Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.310
Filter
1.
Int J Mol Sci ; 25(17)2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39273087

ABSTRACT

Activating enhancer-binding protein 2 (AP-2) is a family of transcription factors (TFs) that play crucial roles in regulating embryonic and oncogenic development. In addition to splice isoforms, five major family members encoded by the TFAP2A/B/C/D/E genes have been identified in humans, i.e., AP-2α/ß/γ/δ/ε. In general, the first three TFs have been studied more thoroughly than AP-2δ or AP-2ε. Currently, there is a relatively limited body of literature focusing on the AP-2 family in the context of gastroenterological research, and a comprehensive overview of the existing knowledge and recommendations for further research directions is lacking. Herein, we have collected available gastroenterological data on AP-2 TFs, discussed the latest medical applications of each family member, and proposed potential future directions. Research on AP-2 in gastrointestinal tumors has predominantly been focused on the two best-described family members, AP-2α and AP-2γ. Surprisingly, research in the past decade has highlighted the importance of AP-2ε in the drug resistance of gastric cancer (GC) and colorectal cancer (CRC). While numerous questions about gastroenterological disorders await elucidation, the available data undoubtedly open avenues for anti-cancer targeted therapy and overcoming chemotherapy resistance. In addition to gastrointestinal cancers, AP-2 family members (primarily AP-2ß and marginally AP-2γ) have been associated with other health issues such as obesity, type 2 diabetes, liver dysfunction, and pseudo-obstruction. On the other hand, AP-2δ has been poorly investigated in gastroenterological disorders, necessitating further research to delineate its role. In conclusion, despite the limited attention given to AP-2 in gastroenterology research, pivotal functions of these transcription factors have started to emerge and warrant further exploration in the future.


Subject(s)
Transcription Factor AP-2 , Humans , Transcription Factor AP-2/metabolism , Transcription Factor AP-2/genetics , Gastrointestinal Diseases/genetics , Gastrointestinal Diseases/metabolism , Animals
2.
Proc Natl Acad Sci U S A ; 121(39): e2406479121, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39284050

ABSTRACT

Parkinson's disease (PD) is typically a sporadic late-onset disorder, which has made it difficult to model in mice. Several transgenic mouse models bearing mutations in SNCA, which encodes alpha-Synuclein (α-Syn), have been made, but these lines do not express SNCA in a physiologically accurate spatiotemporal pattern, which limits the ability of the mice to recapitulate the features of human PD. Here, we generated knock-in mice bearing the G51D SNCA mutation. After establishing that their motor symptoms begin at 9 mo of age, we then sought earlier pathologies. We assessed the phosphorylation at Serine 129 of α-Syn in different tissues and detected phospho-α-Syn in the olfactory bulb and enteric nervous system at 3 mo of age. Olfactory deficit and impaired gut transit followed at 6 mo, preceding motor symptoms. The SncaG51D mice thus parallel the progression of human PD and will enable us to study PD pathogenesis and test future therapies.


Subject(s)
Disease Models, Animal , Gene Knock-In Techniques , Parkinson Disease , alpha-Synuclein , Animals , alpha-Synuclein/metabolism , alpha-Synuclein/genetics , Mice , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/physiopathology , Parkinson Disease/pathology , Mice, Transgenic , Phosphorylation , Olfaction Disorders/genetics , Olfaction Disorders/metabolism , Olfaction Disorders/physiopathology , Olfactory Bulb/metabolism , Olfactory Bulb/pathology , Gastrointestinal Diseases/genetics , Gastrointestinal Diseases/metabolism , Gastrointestinal Diseases/pathology , Enteric Nervous System/metabolism , Enteric Nervous System/physiopathology , Humans , Male
3.
Front Immunol ; 15: 1431495, 2024.
Article in English | MEDLINE | ID: mdl-39206201

