive colitis [131]. The absence of thorough donor screen- Brain Axis, which manifests as encephalomyelitis, depres-
ing parameters could be the reason why FMT in a short sion and nervousnes. Further studies should concentrate on
pilot experiment on canine severe hemorrhagic diarrhoea the potential impacts of FMT on the regulation of encephalic
syndrome solely increased microbiome diversity and pro- areas associated with metabolism so as to promote the appli-
vided no therapeutic advantages[132]. Therefore, the method cation of FMT for the management of metabolic ailments.
necessitates standardization in the setting of the study as the It is noteworthy that FMT exhibits an extensive number of
host's inappropriate details executed by the fecal microbiota indications in experiments with animals that can serve as
could hinder the equilibrium of the inner surroundings inside motivation for researchers [135]. Therefore, the most crucial
the recipients; for instance, constipation-induced GM dys- challenge to be addressed is turning fundamental research
biosis exacerbates the controlled T cell (Treg)/T helper discoveries into therapeutic applications.
Neurochemical Research

Fig. 3  Regulation of dietary


intake by gut-brain axis

Fig. 4  Therapeutic treatment


methods for metabolic disorders

Metabolic Therapy the GM and physiological metabolites (including endo-


toxins and digestive acids) can be additional side effects
The physiological manifestations of obesity, includ- of metabolic therapy.
ing hypertension, hyperglycemia and hyperglycemia are In a research investigation by De Jonge et al. 17 obese
capable of being addressed medically. While altering individuals with T2DM had a noninvasive duodenojeju-
nutritional consumption, gastric elimination, and gastric nostomy bypass [136]. The individuals' body weight and
acid production, metabolic therapy has an impact on the haemoglobin content rose six months into the regimen.
gut flora and the molecular makeup of cholic acid [136]. The amount of predominant small intestinal microbes also
Gut redirection, vagus nerve control, and regulation of varied differently in the excrement. Proteobacteria, Lac-
gut hormones comprise a few of the conventional meta- tobacillus and Veillonella spp were more prevalent in the
bolic therapy procedures. Consequently, modifications in faeces than usual [137]A reduction in weight following
Neurochemical Research

metabolic therapy can be driven by nutritional limits or the gut microbiome, monitoring regular shifts within the
dietary constraints, a smaller gastrointestinal amount, or microbiome, and correlating those alterations to variations
alterations to the levels of gut peptides [137]. Following in illness. Further research into the metabolic impacts of
metabolic treatment, one of the potential pathways for GM with regard to the CNS and the mechanisms governing
shedding pounds could involve the restoration of variation energy consumption and expenditure, as well as additional
in the gut flora. Research involving a substantial number identification of the microbiome of human beings, are neces-
of participants remains crucial on this aspect of the GM's sary. Consequently, the GM may shed light on the intricate
function in outcome post metabolic treatment in order to relationship between mental and physical health, possibly
corroborate these presumptions. leading to new therapeutics for metabolic disorders.

Acknowledgements We thank Department of Scientific Research and


Education, BioNome for helping us with Scientific Writing

Future Directions and Challenges Author Contribution All the authors have equally contributed to the
manuscript.
It has become widely recognized how the integrity Funding The present study is funded by the Department of Scien-
between the GM and cognitive health interacts symbioti- tific Research and Education, BioNome. (Funding ID: DSRE/BNM/
cally and is crucial for maintaining homeostasis. Animals SR/2023/A0108).
and humans both exhibit physiological and behavioral
Data Availability There are no data associated with this study.
alterations as a result of alterations in the typical gut flora.
A novel, potential treatment approach for stress-related Declarations
ailments involves the therapeutic modification of the GM,
especially in the context of inflammatory GI diseases like Conflict of interest The authors declare no conflict of interest.
IBS. There is an imperative to investigate the possible ben-
Ethical Approval Not Applicable.
efits of prebiotics and probiotics towards the administra-
tion of metabolic diseases like T2DM and obesity. Probi- Consent for Publication Not applicable.
otics have demonstrated favorable outcomes with regard
to the treatment of mood disorders such as depression
and anxiety. Furthermore, targeted research is necessary
to investigate how herbal therapies interact with GBA.
Prospective studies should concentrate extensively on the
References
colony-derived impacts of GM on the host as well as itself. 1. Romijn JA, Corssmit EP, Havekes LM, Pijl H (2008) Gut-brain
The variation among individuals in the microbiome axis. Curr Opin Clin Nutr Metab Care 11(4):518–521. https://​
constitutes one of the main obstacles and challenges faced doi.​org/​10.​1097/​MCO.​0b013​e3283​02c9b0
by this particular field of study. Future studies must incor- 2. Ma Q, Xing C, Long W, Wang HY, Liu Q, Wang R-F (2019)
Impact of microbiota on central nervous system and neurological
porate the most recent developments in monitoring the diseases: the gut-brain axis. J Neuroinflamm 16(1):53. https://​
fluctuations of the microbiome and the metabolic changes. doi.​org/​10.​1186/​s12974-​019-​1434-3
In order to examine the metabolic consequences of the 3. Carabotti M, Scirocco A, Maselli MA, Severi C (2015) The gut-
GBA while developing tailored therapies for metabolic brain axis: interactions between enteric microbiota, central and
enteric nervous systems. Ann Gastroenterol 28(2):203–209
disorders, extensive mapping of the human intestinal 4. Jandhyala SM, Talukdar R, Subramanyam C, Vuyyuru H, Sasi-
microbiome is vital. kala M, Nageshwar Reddy D (2015) Role of the normal gut
microbiota. World J Gastroenterol 21(29):8787–8803. https://​
doi.​org/​10.​3748/​wjg.​v21.​i29.​8787
5. Lin Y, Sun Z (2010) Current views on type 2 diabetes. J Endo-
Conclusion crinol 204(1):1–11. https://​doi.​org/​10.​1677/​JOE-​09-​0260
6. Copeland KC, Silverstein J, Moore KR, Prazar GE, Raymer T,
In conclusion, there is a medical imperative for the crea- Shiffman RN, Springer SC, Thaker VV, Anderson M, Spann SJ,
tion of new and stronger medications for metabolic disor- Flinn SK, American Academy of Pediatrics (2013) Management
of newly diagnosed type 2 diabetes mellitus (T2DM) in children
ders. Experts can confirm that there tends to be a recip- and adolescents. Pediatrics 131(2):364–382. https://​doi.​org/​10.​
rocal connection between GM and metabolic disorder by 1542/​peds.​2012-​3494
employing animal models in fecal transference investiga- 7. Fridovich-Keil JL (2006) Galactosemia: the good, the bad, and
tions. The rapid alterations in the genetic makeup of GM and the unknown. J Cell Physiol 209(3):701–705. https://​doi.​org/​10.​
1002/​jcp.​20820
their consequences on human metabolism should be inte- 8. Seyedyazdani R (2008) Inborn errors of amino acid metabo-
grated into future research. It may be advisable to differ- lism sugar and mucopolysaccharides in children with mental
entiate the sequence of these events in humans by imaging
Neurochemical Research

