Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46.092
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 42(1): 153-178, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38941602

ABSTRACT

The intestine is the largest peripheral lymphoid organ in animals, including humans, and interacts with a vast array of microorganisms called the gut microbiota. Comprehending the symbiotic relationship between the gut microbiota and our immune system is essential not only for the field of immunology but also for understanding the pathogenesis of various systemic diseases, including cancer, cardiometabolic disorders, and extraintestinal autoimmune conditions. Whereas microbe-derived antigens are crucial for activating the intestinal immune system, particularly T and B cells, as environmental cues, microbes and their metabolites play a critical role in directing the differentiation of these immune cells. Microbial metabolites are regarded as messengers from the gut microbiota, since bacteria have the ability to produce unique molecules that humans cannot, and many immune cells in the intestine express receptors for these molecules. This review highlights the distinct relationships between microbial metabolites and the differentiation and function of the immune system.


Subject(s)
Gastrointestinal Microbiome , Humans , Animals , Gastrointestinal Microbiome/immunology , Cell Differentiation , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Bacteria/immunology , Bacteria/metabolism
2.
Annu Rev Immunol ; 42(1): 259-288, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38277692

ABSTRACT

Gastrointestinal nematode (GIN) infection has applied significant evolutionary pressure to the mammalian immune system and remains a global economic and human health burden. Upon infection, type 2 immune sentinels activate a common antihelminth response that mobilizes and remodels the intestinal tissue for effector function; however, there is growing appreciation of the impact GIN infection also has on the distal tissue immune state. Indeed, this effect is observed even in tissues through which GINs never transit. This review highlights how GIN infection modulates systemic immunity through (a) induction of host resistance and tolerance responses, (b) secretion of immunomodulatory products, and (c) interaction with the intestinal microbiome. It also discusses the direct consequences that changes to distal tissue immunity can have for concurrent and subsequent infection, chronic noncommunicable diseases, and vaccination efficacy.


Subject(s)
Gastrointestinal Microbiome , Nematoda , Nematode Infections , Animals , Humans , Nematode Infections/immunology , Nematoda/immunology , Nematoda/physiology , Gastrointestinal Microbiome/immunology , Immunomodulation , Host-Parasite Interactions/immunology , Intestinal Diseases, Parasitic/immunology , Immune Tolerance , Gastrointestinal Tract/immunology , Gastrointestinal Tract/parasitology
3.
Annu Rev Immunol ; 42(1): 489-519, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38941607

ABSTRACT

Recent advances have contributed to a mechanistic understanding of neuroimmune interactions in the intestine and revealed an essential role of this cross talk for gut homeostasis and modulation of inflammatory and infectious intestinal diseases. In this review, we describe the innervation of the intestine by intrinsic and extrinsic neurons and then focus on the bidirectional communication between neurons and immune cells. First, we highlight the contribution of neuronal subtypes to the development of colitis and discuss the different immune and epithelial cell types that are regulated by neurons via the release of neuropeptides and neurotransmitters. Next, we review the role of intestinal inflammation in the development of visceral hypersensitivity and summarize how inflammatory mediators induce peripheral and central sensitization of gut-innervating sensory neurons. Finally, we outline the importance of immune cells and gut microbiota for the survival and function of different neuronal populations at homeostasis and during bacterial and helminth infection.


Subject(s)
Neuroimmunomodulation , Humans , Animals , Intestines/immunology , Homeostasis , Gastrointestinal Microbiome/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Neurons/metabolism , Neurons/immunology , Neuropeptides/metabolism , Enteric Nervous System/immunology , Enteric Nervous System/metabolism
4.
Annu Rev Immunol ; 40: 143-167, 2022 04 26.
Article in English | MEDLINE | ID: mdl-34990209