ABSTRACT

Gastrointestinal mucosal surface is frequently under challenge due to it's the large surface area and most common entry of microbes. IL-37, an anti-inflammatory cytokine, regulates local and systemic host immunity. H. pylori infection leads to the inhibition of IL-37 in the gastric mucosa, contributing to heightened mucosal inflammation and destruction, thereby facilitating increased proliferation of H. pylori. Food allergy, due to immune dysregulation, also contribute to GI injury. On the other hand, elevated levels of IL-37 observed in gastric cancer patients align with reduced host immunity at the cellular and humoral levels, indicating that IL-37 may contribute to the development of gastric cancer via suppressing pro-inflammatory responses. While IL-37 provides protection in an IBD animal model, the detection of highly produced IL-37 in IBD patients suggests a stage-dependent role, being protective in acute inflammation but potentially exacerbates the development of IBD in chronic conditions. Moreover, elevated colonic IL-37 in CRC correlates with overall survival time and disease time, indicating a protective role for IL-37 in CRC. The differential regulation and expression of IL-37 between upper- and lower-GI organs may be attributed to variations in the microbial flora. This information suggests that IL-37 could be a potential therapeutic agent, depending on the stage and location.


Subject(s)
Gastrointestinal Diseases , Interleukin-1 , Humans , Interleukin-1/metabolism , Animals , Gastrointestinal Diseases/immunology , Gastrointestinal Diseases/metabolism , Helicobacter Infections/immunology , Helicobacter Infections/metabolism , Gastric Mucosa/metabolism , Gastric Mucosa/immunology , Gastric Mucosa/microbiology , Helicobacter pylori/immunology , Gastrointestinal Microbiome/immunology
4.
World J Gastroenterol ; 30(27): 3273-3277, 2024 Jul 21.
Article in English | MEDLINE | ID: mdl-39086749

ABSTRACT

In this editorial, we comment on three articles published in a recent issue of World Journal of Gastroenterology. There is a pressing need for new research on autophagy's role in gastrointestinal (GI) disorders, and also novel insights into some liver conditions, such as metabolic dysfunction-associated fatty liver disease (MAFLD) and acute liver failure (ALF). Despite advancements, understanding autophagy's intricate mechanisms and implications in these diseases remains incomplete. Moreover, MAFLD's pathogenesis, encompassing hepatic steatosis and metabolic dysregulation, require further elucidation. Similarly, the mechanisms underlying ALF, a severe hepatic dysfunction, are poorly understood. Innovative studies exploring the interplay between autophagy and GI disorders, as well as defined mechanisms of MAFLD and ALF, are crucial for identifying therapeutic targets and enhancing diagnostic and treatment strategies to mitigate the global burden of these diseases.


Subject(s)
Autophagy , Liver Failure, Acute , Humans , Liver Failure, Acute/metabolism , Liver Failure, Acute/pathology , Liver Failure, Acute/etiology , Liver/pathology , Liver/metabolism , Animals , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Gastrointestinal Diseases/metabolism , Gastrointestinal Diseases/pathology , Gastrointestinal Diseases/etiology , Fatty Liver/metabolism , Fatty Liver/pathology
5.
Postepy Biochem ; 69(4): 298-309, 2024 01 30.
Article in Polish | MEDLINE | ID: mdl-39012695

ABSTRACT

The main function of bile acids (BA) is participation in the emulsification of dietary fats. Recently it has been discovered that BAs can also act as signaling molecules regulating the processes of their own synthesis and metabolism, as well as glucose and lipid metabolism. In addition, they affect the motility of the digestive tract and food intake. BA also interacts with the gut microbiota, a major player in their metabolism. Most of the regulatory actions of BAs are mediated by their receptors, the most important of which are the farnesoid X receptor (FXR) and the G protein-coupled receptor -TGR5, found in large amounts in the intestine, liver, adipose tissue and other tissues of the body. Recently, much attention has been paid to the influence of BA on various diseases and the possibility of using them in the treatment of e.g. inflammatory bowel disease, liver diseases, type 2 diabetes and obesity.