retardation. Clin Genet 14(5):310–310. https://d​ oi.o​ rg/1​ 0.1​ 111/j.​ A, Reygner J, Samczuk P, Serino M, Sikora M, Terranegra A,
1399-​0004.​1978.​tb021​94.x Ufnal M, Villeger R, Pichon C, Konturek P, Edeas M (2022)
9. Özen H (2007) Glycogen storage diseases: new perspectives. Microbiota medicine: towards clinical revolution. J Transl Med
World J Gastroenterol 13(17):2541. https://​doi.​org/​10.​3748/​wjg.​ 20(1):111. https://​doi.​org/​10.​1186/​s12967-​022-​03296-9
v13.​i18.​2541 26. Wingler LM, Lefkowitz RJ (2020) Conformational basis of G
10. Castro RG, Pérez AMG, Curto MCR, Álvarez JC, Ferreirós AC, protein-coupled receptor signaling versatility. Trends Cell Biol
Cuadros AV, Bueno DM, Fernández AJC (2019) A new case of 30(9):736–747. https://​doi.​org/​10.​1016/j.​tcb.​2020.​06.​002
Schindler disease. Eur J Case Rep Intern Med 6(11):001269 27. Gautier-Stein A, Rajas F, Mithieux G (2021) Intestinal gluco-
11. Karimzadeh P, Jafari N, Biglari HN, Dari SJ, Abadi FA, Alaee neogenesis and protein diet: future directions. Proc Nutr Soc
MR et al (2014) GM2-Gangliosidosis (Sandhoff and Tay Sachs 80(2):118–125. https://​doi.​org/​10.​1017/​S0029​66512​00079​22
disease): diagnosis and neuroimaging findings (an Iranian pedi- 28. Legan TB, Lavoie B, Mawe GM (2022) Direct and indirect
atric case series). Iran J Child Neurol 8(3):55 mechanisms by which the gut microbiota influence host seroto-
12. Suzuki Y (2013) Chaperone therapy update: fabry disease, GM1- nin systems. Neurogastroenterol Motil 34(10):e14346. https://​
gangliosidosis and gaucher disease. Brain Dev 35(6):515–523. doi.​org/​10.​1111/​nmo.​14346
https://​doi.​org/​10.​1016/j.​brain​dev.​2012.​12.​002 29. Vlachos D, Malisova S, Lindberg FA, Karaniki G (2020) Glyce-
13. Vanier MT (2010) Niemann-pick disease type C. Orphanet J Rare mic index (GI) or glycemic load (GL) and dietary interventions
Dis 5:16. https://​doi.​org/​10.​1186/​1750-​1172-5-​16 for optimizing postprandial hyperglycemia in patients with T2
14. László A, Nagy I, Szücs L, Havass Z, Sztriha L, Svékus A, diabetes: a review. Nutrients 12(6):1561. https://d​ oi.o​ rg/1​ 0.3​ 390/​
Veres E (1992) Biochemical, clinical and genetic analysis of nu120​61561
various aminoacidopathies (non-ketotic hyperglycemia, maple 30. Francque S, Szabo G, Abdelmalek MF, Byrne CD, Cusi K,
syrup urine disease, histidinemia, tyrosinemia). Orv Hetil Dufour J-F, Roden M, Sacks F, Tacke F (2021) Nonalcoholic
133(48):3075–3080 steatohepatitis: the role of peroxisome proliferator-activated
15. Blackburn P, Gass J, Vairo F, Farnham K, Atwal H, Macklin S, receptors. Nat Rev Gastroenterol Hepatol 18(1):24–39. https://​
Klee E, Atwal P (2017) Maple syrup urine disease: mechanisms doi.​org/​10.​1038/​s41575-​020-​00366-5
and management. Appl Clin Genet 10:57–66 31. Katafuchi T, Makishima M (2022) Molecular basis of bile
16. Zielonka M, Kölker S, Gleich F, Stützenberger N, Nagamani acid-FXR-FGF15/19 signaling axis. Int J Mol Sci 23(11):6046.
SCS, Gropman AL, Hoffmann GF, Garbade SF, Posset R (2019) https://​doi.​org/​10.​3390/​ijms2​31160​46
Urea Cycle Disorders Consortium (UCDC) and the European 32. Anderson KM, Gayer CP (2021) The pathophysiology of
Registry and Network for Intoxication type Metabolic Diseases farnesoid X receptor (FXR) in the GI tract: inflammation, bar-
(E-IMD) Consortia Study Group. Early Prediction of Pheno- rier function and innate immunity. Cells 10(11):3206. https://d​ oi.​
typic Severity in Citrullinemia Type 1. Ann Clin Transl Neurol org/​10.​3390/​cells​10113​206
6(9):1858–1871. https://​doi.​org/​10.​1002/​acn3.​50886 33. Wei S, He T, Zhao X, Jing M, Li H, Chen L, Zheng R, Zhao
17. Ribas GS, Lopes FF, Deon M, Vargas CR (2022) Hyperam- Y (2023) Alterations in the gut microbiota and serum metabo-