ABSTRACT

The gut microbiome influences many host physiologies, spanning gastrointestinal function, metabolism, immune homeostasis, neuroactivity, and behavior. Many microbial effects on the host are orchestrated by bidirectional interactions between the microbiome and immune system. Imbalances in this dialogue can lead to immune dysfunction and immune-mediated conditions in distal organs including the brain. Dysbiosis of the gut microbiome and dysregulated neuroimmune responses are common comorbidities of neurodevelopmental, neuropsychiatric, and neurological disorders, highlighting the importance of the gut microbiome-neuroimmune axis as a regulator of central nervous system homeostasis. In this review, we discuss recent evidence supporting a role for the gut microbiome in regulating the neuroimmune landscape in health and disease.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Animals , Brain , Dysbiosis , Humans , Neuroimmunomodulation
5.
Annu Rev Immunol ; 38: 649-671, 2020 04 26.
Article in English | MEDLINE | ID: mdl-32040356

ABSTRACT

A plethora of experimental and epidemiological evidence supports a critical role for inflammation and adaptive immunity in the onset of cancer and in shaping its response to therapy. These data are particularly robust for gastrointestinal (GI) cancers, such as those affecting the GI tract, liver, and pancreas, on which this review is focused. We propose a unifying hypothesis according to which intestinal barrier disruption is the origin of tumor-promoting inflammation that acts in conjunction with tissue-specific cancer-initiating mutations. The gut microbiota and its products impact tissue-resident and recruited myeloid cells that promote tumorigenesis through secretion of growth- and survival-promoting cytokines that act on epithelial cells, as well as fibrogenic and immunosuppressive cytokines that interfere with the proper function of adaptive antitumor immunity. Understanding these relationships should improve our ability to prevent cancer development and stimulate the immune system to eliminate existing malignancies.


Subject(s)
Gastric Mucosa/immunology , Gastric Mucosa/metabolism , Gastrointestinal Microbiome , Gastrointestinal Neoplasms/etiology , Gastrointestinal Neoplasms/metabolism , Host-Pathogen Interactions/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Adaptive Immunity , Animals , Gastric Mucosa/pathology , Gastrointestinal Microbiome/immunology , Gastrointestinal Neoplasms/pathology , Humans , Immunity, Innate , Intestinal Mucosa/pathology , Liver/immunology , Liver/metabolism , Liver/pathology
6.
Annu Rev Immunol ; 38: 23-48, 2020 04 26.
Article in English | MEDLINE | ID: mdl-32340570

ABSTRACT

The gastrointestinal tract harbors numerous commensal bacteria, referred to as the microbiota, that benefit host health by digesting dietary components and eliminating pathogens. The intestinal microbiota maintains epithelial barrier integrity and shapes the mucosal immune system, balancing host defense and oral tolerance with microbial metabolites, components, and attachment to host cells. To avoid aberrant immune responses, epithelial cells segregate the intestinal microbiota from immune cells by constructing chemical and physical barriers, leading to the establishment of host-commensal mutualism. Furthermore, intestinal immune cells participate in the maintenance of a healthy microbiota community and reinforce epithelial barrier functions. Perturbations of the microbiota composition are commonly observed in patients with autoimmune diseases and chronic inflammatory disorders. An understanding of the intimate interactions between the intestinal microbiota, epithelial cells, and immune cells that are crucial for the maintenance of intestinal homeostasis might promote advances in diagnostic and therapeutic approaches for various diseases.


Subject(s)
Gastrointestinal Microbiome/immunology , Host-Pathogen Interactions/immunology , Immunity, Mucosal , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Animals , Disease Susceptibility , Humans , Immune System/immunology , Immune System/metabolism , Peyer's Patches/immunology , Peyer's Patches/metabolism
7.
Annu Rev Immunol ; 37: 377-403, 2019 04 26.
Article in English | MEDLINE | ID: mdl-31026410

ABSTRACT

The gut-associated lymphoid tissue (GALT) faces a considerable challenge. It encounters antigens derived from an estimated 1014 commensal microbes and greater than 30 kg of food proteins yearly. It must distinguish these harmless antigens from potential pathogens and mount the appropriate host immune response. Local and systemic hyporesponsiveness to dietary antigens, classically referred to as oral tolerance, comprises a distinct complement of adaptive cellular and humoral immune responses. It is increasingly evident that a functional epithelial barrier engaged in intimate interplay with innate immune cells and the resident microbiota is critical to establishing and maintaining oral tolerance. Moreover, innate immune cells serve as a bridge between the microbiota, epithelium, and the adaptive immune system, parlaying tonic microbial stimulation into signals critical for mucosal homeostasis. Dysregulation of gut homeostasis and the subsequent disruption of tolerance therefore have clinically significant consequences for the development of food allergy.