Subject(s)
Bile Acids and Salts , Diabetes Mellitus, Type 2 , Gastrointestinal Microbiome , Obesity , Humans , Bile Acids and Salts/metabolism , Obesity/metabolism , Obesity/drug therapy , Gastrointestinal Microbiome/physiology , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Animals , Receptors, Cytoplasmic and Nuclear/metabolism , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/metabolism , Gastrointestinal Diseases/drug therapy , Gastrointestinal Diseases/metabolism , Receptors, G-Protein-Coupled/metabolism , Lipid Metabolism/physiology
6.
Nat Commun ; 15(1): 5543, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39019879

ABSTRACT

Meconium, a non-invasive biomaterial reflecting prenatal substance accumulation, could provide valuable insights into neonatal health. However, the comprehensive protein profile of meconium across gestational ages remains unclear. Here, we conducted an extensive proteomic analysis of first meconium from 259 newborns across varied gestational ages to delineate protein composition and elucidate its relevance to neonatal diseases. The first meconium samples were collected, with the majority obtained before feeding, and the mean time for the first meconium passage from the anus was 11.9 ± 9.47 h. Our analysis revealed 5370 host-derived meconium proteins, which varied depending on sex and gestational age. Specifically, meconium from preterm infants exhibited elevated concentrations of proteins associated with the extracellular matrix. Additionally, the protein profiles of meconium also exhibited unique variations depending on both specific diseases, including gastrointestinal diseases, congenital heart diseases, and maternal conditions. Furthermore, we developed a machine learning model to predict gestational ages using meconium proteins. Our model suggests that newborns with gastrointestinal diseases and congenital heart diseases may have immature gastrointestinal systems. These findings highlight the intricate relationship between clinical parameters and meconium protein composition, offering potential for a novel approach to assess neonatal gastrointestinal health.


Subject(s)
Gestational Age , Machine Learning , Meconium , Proteomics , Humans , Meconium/metabolism , Infant, Newborn , Female , Male , Proteomics/methods , Infant, Premature/metabolism , Gastrointestinal Diseases/metabolism , Heart Defects, Congenital/metabolism , Pregnancy , Proteome/metabolism
7.
Biomolecules ; 14(7)2024 Jul 07.
Article in English | MEDLINE | ID: mdl-39062518

ABSTRACT

The gastrointestinal (GI) tract is an organ actively involved in mechanical processes, where it detects forces via a mechanosensation mechanism. Mechanosensation relies on specialized cells termed mechanoreceptors, which convert mechanical forces into electrochemical signals via mechanosensors. The mechanosensitive Piezo1 and Piezo2 are widely expressed in various mechanosensitive cells that respond to GI mechanical forces by altering transmembrane ionic currents, such as epithelial cells, enterochromaffin cells, and intrinsic and extrinsic enteric neurons. This review highlights recent research advances on mechanosensitive Piezo channels in GI physiology and pathology. Specifically, the latest insights on the role of Piezo channels in the intestinal barrier, GI motility, and intestinal mechanosensation are summarized. Additionally, an overview of Piezo channels in the pathogenesis of GI disorders, including irritable bowel syndrome, inflammatory bowel disease, and GI cancers, is provided. Overall, the presence of mechanosensitive Piezo channels offers a promising new perspective for the treatment of various GI disorders.


Subject(s)
Gastrointestinal Tract , Ion Channels , Mechanotransduction, Cellular , Humans , Ion Channels/metabolism , Animals , Gastrointestinal Tract/metabolism , Gastrointestinal Diseases/metabolism , Gastrointestinal Diseases/physiopathology , Gastrointestinal Diseases/pathology , Gastrointestinal Motility/physiology
8.
World J Gastroenterol ; 30(23): 2934-2946, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38946875