monemia in inherited metabolic diseases. Cell Mol Neurobiol lomics of spontaneous cholestasis caused by loss of FXR signal
42(8):2593–2610. https://​doi.​org/​10.​1007/​s10571-​021-​01156-6 in mice. Front Pharmacol 14:1197847. https://​doi.​org/​10.​3389/​
18. Fan Y, Pedersen O (2021) Gut microbiota in human metabolic fphar.​2023.​11978​47
health and disease. Nat Rev Microbiol 19(1):55–71. https://​doi.​ 34. Xu X, Poulsen KL, Wu L, Liu S, Miyata T, Song Q, Wei Q, Zhao
org/​10.​1038/​s41579-​020-​0433-9 C, Lin C, Yang J (2022) Targeted therapeutics and novel signal-
19. Loos RJF, Yeo GSH (2022) The genetics of obesity: from discov- ing pathways in non-alcohol-associated fatty liver/steatohepatitis
ery to biology. Nat Rev Genet 23(2):120–133. https://​doi.​org/​10.​ (NAFL/NASH). Signal Transduct Target Ther 7(1):287. https://​
1038/​s41576-​021-​00414-z doi.​org/​10.​1038/​s41392-​022-​01119-3
20. Ecker BL, Lee JY, Sterner CJ, Solomon AC, Pant DK, Shen F, 35. McCarty TR, Jirapinyo P, Thompson CC (2020) Effect of sleeve
Peraza J, Vaught L, Mahendra S, Belka GK, Pan T-C, Schmitz gastrectomy on ghrelin, GLP-1, PYY, and GIP gut hormones: a
KH, Chodosh LA (2019) Impact of obesity on breast cancer systematic review and meta-analysis: a systematic review and
recurrence and minimal residual disease. Breast Cancer Res meta-analysis. Ann Surg 272(1):72–80. https://​doi.​org/​10.​1097/​
21(1):41. https://​doi.​org/​10.​1186/​s13058-​018-​1087-7 SLA.​00000​00000​003614
21. Zafar H, Saier Jr. MH (2021) Gut bacteroides species in health 36. El-Salhy M, Hatlebakk JG, Hausken T (2020) Possible role of
and disease. Gut Microbes 13(1):1–20. https://​doi.​org/​10.​1080/​ peptide YY (PYY) in the pathophysiology of irritable bowel syn-
19490​976.​2020.​18481​58 drome (IBS). Neuropeptides 79(101973):101973. https://d​ oi.o​ rg/​
22. Castaner O, Goday A, Park Y-M, Lee S-H, Magkos F, Shiow 10.​1016/j.​npep.​2019.​101973
S-ATE, Schröder H (2018) The gut microbiome profile in obe- 37. Conlon JM (2002) The origin and evolution of peptide YY (PYY)
sity: a systematic review. Int J Endocrinol 2018:1–9. https://​doi.​ and pancreatic polypeptide (PP). Peptides 23(2):269–278. https://​
org/​10.​1155/​2018/​40957​89 doi.​org/​10.​1016/​s0196-​9781(01)​00608-8
23. Lindsay EC, Metcalfe NB, Llewellyn MS (2020) The potential 38. Ballantyne GH (2006) Peptide YY (1–36) and peptide YY
role of the gut microbiota in shaping host energetics and meta- (3–36): Part I. Distribution, release and actions. Obes Surg
bolic rate. J Anim Ecol 89(11):2415–2426. https://​doi.​org/​10.​ 16(5):651–658
1111/​1365-​2656.​13327 39. Vrang N, Larsen PJ (2010) Preproglucagon derived peptides
24. Müller TD, Finan B, Bloom SR, D’Alessio D, Drucker DJ, Flatt GLP-1, GLP-2 and oxyntomodulin in the CNS: role of periph-
PR, Fritsche A, Gribble F, Grill HJ, Habener JF, Holst JJ, Lang- erally secreted and centrally produced peptides. Prog Neuro-
hans W, Meier JJ, Nauck MA, Perez-Tilve D, Pocai A, Reimann biol 92(3):442–462. https://​doi.​org/​10.​1016/j.​pneur​obio.​2010.​
F, Sandoval DA, Schwartz TW, Seeley RJ, Stemmer K, Tang- 07.​003
Christensen M, Woods SC, DiMarchi RD, Tschöp MH (2019) 40. Kim ER, Park JS, Kim JH, Oh JY, Oh IJ, Choi DH, Lee YS,
Glucagon-like peptide 1 (GLP-1). Mol Metab 30:72–130. https://​ Park IS, Kim S, Lee DH, Cheon JH, Bae J-W, Lee M, Cho JW,
doi.​org/​10.​1016/j.​molmet.​2019.​09.​010 An IB, Nam EJ, Yang S-I, Lee M-S, Bae SH, Lee Y-H (2022)
25. Gebrayel P, Nicco C, Al Khodor S, Bilinski J, Caselli E, Comelli A GLP-1/GLP-2 receptor dual agonist to treat NASH: targeting
EM, Egert M, Giaroni C, Karpinski TM, Loniewski I, Mulak
Neurochemical Research