Subject(s)
Dysbiosis/immunology , Food Hypersensitivity/immunology , Gastrointestinal Microbiome/immunology , Intestinal Mucosa/immunology , Administration, Oral , Allergens/immunology , Animals , Food , Food Hypersensitivity/microbiology , Homeostasis , Humans , Immune Tolerance , Immunity, Innate , Intestinal Mucosa/microbiology
8.
Annu Rev Immunol ; 37: 599-624, 2019 04 26.
Article in English | MEDLINE | ID: mdl-31026411

ABSTRACT

The intestinal microbiota plays a crucial role in influencing the development of host immunity, and in turn the immune system also acts to regulate the microbiota through intestinal barrier maintenance and immune exclusion. Normally, these interactions are homeostatic, tightly controlled, and organized by both innate and adaptive immune responses. However, a combination of environmental exposures and genetic defects can result in a break in tolerance and intestinal homeostasis. The outcomes of these interactions at the mucosal interface have broad, systemic effects on host immunity and the development of chronic inflammatory or autoimmune disease. The underlying mechanisms and pathways the microbiota can utilize to regulate these diseases are just starting to emerge. Here, we discuss the recent evidence in this area describing the impact of microbiota-immune interactions during inflammation and autoimmunity, with a focus on barrier function and CD4+ T cell regulation.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/microbiology , Gastrointestinal Microbiome/immunology , Inflammation/microbiology , Inflammatory Bowel Diseases/microbiology , Intestinal Mucosa/microbiology , Animals , Autoimmunity , Diabetes Mellitus, Type 1/immunology , Homeostasis , Humans , Immune Tolerance , Immunomodulation , Inflammation/immunology , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/immunology
9.
Annu Rev Immunol ; 36: 359-381, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29400985

ABSTRACT

IgA is the dominant immunoglobulin isotype produced in mammals, largely secreted across the intestinal mucosal surface. Although induction of IgA has been a hallmark feature of microbiota colonization following colonization in germ-free animals, until recently appreciation of the function of IgA in host-microbial mutualism has depended mainly on indirect evidence of alterations in microbiota composition or penetration of microbes in the absence of somatic mutations in IgA (or compensatory IgM). Highly parallel sequencing techniques that enable high-resolution analysis of either microbial consortia or IgA sequence diversity are now giving us new perspectives on selective targeting of microbial taxa and the trajectory of IgA diversification according to induction mechanisms, between different individuals and over time. The prospects are to link the range of diversified IgA clonotypes to specific antigenic functions in modulating the microbiota composition, position and metabolism to ensure host mutualism.


Subject(s)
Gastrointestinal Microbiome/immunology , Immunoglobulin A/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Age Factors , Animals , Disease Susceptibility , Host-Pathogen Interactions/immunology , Humans , Intestinal Mucosa/metabolism , Protein Binding
10.
Annu Rev Immunol ; 36: 755-781, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29677472

ABSTRACT

Inflammatory bowel disease (IBD) defines a spectrum of complex disorders. Understanding how environmental risk factors, alterations of the intestinal microbiota, and polygenetic and epigenetic susceptibility impact on immune pathways is key for developing targeted therapies. Mechanistic understanding of polygenic IBD is complemented by Mendelian disorders that present with IBD, pharmacological interventions that cause colitis, autoimmunity, and multiple animal models. Collectively, this multifactorial pathogenesis supports a concept of immune checkpoints that control microbial-host interactions in the gut by modulating innate and adaptive immunity, as well as epithelial and mesenchymal cell responses. In addition to classical immunosuppressive strategies, we discuss how resetting the microbiota and restoring innate immune responses, in particular autophagy and epithelial barrier function, might be key for maintaining remission or preventing IBD. Targeting checkpoints in genetically stratified subgroups of patients with Mendelian disorder-associated IBD increasingly directs treatment strategies as part of personalized medicine.