ABSTRACT

In this editorial, we comment on an article titled "Morphological and biochemical characteristics associated with autophagy in gastrointestinal diseases", which was published in a recent issue of the World Journal of Gastroenterology. We focused on the statement that "autophagy is closely related to the digestion, secretion, and regeneration of gastrointestinal cells". With advancing research, autophagy, and particularly the pivotal role of the macroautophagy in maintaining cellular equilibrium and stress response in the gastrointestinal system, has garnered extensive study. However, the significance of mitophagy, a unique selective autophagy pathway with ubiquitin-dependent and independent variants, should not be overlooked. In recent decades, mitophagy has been shown to be closely related to the occurrence and development of gastrointestinal diseases, especially inflammatory bowel disease, gastric cancer, and colorectal cancer. The interplay between mitophagy and mitochondrial quality control is crucial for elucidating disease mechanisms, as well as for the development of novel treatment strategies. Exploring the pathogenesis behind gastrointestinal diseases and providing individualized and efficient treatment for patients are subjects we have been exploring. This article reviews the potential mechanism of mitophagy in gastrointestinal diseases with the hope of providing new ideas for diagnosis and treatment.


Subject(s)
Autophagy , Gastrointestinal Diseases , Mitochondria , Mitophagy , Humans , Autophagy/physiology , Gastrointestinal Diseases/pathology , Gastrointestinal Diseases/metabolism , Gastrointestinal Diseases/physiopathology , Mitochondria/metabolism , Mitochondria/pathology , Gastrointestinal Tract/pathology , Gastrointestinal Tract/metabolism , Animals
9.
Int J Mol Sci ; 25(12)2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38928387

ABSTRACT

Cannabinoids and their receptors play a significant role in the regulation of gastrointestinal (GIT) peristalsis and intestinal barrier permeability. This review critically evaluates current knowledge about the mechanisms of action and biological effects of endocannabinoids and phytocannabinoids on GIT functions and the potential therapeutic applications of these compounds. The results of ex vivo and in vivo preclinical data indicate that cannabinoids can both inhibit and stimulate gut peristalsis, depending on various factors. Endocannabinoids affect peristalsis in a cannabinoid (CB) receptor-specific manner; however, there is also an important interaction between them and the transient receptor potential cation channel subfamily V member 1 (TRPV1) system. Phytocannabinoids such as Δ9-tetrahydrocannabinol (THC) and cannabidiol (CBD) impact gut motility mainly through the CB1 receptor. They were also found to improve intestinal barrier integrity, mainly through CB1 receptor stimulation but also via protein kinase A (PKA), mitogen-associated protein kinase (MAPK), and adenylyl cyclase signaling pathways, as well as by influencing the expression of tight junction (TJ) proteins. The anti-inflammatory effects of cannabinoids in GIT disorders are postulated to occur by the lowering of inflammatory factors such as myeloperoxidase (MPO) activity and regulation of cytokine levels. In conclusion, there is a prospect of utilizing cannabinoids as components of therapy for GIT disorders.


Subject(s)
Cannabinoids , Gastrointestinal Diseases , Gastrointestinal Motility , Permeability , Humans , Cannabinoids/pharmacology , Cannabinoids/therapeutic use , Gastrointestinal Motility/drug effects , Animals , Gastrointestinal Diseases/drug therapy , Gastrointestinal Diseases/metabolism , Permeability/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Endocannabinoids/metabolism
10.
Int Immunopharmacol ; 137: 112416, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-38852521

ABSTRACT

Accumulating evidence emphasizes the critical reciprocity between gut microbiota and intestinal barrier function in maintaining the gastrointestinal homeostasis. Given the fundamental role caused by intestinal permeability, which has been scrutinized as a measurable potential indicator of perturbed barrier function in clinical researches, it seems not surprising that recent decades have been marked by augmented efforts to determine the interaction between intestinal microbes and permeability of the individual. However, despite the significant progress in characterizing intestinal permeability and the commensal bacteria in the intestine, the mechanisms involved are still far from being thoroughly revealed. In the present review, based on multiomic methods, high-throughput sequencing and molecular biology techniques, the impacts of gut microbiota on intestinal permeability as well as their complex interaction networks are systematically summarized. Furthermore, the diseases related to intestinal permeability and main causes of changes in intestinal permeability are briefly introduced. The purpose of this review is to provide a novel prospection to elucidate the correlation between intestinal microbiota and permeability, and to explore a promising solution for diagnosis and treatment of gastrointestinal related diseases.