the gut-liver axis and microbiome. Hepatology 75(6):1523–1538. 56. Date Y, Murakami N, Toshinai K, Matsukura S, Niijima A,
https://​doi.​org/​10.​1002/​hep.​32235 Matsuo H, Kangawa K, Nakazato M (2002) The role of the gas-
41. Parkes D, Jodka C, Smith P, Nayak S, Rinehart L, Gingerich R, tric afferent vagal nerve in ghrelin-induced feeding and growth
Chen K, Young A (2001) Pharmacokinetic actions of exendin-4 hormone secretion in rats. Gastroenterology 123(4):1120–1128.
in the rat: comparison with glucagon-like peptide-1. Drug Dev https://​doi.​org/​10.​1053/​gast.​2002.​35954
Res 53(4):260–267. https://​doi.​org/​10.​1002/​ddr.​1195 57. Gabe MBN, Gasbjerg LS, Gadgaard S, Lindquist P, Holst JJ,
42. Nachnani JS, Bulchandani DG, Nookala A, Herndon B, Molteni Rosenkilde MM (2022) N-Terminal alterations turn the gut
A, Pandya P, Taylor R, Quinn T, Weide L, Alba LM (2010) Bio- hormone GLP-2 into an antagonist with gradual loss of GLP-2
chemical and histological effects of exendin-4 (Exenatide) on receptor selectivity towards more GLP-1 receptor interaction. Br
the rat pancreas. Diabetologia 53(1):153–159. https://d​ oi.o​ rg/1​ 0.​ J Pharmacol 179(18):4473–4485. https://​doi.​org/​10.​1111/​bph.​
1007/​s00125-​009-​1515-4 15866
43. Nuche-Berenguer B, Moreno P, Portal-Nuñez S, Dapía S, Esbrit 58. Kaelberer MM, Bohórquez DV (2018) The now and then of gut-
P, Villanueva-Peñacarrillo ML (2010) Exendin-4 exerts osteo- brain signaling. Brain Res 1693:192–196. https://​doi.​org/​10.​
genic actions in insulin-resistant and type 2 diabetic states. Regul 1016/j.​brain​res.​2018.​03.​027
Pept 159(1–3):61–66. https://​doi.​org/​10.​1016/j.​regpep.​2009.​06.​ 59. Shaaban Z, Khoradmehr A, Amiri-Yekta A, Nowzari F, Jafarza-
010 deh Shirazi MR, Tamadon A (2021) Pathophysiologic mecha-
44. Grandt D, Schimiczek M, Beglinger C, Layer P, Goebell H, Eys- nisms of insulin secretion and signaling-related genes in etiology
selein VE, Reeve Jr. JR (1994) Two molecular forms of peptide of polycystic ovary syndrome. Genet Res (Camb) 2021:7781823.
YY (PYY) are abundant in human blood: characterization of a https://​doi.​org/​10.​1155/​2021/​77818​23
radioimmunoassay recognizing PYY 1–36 and PYY 3–36. Regul 60. Papamichael K, Cheifetz AS (2016) Use of anti-TNF drug lev-
Pept 51(2):151–159. https://​doi.​org/​10.​1016/​0167-​0115(94)​ els to optimise patient management. Frontline Gastroenterol
90204-6 7(4):289–300. https://​doi.​org/​10.​1136/​flgas​tro-​2016-​100685
45. Foster JA, Rinaman L, Cryan JF (2017) Stress & the gut-brain 61. Kontoyiannis D, Pasparakis M, Pizarro TT, Cominelli F, Kol-
axis: regulation by the microbiome. Neurobiol Stress 7:124–136. lias G (1999) Impaired on/off regulation of TNF biosynthesis in
https://​doi.​org/​10.​1016/j.​ynstr.​2017.​03.​001 mice lacking TNF AU-rich elements: implications for joint and
46. Hyland NP, Cryan JF (2016) Microbe-host interactions: influence gut-associated immunopathologies. Immunity 10(3):387–398.
of the gut microbiota on the enteric nervous system. Dev Biol https://​doi.​org/​10.​1016/​s1074-​7613(00)​80038-2
417(2):182–187. https://​doi.​org/​10.​1016/j.​ydbio.​2016.​06.​027 62. Galgani JE, Kelley DE, Albu JB, Krakoff J, Smith SR, Bray GA,
47. Arneth BM (2018) Gut-brain axis biochemical signalling from Ravussin E, Look AHEAD Adipose Research Group (2013) Adi-
the gastrointestinal tract to the central nervous system: gut dys- pose tissue expression of adipose (WDTC1) gene is associated
biosis and altered brain function. Postgrad Med J 94(1114):446– with lower fat mass and enhanced insulin sensitivity in humans:
452. https://​doi.​org/​10.​1136/​postg​radme​dj-​2017-​135424 adipose gene and obesity phenotype. Obesity (Silver Spring)