Subject(s)
Disease Susceptibility/immunology , Inflammatory Bowel Diseases/etiology , Inflammatory Bowel Diseases/therapy , Animals , Biomarkers , Chronic Disease , Disease Management , Disease Models, Animal , Drug-Related Side Effects and Adverse Reactions , Dysbiosis , Gastrointestinal Microbiome , Genetic Predisposition to Disease , Humans , Inflammatory Bowel Diseases/prevention & control , Molecular Targeted Therapy , Translational Research, Biomedical
11.
Annu Rev Immunol ; 35: 371-402, 2017 04 26.
Article in English | MEDLINE | ID: mdl-28446062

ABSTRACT

Nutrition and the gut microbiome regulate many systems, including the immune, metabolic, and nervous systems. We propose that the host responds to deficiency (or sufficiency) of dietary and bacterial metabolites in a dynamic way, to optimize responses and survival. A family of G protein-coupled receptors (GPCRs) termed the metabolite-sensing GPCRs bind to various metabolites and transmit signals that are important for proper immune and metabolic functions. Members of this family include GPR43, GPR41, GPR109A, GPR120, GPR40, GPR84, GPR35, and GPR91. In addition, bile acid receptors such as GPR131 (TGR5) and proton-sensing receptors such as GPR65 show similar features. A consistent feature of this family of GPCRs is that they provide anti-inflammatory signals; many also regulate metabolism and gut homeostasis. These receptors represent one of the main mechanisms whereby the gut microbiome affects vertebrate physiology, and they also provide a link between the immune and metabolic systems. Insufficient signaling through one or more of these metabolite-sensing GPCRs likely contributes to human diseases such as asthma, food allergies, type 1 and type 2 diabetes, hepatic steatosis, cardiovascular disease, and inflammatory bowel diseases.


Subject(s)
Cardiovascular Diseases/immunology , Diabetes Mellitus, Type 1/immunology , Gastrointestinal Microbiome/immunology , Hypersensitivity/immunology , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Diet , Homeostasis , Humans , Immunity , Receptors, G-Protein-Coupled/immunology
12.
Annu Rev Immunol ; 35: 119-147, 2017 04 26.
Article in English | MEDLINE | ID: mdl-28125357

ABSTRACT

The intestinal epithelial barrier includes columnar epithelial, Paneth, goblet, enteroendocrine, and tuft cells as well as other cell populations, all of which contribute properties essential for gastrointestinal homeostasis. The intestinal mucosa is covered by mucin, which contains antimicrobial peptides and secretory IgA and prevents luminal bacteria, fungi, and viruses from stimulating intestinal immune responses. Conversely, the transport of luminal microorganisms-mediated by M, dendritic, and goblet cells-into intestinal tissues facilitates the harmonization of active and quiescent mucosal immune responses. The bacterial population within gut-associated lymphoid tissues creates the intratissue cohabitations for harmonized mucosal immunity. Intermolecular and intercellular communication among epithelial, immune, and mesenchymal cells creates an environment conducive for epithelial regeneration and mucosal healing. This review summarizes the so-called intestinal mucosal ecological network-the complex but vital molecular and cellular interactions of epithelial mesenchymal cells, immune cells, and commensal microbiota that achieve intestinal homeostasis, regeneration, and healing.


Subject(s)
Epithelial Cells/physiology , Gastrointestinal Microbiome/immunology , Intestinal Mucosa/immunology , Animals , Cell Communication , Homeostasis , Humans , Immunity, Innate , Immunoglobulin A/metabolism , Intestinal Mucosa/pathology , Wound Healing
13.
Cell ; 187(13): 3231-3232, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38906099

ABSTRACT

Numerous studies have evaluated the gut microbiome as a biomarker for predicting cancer immunotherapy, but the heterogeneity among different studies has hindered its applications. In this issue of Cell, Derosa et al. report a biomarker based on the ecological topology of the gut microbiota that can predict immunotherapy efficacy effectively.