Subject(s)
Gastrointestinal Diseases , Gastrointestinal Microbiome , Intestinal Mucosa , Permeability , Humans , Animals , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Gastrointestinal Diseases/microbiology , Gastrointestinal Diseases/therapy , Gastrointestinal Diseases/metabolism , Intestinal Barrier Function
11.
Mol Biol Rep ; 51(1): 741, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38874869

ABSTRACT

Gastrointestinal cancer is the most fatal cancer worldwide. The etiology of gastrointestinal cancer has yet to be fully characterized. Alcohol consumption, obesity, tobacco, Helicobacter pylori and gastrointestinal disorders, including gastroesophageal reflux disease, gastric ulcer, colon polyps and non-alcoholic fatty liver disease are among the several risks factors for gastrointestinal cancers. Phycocyanin which is abundant in Spirulina. Phycocyanin, a member of phycobiliprotein family with intense blue color, is an anti-diabetic, neuroprotective, anti-oxidative, anti-inflammatory, and anticancer compound. Evidence exists supporting that phycocyanin has antitumor effects, exerting its pharmacological effects by targeting a variety of cellular and molecular processes, i.e., apoptosis, cell-cycle arrest, migration and Wnt/ß-catenin signaling. Phycocyanin has also been applied in treatment of several gastrointestinal disorders such as, gastric ulcer, ulcerative colitis and fatty liver that is known as a risk factor for progression to cancer. Herein, we summarize various cellular and molecular pathways that are affected by phycocyanin, its efficacy upon combined drug treatment, and the potential for nanotechnology in its gastrointestinal cancer therapy.


Subject(s)
Gastrointestinal Neoplasms , Phycocyanin , Humans , Phycocyanin/pharmacology , Phycocyanin/therapeutic use , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Animals , Apoptosis/drug effects , Gastrointestinal Diseases/drug therapy , Gastrointestinal Diseases/metabolism
12.
Saudi J Gastroenterol ; 30(5): 283-293, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38708898

ABSTRACT

ABSTRACT: Bile acids serve as endogenous ligands for nuclear and cell membrane receptors and play a crucial role in bile acid and lipid metabolism. These detergent-like compounds promote bile flow and aid in the absorption of dietary fats and fat-soluble vitamins in the intestine. Synthesized in the liver as end products of cholesterol catabolism, bile acids exhibit a chemical structure comprising a nucleus and a side chain featuring a carboxyl group, with diverse steric arrangements and potential polar substituents. Critical interactions occur between bile acid species and various nuclear and cell membrane receptors, including the farnesoid X receptor and G-protein-coupled bile acid receptor 1. This research aimed to review the literature on bile acids and their roles in treating different diseases. Currently, numerous investigations are concentrating on specific bile acid species that target nuclear receptors in the gastrointestinal system, aiming to improve the treatment of conditions such as nonalcoholic fatty liver disease. Given the global attention this topic has garnered from research groups, it is considered relatively new, thus anticipating some gaps or incomplete data. Bile acid species have a significant therapeutic promise, especially in their ability to activate or inhibit nuclear receptors, such as farnesoid X receptor. This research provides to offer essential information for scientists and medical practitioners interested in discovering new studies that underscore the importance of bile acids in ameliorating and impeding the progression of disorders. Furthermore, it opens avenues for previously overlooked bile acid-based therapies.


Subject(s)
Bile Acids and Salts , Gastrointestinal Diseases , Receptors, Cytoplasmic and Nuclear , Humans , Bile Acids and Salts/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Gastrointestinal Diseases/drug therapy , Gastrointestinal Diseases/metabolism , Gastrointestinal Diseases/therapy , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Receptors, G-Protein-Coupled/metabolism , Lipid Metabolism/drug effects , Animals
13.
Stem Cell Rev Rep ; 20(6): 1441-1458, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38758462

ABSTRACT

Organoid models have recently been utilized to study 3D human-derived tissue systems to uncover tissue architecture and adult stem cell biology. Patient-derived organoids unambiguously provide the most suitable in vitro system to study disease biology with the actual genetic background. With the advent of much improved and innovative approaches, patient-derived organoids can potentially be used in regenerative medicine. Various human tissues were explored to develop organoids due to their multifold advantage over the conventional in vitro cell line culture approach and in vivo models. Gastrointestinal (GI) tissues have been widely studied to establish organoids and organ-on-chip for screening drugs, nutraceuticals, and other small molecules having therapeutic potential. The function of channel proteins, transporters, and transmembrane proteins was also explained. The successful application of genome editing in organoids using the CRISPR-Cas approach has been reported recently. GI diseases such as Celiac disease (CeD), Inflammatory bowel disease (IBD), and common GI cancers have been investigated using several patient-derived organoid models. Recent advancements on organoid bio-banking and 3D bio-printing contributed significantly in personalized disease management and therapeutics. This article reviews the available literature on investigations and translational applications of patient-derived GI organoid models, notably on elucidating gut-microbial interaction and epigenetic modifications.


Subject(s)
Gastrointestinal Diseases , Organoids , Humans , Organoids/metabolism , Gastrointestinal Diseases/pathology , Gastrointestinal Diseases/therapy , Gastrointestinal Diseases/metabolism , Translational Research, Biomedical , Animals , Biomedical Research , Gene Editing/methods , Models, Biological
14.
Brain Behav Immun ; 119: 867-877, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38750700

ABSTRACT

The gastrointestinal tract is one of the main organs affected during systemic inflammation and disrupted gastrointestinal motility is a major clinical manifestation. Many studies have investigated the involvement of neuroimmune interactions in regulating colonic motility during localized colonic inflammation, i.e., colitis. However, little is known about how the enteric nervous system and intestinal macrophages contribute to dysregulated motility during systemic inflammation. Given that systemic inflammation commonly results from the innate immune response against bacterial infection, we mimicked bacterial infection by administering lipopolysaccharide (LPS) to rats and assessed colonic motility using ex vivo video imaging techniques. We utilized the Cx3cr1-Dtr rat model of transient depletion of macrophages to investigate the role of intestinal macrophages in regulating colonic motility during LPS infection. To investigate the role of inhibitory enteric neurotransmission on colonic motility following LPS, we applied the nitric oxide synthase inhibitor, Nω-nitro-L-arginine (NOLA). Our results confirmed an increase in colonic contraction frequency during LPS-induced systemic inflammation. However, neither the depletion of intestinal macrophages, nor the suppression of inhibitory enteric nervous system activity impacted colonic motility disruption during inflammation. This implies that the interplay between the enteric nervous system and intestinal macrophages is nuanced, and complex, and further investigation is needed to clarify their joint roles in colonic motility.


Subject(s)
Enteric Nervous System , Gastrointestinal Motility , Inflammation , Lipopolysaccharides , Macrophages , Animals , Lipopolysaccharides/pharmacology , Rats , Gastrointestinal Motility/physiology , Macrophages/metabolism , Inflammation/metabolism , Inflammation/physiopathology , Enteric Nervous System/physiopathology , Enteric Nervous System/metabolism , Male , Brain-Gut Axis/physiology , Colon/metabolism , Gastrointestinal Tract/metabolism , Colitis/physiopathology , Colitis/metabolism , Colitis/chemically induced , Brain/metabolism , Rats, Sprague-Dawley , Gastrointestinal Diseases/physiopathology , Gastrointestinal Diseases/metabolism
15.
Int J Mol Sci ; 25(8)2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38673946

ABSTRACT

Inflammatory diseases commonly associated with humans are chronic inflammatory gastrointestinal diseases (CIGDs) [...].


Subject(s)
Inflammation , Humans , Inflammation/metabolism , Gastrointestinal Diseases/metabolism , Gastrointestinal Diseases/therapy , Gastrointestinal Diseases/drug therapy , Animals
16.
Gastroenterology ; 167(2): 231-249, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38431204

ABSTRACT

Ferroptosis is a form of nonapoptotic cell death that involves iron-dependent phospholipid peroxidation induced by accumulation of reactive oxygen species, and results in plasma membrane damage and the release of damage-associated molecular patterns. Ferroptosis has been implicated in aging and immunity, as well as disease states including intestinal and liver conditions and cancer. To date, several ferroptosis-associated genes and pathways have been implicated in liver disease. Although ferroptotic cell death is associated with dysfunction of the intestinal epithelium, the underlying molecular basis is poorly understood. As the mechanisms regulating ferroptosis become further elucidated, there is clear potential to use ferroptosis to achieve therapeutic benefit.


Subject(s)
Ferroptosis , Gastrointestinal Diseases , Reactive Oxygen Species , Humans , Gastrointestinal Diseases/metabolism , Gastrointestinal Diseases/pathology , Gastrointestinal Diseases/physiopathology , Reactive Oxygen Species/metabolism , Animals , Iron/metabolism , Signal Transduction , Lipid Peroxidation
17.
Naunyn Schmiedebergs Arch Pharmacol ; 397(8): 5335-5362, 2024 08.
Article in English | MEDLINE | ID: mdl-38358468

ABSTRACT

Most animals have large amounts of the special substance melatonin, which is controlled by the light/dark cycle in the suprachiasmatic nucleus. According to what is now understood, the gastrointestinal tract (GIT) and other areas of the body are sites of melatonin production. According to recent studies, the GIT and adjacent organs depend critically on a massive amount of melatonin. Not unexpectedly, melatonin's many biological properties, such as its antioxidant, anti-inflammatory, pro-apoptotic, anti-proliferative, anti-metastasis, and antiangiogenic properties, have drawn the attention of researchers more and more. Because melatonin is an antioxidant, it produces a lot of secretions in the GIT's mucus and saliva, which shields cells from damage and promotes the development of certain GIT-related disorders. Melatonin's ability to alter cellular behavior in the GIT and other associated organs, such as the liver and pancreas, is another way that it functions. This behavior alters the secretory and metabolic activities of these cells. In this review, we attempted to shed fresh light on the many roles that melatonin plays in the various regions of the gastrointestinal tract by focusing on its activities for the first time.


Subject(s)
Appetite , Gastrointestinal Diseases , Melatonin , Melatonin/metabolism , Melatonin/therapeutic use , Melatonin/pharmacology , Humans , Animals , Gastrointestinal Diseases/drug therapy , Gastrointestinal Diseases/metabolism , Gastrointestinal Diseases/physiopathology , Appetite/drug effects , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/drug effects , Antioxidants/therapeutic use , Antioxidants/pharmacology
18.
Trends Endocrinol Metab ; 35(5): 364-366, 2024 May.
Article in English | MEDLINE | ID: mdl-38418280

ABSTRACT

Mitochondrial dysfunctions predominantly cause encephalomyopathies with muscle atrophy and neurodegeneration. However, their impact on other tissues, particularly the gastrointestinal tract, requires further investigation. In a recent report in Nature, Moschandrea et al. used mice deficient in the mitochondrial aminoacyl-tRNA synthetase DARS2 to investigate the role of enterocytic mitochondria in dietary lipid processing and transport. Their work sheds light on the development of gastrointestinal disorders as a result of mitochondrial dysfunction.


Subject(s)
Gastrointestinal Diseases , Mitochondria , Animals , Humans , Mice , Gastrointestinal Diseases/metabolism , Lipid Metabolism/physiology , Mitochondria/metabolism
19.
Cell Mol Life Sci ; 81(1): 34, 2024 Jan 12.
Article in English | MEDLINE | ID: mdl-38214802

ABSTRACT

This review provides an update on recent findings from basic, translational, and clinical studies on the molecular mechanisms of mitochondrial dysfunction and apoptosis of hepatocytes in multiple liver diseases, including but not limited to alcohol-associated liver disease (ALD), metabolic dysfunction-associated steatotic liver disease (MASLD), and drug-induced liver injury (DILI). While the ethanol-inducible cytochrome P450-2E1 (CYP2E1) is mainly responsible for oxidizing binge alcohol via the microsomal ethanol oxidizing system, it is also responsible for metabolizing many xenobiotics, including pollutants, chemicals, drugs, and specific diets abundant in n-6 fatty acids, into toxic metabolites in many organs, including the liver, causing pathological insults through organelles such as mitochondria and endoplasmic reticula. Oxidative imbalances (oxidative stress) in mitochondria promote the covalent modifications of lipids, proteins, and nucleic acids through enzymatic and non-enzymatic mechanisms. Excessive changes stimulate various post-translational modifications (PTMs) of mitochondrial proteins, transcription factors, and histones. Increased PTMs of mitochondrial proteins inactivate many enzymes involved in the reduction of oxidative species, fatty acid metabolism, and mitophagy pathways, leading to mitochondrial dysfunction, energy depletion, and apoptosis. Unique from other organelles, mitochondria control many signaling cascades involved in bioenergetics (fat metabolism), inflammation, and apoptosis/necrosis of hepatocytes. When mitochondrial homeostasis is shifted, these pathways become altered or shut down, likely contributing to the death of hepatocytes with activation of inflammation and hepatic stellate cells, causing liver fibrosis and cirrhosis. This review will encapsulate how mitochondrial dysfunction contributes to hepatocyte apoptosis in several types of liver diseases in order to provide recommendations for targeted therapeutics.


Subject(s)
Gastrointestinal Diseases , Liver Diseases, Alcoholic , Mitochondrial Diseases , Humans , Liver/metabolism , Ethanol/pharmacology , Apoptosis , Oxidative Stress , Inflammation/pathology , Gastrointestinal Diseases/metabolism , Hepatocytes/metabolism , Protein Processing, Post-Translational , Mitochondrial Proteins/metabolism , Mitochondrial Diseases/metabolism
20.
J Biol Chem ; 300(1): 105505, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38029963

ABSTRACT

Mutations in receptor guanylyl cyclase C (GC-C) cause severe gastrointestinal disease, including meconium ileus, early onset acute diarrhea, and pediatric inflammatory bowel disease that continues into adulthood. Agonists of GC-C are US Food and Drug Administration-approved drugs for the treatment of constipation and irritable bowel syndrome. Therapeutic strategies targeting GC-C are tested in preclinical mouse models, assuming that murine GC-C mimics human GC-C in its biochemical properties and downstream signaling events. Here, we reveal important differences in ligand-binding affinity and GC activity between mouse GC-C and human GC-C. We generated a series of chimeric constructs of various domains of human and mouse GC-C to show that the extracellular domain of mouse GC-C contributed to log-orders lower affinity of mouse GC-C for ligands than human GC-C. Further, the Vmax of the murine GC domain was lower than that of human GC-C, and allosteric regulation of the receptor by ATP binding to the intracellular kinase-homology domain also differed. These altered properties are reflected in the high concentrations of ligands required to elicit signaling responses in the mouse gut in preclinical models and the specificity of a GC inhibitor towards human GC-C. Therefore, our studies identify considerations in using the murine model to test molecules for therapeutic purposes that work as either agonists or antagonists of GC-C, and vaccines for the bacterial heat-stable enterotoxin that causes watery diarrhea in humans.


Subject(s)
Receptors, Guanylate Cyclase-Coupled , Animals , Child , Humans , Mice , Diarrhea , Enterotoxins , Guanylate Cyclase/metabolism , Ligands , Receptors, Enterotoxin/genetics , Receptors, Guanylate Cyclase-Coupled/antagonists & inhibitors , Receptors, Guanylate Cyclase-Coupled/genetics , Receptors, Guanylate Cyclase-Coupled/metabolism , Gastrointestinal Diseases/drug therapy , Gastrointestinal Diseases/metabolism , Gastrointestinal Diseases/pathology
SELECTION OF CITATIONS
SEARCH DETAIL