48. Manca C, Boubertakh B, Leblanc N, Deschênes T, Lacroix S, 21(11):2244–2248. https://​doi.​org/​10.​1002/​oby.​20371
Martin C, Houde A, Veilleux A, Flamand N, Muccioli GG, Ray- 63. Murphy KG, Bloom SR (2006) Gut hormones and the regulation
mond F, Cani PD, Di Marzo V, Silvestri C (2020) Germ-free of energy homeostasis. Nature 444(7121):854–859. https://​doi.​
mice exhibit profound gut microbiota-dependent alterations of org/​10.​1038/​natur​e05484
intestinal endocannabinoidome signaling. J Lipid Res 61(1):70– 64. Miller GD (2019) Appetite regulation: hormones, peptides, and
85. https://​doi.​org/​10.​1194/​jlr.​RA119​000424 neurotransmitters and their role in obesity. Am J Lifestyle Med
49. Vogalis F, Harvey JR, Neylon CB, Furness JB (2002) Regula- 13(6):586–601. https://​doi.​org/​10.​1177/​15598​27617​716376
tion of K+ channels underlying the slow afterhyperpolarization 65. Nogaller AM, Abdeeva OV (1991) Allergy and the endocrine sys-
in enteric afterhyperpolarization-generating myenteric neurons: tem of the gastrointestinal tract. Klin Med (Mosk) 69(4):17–23
role of calcium and phosphorylation. Clin Exp Pharmacol Phys- 66. Drucker DJ (2007) The role of gut hormones in glucose homeo-
iol 29(10):935–943. https://​doi.​org/​10.​1046/j.​1440-​1681.​2002.​ stasis. J Clin Invest 117(1):24–32. https://d​ oi.​org/​10.​1172/​JCI30​
03755.x 076
50. Morais LH, Schreiber 4th HL, Mazmanian SK (2021) The 67. Myers MG, Cowley MA, Münzberg H (2008) Mechanisms of
gut microbiota-brain axis in behaviour and brain disorders. leptin action and leptin resistance. Annu Rev Physiol 70(1):537–
Nat Rev Microbiol 19(4):241–255. https://​doi.​org/​10.​1038/​ 556. https://​doi.​org/​10.​1146/​annur​ev.​physi​ol.​70.​113006.​100707
s41579-​020-​00460-0 68. Wang B, Kong Q, Li X, Zhao J, Zhang H, Chen W, Wang G
51. Bonaz B, Bazin T, Pellissier S (2018) The vagus nerve at the (2020) A high-fat diet increases gut microbiota biodiversity
interface of the microbiota-gut-brain axis. Front Neurosci 12:49. and energy expenditure due to nutrient difference. Nutrients
https://​doi.​org/​10.​3389/​fnins.​2018.​00049 12(10):3197. https://​doi.​org/​10.​3390/​nu121​03197
52. Ratcliffe EM, Farrar NR, Fox EA (2011) Development of 69. Friedman JM (1997) Leptin, leptin receptors and the control of
the vagal innervation of the gut: steering the wandering body weight. Eur J Med Res 2(1):7–13
nerve: enteric vagal development. Neurogastroenterol Motil 70. Delort L, Rossary A, Farges M-C, Vasson M-P, Caldefie-
23(10):898–911. https://​doi.​org/​10.​1111/j.​1365-​2982.​2011.​ Chézet F (2015) Leptin, adipocytes and breast cancer: focus
01764.x on inflammation and anti-tumor immunity. Life Sci 140:37–48.
53. Volkers L, Mechioukhi Y, Coste B (2015) Piezo channels: from https://​doi.​org/​10.​1016/j.​lfs.​2015.​04.​012
structure to function. Pflugers Arch 467(1):95–99. https://​doi.​ 71. Wei Y, Wang L, Clark JCM, Dass CR, Choong PFM (2008)
org/​10.​1007/​s00424-​014-​1578-z Elevated leptin expression in a rat model of fracture and trau-
54. Browning KN, Travagli RA (2011) Plasticity of vagal brainstem matic brain injury. J Pharm Pharmacol 60(12):1667–1672.
circuits in the control of gastrointestinal function. Auton Neuro- https://​doi.​org/​10.​1211/​jpp/​60.​12.​0013
sci 161(1–2):6–13. https://​doi.​org/​10.​1016/j.​autneu.​2010.​11.​001 72. Duggal P (2002) Expression of the long (OB-RB) and short
55. Gribble FM, Reimann F (2016) Enteroendocrine cells: chemosen- (OB-RA) forms of the leptin receptor throughout the oestrous
sors in the intestinal epithelium. Annu Rev Physiol 78(1):277– cycle in the mature rat ovary. J Reprod Fertil 123(6):899–905.
299. https://​doi.​org/​10.​1146/​annur​ev-​physi​ol-​021115-​105439 https://​doi.​org/​10.​1530/​reprod/​123.6.​899
Neurochemical Research

73. Beach KM, Wang J, Otteson DC (2017) Regulation of stem cell strategies. Nat Med 24(7):908–922. https://​doi.​org/​10.​1038/​
properties of Müller Glia by JAK/STAT and MAPK signaling s41591-​018-​0104-9
in the mammalian retina. Stem Cells Int 2017:1610691. https://​ 90. Rinella ME, Sanyal AJ (2016) Management of NAFLD: a stage-
doi.​org/​10.​1155/​2017/​16106​91 based approach. Nat Rev Gastroenterol Hepatol 13(4):196–205.
74. Pardanani A (2008) JAK2 inhibitor therapy in myeloprolifera- https://​doi.​org/​10.​1038/​nrgas​tro.​2016.3
tive disorders: rationale, preclinical studies and ongoing clini- 91. Arias N, De Chiara F, Habtesion A, Davies N, Thomsen K, Jalan
cal trials. Leukemia 22(1):23–30. https://​doi.​org/​10.​1038/​sj.​ R (2017) Neurobehavioral disturbances and hyperammonemia
leu.​24049​48 are evident in rodent model of NAFLD prior to cirrhosis stages.
75. Carow B, Rottenberg ME (2014) SOCS3, a major regulator of J Hepatol 66(1):S601–S602. https://​doi.​org/​10.​1016/​s0168-​
infection and inflammation. Front Immunol 5:58. https://​doi.​ 8278(17)​31635-5
org/​10.​3389/​fimmu.​2014.​00058 92. Mantovani A, Dalbeni A (2021) Treatments for NAFLD: state of
76. Levine RL, Pardanani A, Tefferi A, Gilliland DG (2007) Role art. Int J Mol Sci 22(5):2350. https://d​ oi.o​ rg/1​ 0.3​ 390/i​ jms22​ 0523​
of JAK2 in the pathogenesis and therapy of myeloproliferative 50
disorders. Nat Rev Cancer 7(9):673–683. https://​doi.​org/​10.​ 93. Hu K et al (2022) Effects of the lipid metabolites and the gut
1038/​nrc22​10 microbiota in ApoE−/− mice on atherosclerosis co-depression
77. Berthoud H-R, Sutton GM, Townsend RL, Patterson LM, from the microbiota-gut-brain axis. Front Mol Biosci 9:786492
Zheng H (2006) Brainstem mechanisms integrating gut-derived 94. Wan X, Xu C, Yu C, Li Y (2016) Role of NLRP3 inflammasome
satiety signals and descending forebrain information in the in the progression of NAFLD to NASH. Can J Gastroenterol
control of meal size. Physiol Behav 89(4):517–524. https://​ Hepatol 2016:6489012. https://​doi.​org/​10.​1155/​2016/​64890​12
doi.​org/​10.​1016/j.​physb​eh.​2006.​08.​018 95. Romero-Gómez M, Zelber-Sagi S, Trenell M (2017) Treatment
78. Fetissov SO, Hökfelt T (2019) On the origin of eating dis- of NAFLD with diet, physical activity and exercise. J Hepatol
orders: altered signaling between gut microbiota, adaptive 67(4):829–846. https://​doi.​org/​10.​1016/j.​jhep.​2017.​05.​016
immunity and the brain melanocortin system regulating feed- 96. Song Q, Zhang X (2022) The role of gut-liver axis in gut microbi-
ing behavior. Curr Opin Pharmacol 48:82–91. https://​doi.​org/​ ome dysbiosis associated NAFLD and NAFLD-HCC. Biomedi-
10.​1016/j.​coph.​2019.​07.​004 cines 10(3):524. https://​doi.​org/​10.​3390/​biome​dicin​es100​30524
79. Vaisse C, Halaas JL, Horvath CM, Darnell Jr JE, Stoffel M, 97. Chen X, D’Souza R, Hong S-T (2013) The role of gut microbiota
Friedman JM (1996) Leptin activation of Stat3 in the hypo- in the gut-brain axis: current challenges and perspectives. Protein
thalamus of wild-type and Ob/Ob mice but not Db/Db mice. Cell 4(6):403–414. https://​doi.​org/​10.​1007/​s13238-​013-​3017-x
Nat Genet 14(1):95–97. https://​doi.​org/​10.​1038/​ng0996-​95 98. Serrano-Pozo A, Frosch MP, Masliah E, Hyman BT (2011) Neu-
80. Kojima M, Kangawa K (2008) Structure and function of ghre- ropathological alterations in Alzheimer disease. Cold Spring
lin. Results Probl Cell Differ 46:89–115. https://​doi.​org/​10.​ Harb Perspect Med 1(1):a006189. https://​doi.​org/​10.​1101/​cshpe​
1007/​400_​2007_​049 rspect.​a0061​89
81. Martínez-Sánchez N (2020) There and back again: leptin 99. Mount C, Downton C (2006) Alzheimer disease: progress
actions in white adipose tissue. Int J Mol Sci 21(17):6039. or profit? Nat Med 12(7):780–784. https://​doi.​org/​10.​1038/​
https://​doi.​org/​10.​3390/​ijms2​11760​39 nm0706-​780
82. Wren AM (2001) Ghrelin enhances appetite and increases food 100. Ashe KH, Zahs KR (2010) Probing the biology of Alzheimer’s
intake in humans. J Clin Endocrinol Metab 86(12):5992–5992. disease in mice. Neuron 66(5):631–645. https://​doi.​org/​10.​
https://​doi.​org/​10.​1210/​jc.​86.​12.​5992 1016/j.​neuron.​2010.​04.​031
83. Garin MC, Burns CM, Kaul S, Cappola AR (2013) Clinical 101. Bendlin BB (2022) Gut microbiome alterations in Alzheimer’s
review: the human experience with ghrelin administration. J disease. Alzheimers. https://​doi.​org/​10.​1002/​alz.​069987
Clin Endocrinol Metab 98(5):1826–1837. https://​doi.​org/​10.​ 102. Weiner WJ (2008) Early diagnosis of Parkinson’s disease and
1210/​jc.​2012-​4247 initiation of treatment. Rev Neurol Dis 5(2):46–53; quiz 54–55
84. Noh JY, Wu C-S, DeLuca JAA, Devaraj S, Jayaraman A, 103. Skorvanek M, Goldman JG, Jahanshahi M, Marras C, Rektorova
Alaniz RC, Tan X-D, Allred CD, Sun Y (2022) Novel role of I, Schmand B, van Duijn E, Goetz CG, Weintraub D, Stebbins
ghrelin receptor in gut dysbiosis and experimental colitis in GT, Martinez-Martin P (2018) Global scales for cognitive
aging. Int J Mol Sci 23(4):2219. https://​doi.​org/​10.​3390/​ijms2​ screening in Parkinson’s disease: critique and recommendations.
30422​19 Mov Disord 33(2):208–218
85. Shimazu-Kuwahara S, Harada N, Yamane S, Joo E, Sankoda A, 104. Overvliet GM, Vlaar A, Haan C, Lampe I (2020) Pharmaco-
Kieffer TJ, Inagaki N (2017) Attenuated secretion of glucose- logical treatment of apathy in Parkinson’s disease, review of the
dependent insulinotropic polypeptide (GIP) does not alleviate literature. Res Square. https://​doi.​org/​10.​21203/​rs.2.​21219/​v1
hyperphagic obesity and insulin resistance in Ob/Ob mice. Mol 105. Shprecher DR, Derkinderen P (2012) Parkinson disease: the
Metab 6(3):288–294. https://​doi.​org/​10.​1016/j.​molmet.​2017.​01.​ enteric nervous system spills its guts. Neurology 78(9):683;
006 author reply 683. https://​doi.​org/​10.​1212/​WNL.​0b013​e3182​
86. Santner A, Estelle M (2009) Recent advances and emerging 4bd195.
trends in plant hormone signalling. Nature 459(7250):1071– 106. Petrov VA, Saltykova IV, Zhukova IA, Alifirova VM, Zhukova NG,
1078. https://​doi.​org/​10.​1038/​natur​e08122 Dorofeeva YB, Tyakht AV, Kovarsky BA, Alekseev DG, Kostryu-
87. Heimann E, Nyman M, Pålbrink A-K, Lindkvist-Petersson K, kova ES, Mironova YS, Izhboldina OP, Nikitina MA, Perevoz-
Degerman E (2016) Branched short-chain fatty acids modulate chikova TV, Fait EA, Babenko VV, Vakhitova MT, Govorun VM,
glucose and lipid metabolism in primary adipocytes. Adipocyte Sazonov AE (2017) Analysis of gut microbiota in patients with
5(4):359–368. https://​doi.​org/​10.​1080/​21623​945.​2016.​12520​11 Parkinson’s disease. Bull Exp Biol Med 162(6):734–737. https://​
88. Humayun E, Ali U, Shujaat N (2021) Obesity a multifactorial doi.​org/​10.​1007/​s10517-​017-​3700-7
medical problem, presentation to treatment: a systematic review. 107. McFarthing K, Buff S, Rafaloff G, Fiske B, Mursaleen L, Fuest
Prof Med J 28(1):1–8 R, Wyse RK, Stott SRW (2023) Parkinson’s disease drug thera-
89. Friedman SL, Neuschwander-Tetri BA, Rinella M, Sanyal AJ pies in the clinical trial pipeline: 2023 update. J Parkinsons Dis
(2018) Mechanisms of NAFLD development and therapeutic 13(4):427–439. https://​doi.​org/​10.​3233/​JPD-​239901
Neurochemical Research

108. Gorecki AM, Preskey L, Bakeberg MC, Kenna JE, Gildenhuys C, 123. Matenchuk BA, Mandhane PJ, Kozyrskyj AL (2020) Sleep,
MacDougall G, Dunlop SA, Mastaglia FL, Akkari PA, Koengten F, circadian rhythm, and gut microbiota. Sleep Med Rev
Anderton RS (2019) Altered gut microbiome in Parkinson’s disease 53(101340):101340. https://​doi.​org/​10.​1016/j.​smrv.​2020.​101340
and the influence of lipopolysaccharide in a human α-synuclein 124. Frazier K, Chang EB (2020) Intersection of the gut microbiome
over-expressing mouse model. Front Neurosci 13:839. https://​doi.​ and circadian rhythms in metabolism. Trends Endocrinol Metab
org/​10.​3389/​fnins.​2019.​00839 31(1):25–36. https://​doi.​org/​10.​1016/j.​tem.​2019.​08.​013
109. Xilouri M, Stefanis L (2011) Autophagic pathways in parkinson 125. Ouwehand AC, Salminen S, Isolauri E (2002) Probiotics: an
disease and related disorders. Expert Rev Mol Med 13(e8):e8. overview of beneficial effects. Antonie Van Leeuwenhoek
https://​doi.​org/​10.​1017/​S1462​39941​10018​03 82(1–4):279–289
110. Unger MM, Spiegel J, Dillmann K-U, Grundmann D, Philippeit 126. Roberfroid M (2007) Prebiotics: the concept revisited. J Nutr
H, Bürmann J, Faßbender K, Schwiertz A, Schäfer K-H (2016) 137(3):830S-837S. https://​doi.​org/​10.​1093/​jn/​137.3.​830s
Short Chain Fatty acids and gut microbiota differ between patients 127. Aoun A, Darwish F, Hamod N (2020) The influence of the gut
with Parkinson’s disease and age-matched controls. Parkinsonism microbiome on obesity in adults and the role of probiotics, prebiot-
Relat Disord 32:66–72. https://d​ oi.o​ rg/1​ 0.1​ 016/j.p​ arkre​ ldis.2​ 016.​ ics, and synbiotics for weight loss. Prev Nutr Food Sci 25(2):113–
08.​019 123. https://​doi.​org/​10.​3746/​pnf.​2020.​25.2.​113
111. Epp LM, Mravec B (2006) Chronic polysystemic candidiasis as 128. Cerdó T, García-Santos JA, Bermúdez M, Campoy C (2019) The
a possible contributor to onset of idiopathic Parkinson’s disease. role of probiotics and prebiotics in the prevention and treatment of
Bratisl Lek Listy 107:227 obesity. Nutrients 11(3):635
112. Liu M, Bing G (2011) Lipopolysaccharide animal models for Par- 129. Wegh CAM, Geerlings SY, Knol J, Roeselers G, Belzer C (2019)
kinson’s disease. Parkinsons Dis 2011:327089. https://​doi.​org/​10.​ Postbiotics and their potential applications in early life nutrition and
4061/​2011/​327089 beyond. Int J Mol Sci 20(19):4673. https://​doi.​org/​10.​3390/​ijms2​
113. Disease P, Alonso R, Carrasco L, Alonso R, Carrasco LD (2020) 01946​73
Parkinson’s disease: a comprehensive analysis of fungi and bacteria 130. Szydłowska A, Sionek B (2022) Probiotics and postbiotics as the
in brain tissue. Int J Biol Sci 16(7):1135 functional food components affecting the immune response. Micro-
114. Ding H (2017) Gut microbiota and anxiety: an exploration of key organisms 11(1):104. https://​doi.​org/​10.​3390/​micro​organ​isms1​
findings. J Depress Anxiety. https://​doi.​org/​10.​4172/​2167-​1044.​ 10101​04
10002​97 131. Khoruts A, Sadowsky MJ (2016) Understanding the mechanisms
115. Bear TLK, Dalziel JE, Coad J, Roy NC, Butts CA, Gopal PK of faecal microbiota transplantation. Nat Rev Gastroenterol Hepatol
(2020) The role of the gut microbiota in dietary interventions for 13(9):508–516. https://​doi.​org/​10.​1038/​nrgas​tro.​2016.​98
depression and anxiety. Adv Nutr 11(4):890–907. https://​doi.​org/​ 132. Wang S, Chen H, Wen X, Mu J, Sun M, Song X, Liu B, Chen J,
10.​1093/​advan​ces/​nmaa0​16 Fan X (2021) The efficacy of fecal microbiota transplantation in
116. Jiang H-Y, Zhang X, Yu Z-H, Zhang Z, Deng M, Zhao J-H, Ruan B experimental autoimmune encephalomyelitis: transcriptome and
(2018) Altered gut microbiota profile in patients with generalized gut microbiota profiling. J Immunol Res 2021:4400428. https://​
anxiety disorder. J Psychiatr Res 104:130–136. https://​doi.​org/​10.​ doi.​org/​10.​1155/​2021/​44004​28
1016/j.​jpsyc​hires.​2018.​07.​007 133. Borody TJ, Brandt LJ, Paramsothy S (2014) Therapeutic faecal
117. Cheung SG, Goldenthal AR, Uhlemann A-C, Mann JJ, Miller JM, microbiota transplantation: current status and future developments.
Sublette ME (2019) Systematic review of gut microbiota and major Curr Opin Gastroenterol 30(1):97–105. https://​doi.​org/​10.​1097/​
depression. Front Psychiatry 10:34. https://​doi.​org/​10.​3389/​fpsyt.​ MOG.​00000​00000​000027
2019.​00034 134. Zhang X, Cai X, Zheng X (2021) Gut microbiome-oriented therapy
118. Barandouzi ZA, Starkweather AR, Henderson WA, Gyamfi A, for metabolic diseases: challenges and opportunities towards clini-
Cong XS (2020) Altered composition of gut microbiota in depres- cal translation. Trends Pharmacol Sci 42(12):984–987. https://​doi.​
sion: a systematic review. Front Psychiatry 11:541. https://​doi.​org/​ org/​10.​1016/j.​tips.​2021.​09.​003
10.​3389/​fpsyt.​2020.​00541 135. Swerdlow D, Preiss D, Hingorani AD, Sattar N (2015) HMG-coen-
119. Valles-Colomer M, Falony G, Darzi Y, Tigchelaar EF, Wang J, Tito zyme a reductase inhibition, type 2 diabetes, and bodyweight: evi-
RY, Schiweck C, Kurilshikov A, Joossens M, Wijmenga C, Claes dence from genetic analysis and randomised trials. Atherosclerosis
S, Van Oudenhove L, Zhernakova A, Vieira-Silva S, Raes J (2019) 241(1):e17. https://​doi.​org/​10.​1016/j.​ather​oscle​rosis.​2015.​04.​076
The neuroactive potential of the human gut microbiota in quality 136. Crovesy L, Masterson D, Rosado EL (2020) Profile of the gut
of life and depression. Nat Microbiol 4(4):623–632. https://​doi.​org/​ microbiota of adults with obesity: a systematic review. Eur J Clin
10.​1038/​s41564-​018-​0337-x Nutr 74(9):1251–1262. https://​doi.​org/​10.​1038/​s41430-​020-​0607-6
120. Chang L, Wei Y, Hashimoto K (2022) Brain-gut-microbiota axis 137. Huda MSB, Wilding JPH, Pinkney JH (2006) Gut peptides and the
in depression: a historical overview and future directions. Brain regulation of appetite. Obes Rev 7(2):163–182. https://​doi.​org/​10.​
Res Bull 182:44–56. https://​doi.​org/​10.​1016/j.​brain​resbu​ll.​2022.​ 1111/j.​1467-​789X.​2006.​00245.x
02.​004
121. Parkar SG, Kalsbeek A, Cheeseman JF (2019) Potential role for the Publisher's Note Springer Nature remains neutral with regard to
gut microbiota in modulating host circadian rhythms and metabolic jurisdictional claims in published maps and institutional affiliations.
health. Microorganisms 7(2):41. https://​doi.​org/​10.​3390/​micro​
organ​isms7​020041 Springer Nature or its licensor (e.g. a society or other partner) holds
122. Bishehsari F, Voigt RM, Keshavarzian A (2020) Circadian rhythms exclusive rights to this article under a publishing agreement with the
and the gut microbiota: from the metabolic syndrome to cancer. author(s) or other rightsholder(s); author self-archiving of the accepted
Nat Rev Endocrinol 16(12):731–739. https://​doi.​org/​10.​1038/​ manuscript version of this article is solely governed by the terms of
s41574-​020-​00427-4 such publishing agreement and applicable law.

You might also like