Subject(s)
Gastrointestinal Microbiome , Immunotherapy , Neoplasms , Periodicals as Topic , Humans , Biomarkers/metabolism , Biomarkers, Tumor/metabolism , Gastrointestinal Microbiome/immunology , Immunotherapy/methods , Neoplasms/therapy , Neoplasms/immunology
14.
Cell ; 187(12): 2903-2904, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38848674

ABSTRACT

Gut microbes are known to impact host physiology in several ways. However, key molecular players in host-commensal interactions remain to be uncovered. In this issue of Cell, McCurry et al. reveal that gut bacteria perform 21-dehydroxylation to convert abundant biliary corticoids to neurosteroids using readily available H2 in their environment.


Subject(s)
Gastrointestinal Microbiome , Humans , Animals , Bacteria/metabolism , Neurotransmitter Agents/metabolism
15.
Cell ; 187(7): 1801-1818.e20, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38471500

ABSTRACT

The repertoire of modifications to bile acids and related steroidal lipids by host and microbial metabolism remains incompletely characterized. To address this knowledge gap, we created a reusable resource of tandem mass spectrometry (MS/MS) spectra by filtering 1.2 billion publicly available MS/MS spectra for bile-acid-selective ion patterns. Thousands of modifications are distributed throughout animal and human bodies as well as microbial cultures. We employed this MS/MS library to identify polyamine bile amidates, prevalent in carnivores. They are present in humans, and their levels alter with a diet change from a Mediterranean to a typical American diet. This work highlights the existence of many more bile acid modifications than previously recognized and the value of leveraging public large-scale untargeted metabolomics data to discover metabolites. The availability of a modification-centric bile acid MS/MS library will inform future studies investigating bile acid roles in health and disease.


Subject(s)
Bile Acids and Salts , Gastrointestinal Microbiome , Metabolomics , Tandem Mass Spectrometry , Animals , Humans , Bile Acids and Salts/chemistry , Metabolomics/methods , Polyamines , Tandem Mass Spectrometry/methods , Databases, Chemical
16.
Cell ; 187(1): 62-78.e20, 2024 01 04.
Article in English | MEDLINE | ID: mdl-38096822

ABSTRACT

The microbiota influences intestinal health and physiology, yet the contributions of commensal protists to the gut environment have been largely overlooked. Here, we discover human- and rodent-associated parabasalid protists, revealing substantial diversity and prevalence in nonindustrialized human populations. Genomic and metabolomic analyses of murine parabasalids from the genus Tritrichomonas revealed species-level differences in excretion of the metabolite succinate, which results in distinct small intestinal immune responses. Metabolic differences between Tritrichomonas species also determine their ecological niche within the microbiota. By manipulating dietary fibers and developing in vitro protist culture, we show that different Tritrichomonas species prefer dietary polysaccharides or mucus glycans. These polysaccharide preferences drive trans-kingdom competition with specific commensal bacteria, which affects intestinal immunity in a diet-dependent manner. Our findings reveal unappreciated diversity in commensal parabasalids, elucidate differences in commensal protist metabolism, and suggest how dietary interventions could regulate their impact on gut health.


Subject(s)
Gastrointestinal Microbiome , Parabasalidea , Polysaccharides , Animals , Humans , Mice , Dietary Fiber , Intestine, Small/metabolism , Polysaccharides/metabolism , Parabasalidea/metabolism , Dietary Carbohydrates/metabolism , Biodiversity
17.
Cell ; 187(5): 1191-1205.e15, 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38366592

ABSTRACT

Carbohydrate intolerance, commonly linked to the consumption of lactose, fructose, or sorbitol, affects up to 30% of the population in high-income countries. Although sorbitol intolerance is attributed to malabsorption, the underlying mechanism remains unresolved. Here, we show that a history of antibiotic exposure combined with high fat intake triggered long-lasting sorbitol intolerance in mice by reducing Clostridia abundance, which impaired microbial sorbitol catabolism. The restoration of sorbitol catabolism by inoculation with probiotic Escherichia coli protected mice against sorbitol intolerance but did not restore Clostridia abundance. Inoculation with the butyrate producer Anaerostipes caccae restored a normal Clostridia abundance, which protected mice against sorbitol-induced diarrhea even when the probiotic was cleared. Butyrate restored Clostridia abundance by stimulating epithelial peroxisome proliferator-activated receptor-gamma (PPAR-γ) signaling to restore epithelial hypoxia in the colon. Collectively, these mechanistic insights identify microbial sorbitol catabolism as a potential target for approaches for the diagnosis, treatment, and prevention of sorbitol intolerance.


Subject(s)
Carbohydrate Metabolism, Inborn Errors , Gastrointestinal Microbiome , Sorbitol , Animals , Mice , Anti-Bacterial Agents/pharmacology , Butyrates , Clostridium , Escherichia coli , Sorbitol/metabolism
18.
Cell ; 187(8): 1853-1873.e15, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38574728

ABSTRACT

This study has followed a birth cohort for over 20 years to find factors associated with neurodevelopmental disorder (ND) diagnosis. Detailed, early-life longitudinal questionnaires captured infection and antibiotic events, stress, prenatal factors, family history, and more. Biomarkers including cord serum metabolome and lipidome, human leukocyte antigen (HLA) genotype, infant microbiota, and stool metabolome were assessed. Among the 16,440 Swedish children followed across time, 1,197 developed an ND. Significant associations emerged for future ND diagnosis in general and for specific ND subtypes, spanning intellectual disability, speech disorder, attention-deficit/hyperactivity disorder, and autism. This investigation revealed microbiome connections to future diagnosis as well as early emerging mood and gastrointestinal problems. The findings suggest links to immunodysregulation and metabolism, compounded by stress, early-life infection, and antibiotics. The convergence of infant biomarkers and risk factors in this prospective, longitudinal study on a large-scale population establishes a foundation for early-life prediction and intervention in neurodevelopment.


Subject(s)
Biomarkers , Gastrointestinal Microbiome , Neurodevelopmental Disorders , Child , Female , Humans , Infant , Pregnancy , Autism Spectrum Disorder/microbiology , Longitudinal Studies , Prospective Studies , Feces/microbiology , Mood Disorders/microbiology
19.
Cell ; 187(3): 750-763.e20, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38242132

ABSTRACT

Breastfeeding offers demonstrable benefits to newborns and infants by providing nourishment and immune protection and by shaping the gut commensal microbiota. Although it has been appreciated for decades that breast milk contains complement components, the physiological relevance of complement in breast milk remains undefined. Here, we demonstrate that weanling mice fostered by complement-deficient dams rapidly succumb when exposed to murine pathogen Citrobacter rodentium (CR), whereas pups fostered on complement-containing milk from wild-type dams can tolerate CR challenge. The complement components in breast milk were shown to directly lyse specific members of gram-positive gut commensal microbiota via a C1-dependent, antibody-independent mechanism, resulting in the deposition of the membrane attack complex and subsequent bacterial lysis. By selectively eliminating members of the commensal gut community, complement components from breast milk shape neonate and infant gut microbial composition to be protective against environmental pathogens such as CR.


Subject(s)
Complement System Proteins , Gastrointestinal Microbiome , Milk , Animals , Female , Humans , Infant , Mice , Bacteria , Breast Feeding , Citrobacter rodentium , Complement System Proteins/analysis , Immunologic Factors , Infant Health , Milk, Human , Milk/chemistry , Enterobacteriaceae Infections/immunology
20.
Cell ; 187(11): 2687-2689, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38788691

ABSTRACT

In this issue of Cell, Nie and co-authors report that the microbe-derived bile acid (BA) 3-succinylated cholic acid protects against the progression of metabolic dysfunction-associated liver disease. Intriguingly, its protective mechanism does not involve traditional BA signaling pathways but is instead linked to the proliferation of the commensal microbe Akkermansia muciniphila.


Subject(s)
Akkermansia , Bile Acids and Salts , Periodicals as Topic , Animals , Humans , Mice , Akkermansia/metabolism , Bile Acids and Salts/metabolism , Cholic Acid/metabolism , Gastrointestinal Microbiome , Liver/metabolism , Liver Diseases/metabolism , Liver Diseases/microbiology , Verrucomicrobia/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL