1
|
Gradl K, Richter P, Somoza V. Bitter peptides formed during in-vitro gastric digestion induce mechanisms of gastric acid secretion and release satiating serotonin via bitter taste receptors TAS2R4 and TAS2R43 in human parietal cells in culture. Food Chem 2025; 482:144174. [PMID: 40184744 DOI: 10.1016/j.foodchem.2025.144174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 03/02/2025] [Accepted: 03/30/2025] [Indexed: 04/07/2025]
Abstract
A key barrier in transitioning to plant-based, more satiating diets, is the bitter taste of plant proteins. We hypothesize that both, a more bitter tasting (MBT) and a less bitter tasting (LBT) pea protein hydrolysate (PPH) can be digested in the stomach into bitter tasting peptides that stimulate proton secretion (PS) and serotonin release, as two of the key gastric satiety signals, via the functional involvement of bitter taste receptors (TAS2Rs). Using a sensory-guided LC-MS approach, we identified six bitter peptides that were released from LBT-PPH and MBT-PPH during gastric digestion in vitro. TAS2R4 and TAS2R43 involvement in PS and serotonin release was confirmed via CRISPR-Cas9 knockout experiments. Our hypothesis was proven with all six peptides equally stimulating PS in immortalized human gastric HGT-1 cells, and LBT-PPH-derived peptides eliciting a higher serotonin release in HGT-1 cells than MBT-PPH peptides, indicating a satiating potential of less bitter tasting protein hydrolysates.
Collapse
Affiliation(s)
- Katrin Gradl
- TUM School of Life Sciences, Technical University of Munich, Alte Akademie 8, 85354 Freising, Germany; Leibniz Institute for Food Systems Biology at the Technical University of Munich, Lise-Meitner-Straße 34, 85354 Freising, Germany
| | - Phil Richter
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Lise-Meitner-Straße 34, 85354 Freising, Germany
| | - Veronika Somoza
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Lise-Meitner-Straße 34, 85354 Freising, Germany; Chair of Nutritional Systems Biology, Technical University of Munich, Lise-Meitner-Straße 34, 85354 Freising, Germany; Department of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria.
| |
Collapse
|
2
|
Yue Y, Wan X, Liu G, Zhu T, Xu D, Zhao M, Cai Y, Murayama R, Hashimoto H, Anzai N, Hashimoto K. Subdiaphragmatic vagotomy reduces hypothalamic oxytocin expression and blood levels after oral MDMA administration in male rats. Prog Neuropsychopharmacol Biol Psychiatry 2025; 137:111260. [PMID: 39855536 DOI: 10.1016/j.pnpbp.2025.111260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 01/13/2025] [Accepted: 01/17/2025] [Indexed: 01/27/2025]
Abstract
3,4-Methylenedioxymethamphetamine (MDMA) is a widely recognized entactogen frequently used recreationally. It is known for its interaction with the serotonin and oxytocin systems, which underlie its entactogenic effects in humans. Recently, we demonstrated that the gut-brain axis, mediated by the subdiaphragmatic vagus nerve, contributes to MDMA-induced resilience enhancement in rodents. This study investigates whether subdiaphragmatic vagotomy (SDV) affects plasma oxytocin levels and the expression of oxytocin and c-Fos in the hypothalamus following a single oral dose of MDMA in rats. SDV significantly reduced baseline plasma oxytocin levels and oxytocin expression in the paraventricular and supraoptic nuclei of the hypothalamus. Furthermore, SDV markedly attenuated MDMA-induced increases in plasma oxytocin and the expression of oxytocin and c-Fos in these hypothalamic regions. These findings suggest that the subdiaphragmatic vagus nerve plays a critical role in brain-body communication, mediating MDMA's pharmacological effects on the oxytocin system.
Collapse
Affiliation(s)
- Yong Yue
- Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan; Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Xiayun Wan
- Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan
| | - Guilin Liu
- Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan; Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan; Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao 266100, China
| | - Tingting Zhu
- Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan; Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Dan Xu
- Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan; Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Mingming Zhao
- Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan; Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yi Cai
- Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan; Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Rumi Murayama
- Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan; Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan
| | - Hirofumi Hashimoto
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Naohiko Anzai
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Kenji Hashimoto
- Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan.
| |
Collapse
|
3
|
Ahangari G, Norioun H. The dual role of dopamine and serotonin in cancer progression and inflammation: Mechanisms and therapeutic potential. Neuroscience 2025; 569:184-194. [PMID: 39675693 DOI: 10.1016/j.neuroscience.2024.12.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/20/2024] [Accepted: 12/07/2024] [Indexed: 12/17/2024]
Abstract
This review examines the critical functions of dopamine and serotonin in the regulation of inflammation and cancer, emphasizing their potential as therapeutic targets. Traditionally recognized for their roles in neural communication, these neurotransmitters are now understood to play substantial roles in immune modulation and tumor progression. We conducted a systematic review of studies published between 2013 and 2024, using databases such as PubMed, Google Scholar, and Scopus, to assess dopamine and serotonin synthesis, receptor activity, and involvement in disease pathways. Findings indicate that dopamine, through its D1 and D2 receptors, exerts both pro- and anti-inflammatory effects, influencing tumor growth and immune responses in cancers such as breast and pancreatic. Similarly, serotonin, particularly through receptors HTR2A and HTR2B, has demonstrated dual roles in cancer progression, impacting the growth and metastasis of cancers such as gastric and colorectal. This review also addresses the interaction between dopamine and serotonin signaling pathways, which may collectively alter immune cell function and tumor microenvironment dynamics, suggesting a promising direction for combined therapeutic approaches. By synthesizing current data on dopamine and serotonin pathways, this review aims to inform the development of targeted therapies that modulate immune responses in inflammation-driven cancers. Our findings underscore the potential of neurotransmitter-based interventions as a novel strategy for managing cancer and inflammatory diseases.
Collapse
Affiliation(s)
- Ghasem Ahangari
- Medical Genetics Department, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Iran.
| | - Hamid Norioun
- Medical Genetics Department, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Iran.
| |
Collapse
|
4
|
Navarro-Corcuera A, Martínez-Chantar ML. Histone serotonylation in HCC: Decoding the impact of "happy" histones on liver cancer progression. J Hepatol 2025:S0168-8278(25)00129-1. [PMID: 40023196 DOI: 10.1016/j.jhep.2025.02.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 03/04/2025]
Affiliation(s)
- Amaia Navarro-Corcuera
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - María L Martínez-Chantar
- Liver Disease Lab, CIC bioGUNE, Basque Research and Technology Alliance, BRTA, Derio 48160 Bizkaia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Carlos III National Health Institute, 28029 Madrid, Spain.
| |
Collapse
|
5
|
Kilic M, Karakoy Z, Halici H, Cadirci E, Halici Z. Novel therapeutic approaches targeting 5-HT7 receptors outside the central nervous system. J Recept Signal Transduct Res 2025; 45:55-60. [PMID: 39754608 DOI: 10.1080/10799893.2024.2446401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 01/06/2025]
Abstract
Serotonin (5-HT) is a neurotransmitter found throughout the human body that regulates many physiological events arising from the brain and central nervous system (CNS), such as sleep and appetite. However, it has many other functions in systems outside. In addition to the routine expression of 5-HT7 receptors in CNS regions, such as the pituitary gland, spinal cord, and hippocampus, many studies have reported the expression of these receptors in pathological conditions outside. The role of 5-HT7 receptors outside the CNS has been attracting increased attention in recent years. This review highlights the fact that 5-HT7 receptors are associated with diseases and systems beyond the CNS increasing or decreasing in response to cellular changes. Clinical, basic, in vivo and in vitro studies to date are described, but more research is needed to better understand the role of 5-HT7 receptors outside the CNS.
Collapse
Affiliation(s)
- Melis Kilic
- Department of Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Zeynep Karakoy
- Department of Pharmacology, Faculty of Pharmacy, Erzincan Binali Yıldırım University, Erzincan, Turkey
| | - Hamza Halici
- Department of Hınıs Vocational Training School, Ataturk University, Erzurum, Turkey
| | - Elif Cadirci
- Department of Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Zekai Halici
- Department of Pharmacology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| |
Collapse
|
6
|
Zhu P, Savova MV, Kindt A, Wopereis H, Belzer C, Harms AC, Hankemeier T. Exploring the Fecal Metabolome in Infants With Cow's Milk Allergy: The Distinct Impacts of Cow's Milk Protein Tolerance Acquisition and of Synbiotic Supplementation. Mol Nutr Food Res 2025; 69:e202400583. [PMID: 39665335 PMCID: PMC11704826 DOI: 10.1002/mnfr.202400583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/11/2024] [Accepted: 11/25/2024] [Indexed: 12/13/2024]
Abstract
SCOPE Cow's milk allergy (CMA) is one of the most prevalent food allergies in early childhood, often treated via elimination diets including standard amino acid-based formula or amino acid-based formula supplemented with synbiotics (AAF or AAF-S). This work aimed to assess the effect of cow's milk (CM) tolerance acquisition and synbiotic (inulin, oligofructose, Bifidobacterium breve M-16 V) supplementation on the fecal metabolome in infants with IgE-mediated CMA. METHODS AND RESULTS The CMA-allergic infants received AAF or AAF-S for a year during which fecal samples were collected. The samples were subjected to metabolomics analyses covering gut microbial metabolites including SCFAs, tryptophan metabolites, and bile acids (BAs). Longitudinal data analysis suggested amino acids, BAs, and branched SCFAs alterations in infants who outgrew CMA during the intervention. Synbiotic supplementation significantly modified the fecal metabolome after 6 months of intervention, including altered purine, BA, and unsaturated fatty acid levels, and increased metabolites of infant-type Bifidobacterium species: indolelactic acid and 4-hydroxyphenyllactic acid. CONCLUSION This study offers no clear conclusion on the impact of CM-tolerance acquisition on the fecal metabolome. However, our results show that 6 months of synbiotic supplementation successfully altered fecal metabolome and suggest induced bifidobacteria activity, which subsequently declined after 12 months of intervention.
Collapse
Affiliation(s)
- Pingping Zhu
- Metabolomics and Analytics CentreLeiden Academic Centre for Drug ResearchLeiden UniversityLeidenThe Netherlands
| | - Mariyana V. Savova
- Metabolomics and Analytics CentreLeiden Academic Centre for Drug ResearchLeiden UniversityLeidenThe Netherlands
| | - Alida Kindt
- Metabolomics and Analytics CentreLeiden Academic Centre for Drug ResearchLeiden UniversityLeidenThe Netherlands
| | | | | | - Clara Belzer
- Laboratory of MicrobiologyWageningen UniversityWageningenThe Netherlands
| | - Amy C. Harms
- Metabolomics and Analytics CentreLeiden Academic Centre for Drug ResearchLeiden UniversityLeidenThe Netherlands
| | - Thomas Hankemeier
- Metabolomics and Analytics CentreLeiden Academic Centre for Drug ResearchLeiden UniversityLeidenThe Netherlands
| |
Collapse
|
7
|
Lemmetyinen TT, Viitala EW, Wartiovaara L, Päivinen P, Virtanen HT, Pentinmikko N, Katajisto P, Mäkelä TP, Wang TC, Andressoo JO, Ollila S. Mesenchymal GDNF promotes intestinal enterochromaffin cell differentiation. iScience 2024; 27:111246. [PMID: 39634560 PMCID: PMC11616604 DOI: 10.1016/j.isci.2024.111246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 09/02/2024] [Accepted: 10/22/2024] [Indexed: 12/07/2024] Open
Abstract
Enteroendocrine cells (EECs) differentiate and mature to form functionally distinct populations upon migration along the intestinal crypt-villus axis, but how niche signals affect this process is poorly understood. Here, we identify expression of Glial cell line-derived neurotrophic factor (GDNF) in the intestinal subepithelial myofibroblasts (SEMFs), while the GDNF receptor RET was expressed in a subset of EECs, suggesting GDNF-mediated regulation. Indeed, GDNF-RET signaling induced increased expression of EEC genes including Tph1, encoding for the rate-limiting enzyme for 5-hydroxytryptamine (5-HT, serotonin) biosynthesis, and increased the frequency of 5-HT+ enterochromaffin cells (ECs) in mouse organoid culture experiments and in vivo. Moreover, expression of the 5-HT receptor Htr4 was enriched in Lgr5+ intestinal stem cells (ISCs) and 5-HT reduced the ISC clonogenicity. In summary, our results show that GDNF-RET signaling regulate EEC differentiation, and suggest 5-HT as a potential niche factor regulating Lgr5+ ISC activity, with potential implications in intestinal regeneration.
Collapse
Affiliation(s)
- Toni T. Lemmetyinen
- Translational Cancer Medicine Program, University of Helsinki, 00014 Helsinki, Finland
| | - Emma W. Viitala
- Translational Cancer Medicine Program, University of Helsinki, 00014 Helsinki, Finland
| | - Linnea Wartiovaara
- Translational Cancer Medicine Program, University of Helsinki, 00014 Helsinki, Finland
| | - Pekka Päivinen
- HiLIFE-Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, 00014 Helsinki, Finland
| | - Heikki T. Virtanen
- Department of Pharmacology, Faculty of Medicine, Helsinki Institute of Life Science, University of Helsinki, 00290 Helsinki, Finland
| | - Nalle Pentinmikko
- The Francis Crick Institute, London NW1 1AY, UK
- Institute of Biotechnology, HiLIFE, University of Helsinki, 00014 Helsinki, Finland
| | - Pekka Katajisto
- Institute of Biotechnology, HiLIFE, University of Helsinki, 00014 Helsinki, Finland
- Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences Research Program, University of Helsinki, 00014 Helsinki, Finland
- Department of Cell and Molecular Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Tomi P. Mäkelä
- HiLIFE-Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, 00014 Helsinki, Finland
| | - Timothy C. Wang
- Division of Digestive and Liver Diseases, Department of Medicine, Irving Cancer Research Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Jaan-Olle Andressoo
- Department of Pharmacology, Faculty of Medicine, Helsinki Institute of Life Science, University of Helsinki, 00290 Helsinki, Finland
- Division of Neurogeriatrics, Department of Neurobiology, Care Science and Society (NVS), Karolinska Institutet, 17177 Stockholm, Sweden
| | - Saara Ollila
- Translational Cancer Medicine Program, University of Helsinki, 00014 Helsinki, Finland
- Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences Research Program, University of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|
8
|
Jiang B, Quinn-Bohmann N, Diener C, Nathan VB, Han-Hallett Y, Reddivari L, Gibbons SM, Baloni P. Understanding disease-associated metabolic changes in human colon epithelial cells using i ColonEpithelium metabolic reconstruction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619644. [PMID: 39484551 PMCID: PMC11526933 DOI: 10.1101/2024.10.22.619644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
The colon epithelium plays a key role in the host-microbiome interactions, allowing uptake of various nutrients and driving important metabolic processes. To unravel detailed metabolic activities in the human colon epithelium, our present study focuses on the generation of the first cell-type specific genome-scale metabolic model (GEM) of human colonic epithelial cells, named iColonEpithelium. GEMs are powerful tools for exploring reactions and metabolites at systems level and predicting the flux distributions at steady state. Our cell-type-specific iColonEpithelium metabolic reconstruction captures genes specifically expressed in the human colonic epithelial cells. The iColonEpithelium is also capable of performing metabolic tasks specific to the cell type. A unique transport reaction compartment has been included to allow simulation of metabolic interactions with the gut microbiome. We used iColonEpithelium to identify metabolic signatures associated with inflammatory bowel disease. We integrated single-cell RNA sequencing data from Crohn's Diseases (CD) and ulcerative colitis (UC) samples with the iColonEpithelium metabolic network to predict metabolic signatures of colonocytes between CD and UC compared to healthy samples. We identified reactions in nucleotide interconversion, fatty acid synthesis and tryptophan metabolism were differentially regulated in CD and UC conditions, which were in accordance with experimental results. The iColonEpithelium metabolic network can be used to identify mechanisms at the cellular level, and our network has the potential to be integrated with gut microbiome models to explore the metabolic interactions between host and gut microbiota under various conditions.
Collapse
|
9
|
Liu HN, Nakamura M, Kawashima H. New Role of the Serotonin as a Biomarker of Gut-Brain Interaction. Life (Basel) 2024; 14:1280. [PMID: 39459580 PMCID: PMC11509611 DOI: 10.3390/life14101280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 09/23/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
Serotonin (5-hydroxytryptamine: 5-HT), a neurotransmitter that regulates mood in the brain and signaling in the gut, has receptors throughout the body that serve various functions, especially in the gut and brain. Selective serotonin reuptake inhibitors (SSRIs) are used to treat depression, but their efficacy is uncertain. Depression is often associated with early gastrointestinal symptoms. Gut disorders such as functional dyspepsia (FD), irritable bowel syndrome (IBS) and inflammatory bowel disease (IBD), including ulcerative colitis (UC) and Crohn's disease (CD), are linked to elevated serotonin levels. In this review, we would like to discuss the approach of using serotonin as a biomarker for gut-brain, and body-wide organ communication may lead to the development of preventive and innovative treatments for gut-brain disorders, offering improved visibility and therapeutic monitoring. It could also be used to gauge stress intensity for self-care and mental health improvement.
Collapse
Affiliation(s)
- Hong Nian Liu
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan; (M.N.)
| | | | | |
Collapse
|
10
|
Janova H, Zhao FR, Desai P, Mack M, Thackray LB, Stappenbeck TS, Diamond MS. West Nile virus triggers intestinal dysmotility via T cell-mediated enteric nervous system injury. J Clin Invest 2024; 134:e181421. [PMID: 39207863 PMCID: PMC11527448 DOI: 10.1172/jci181421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
Intestinal dysmotility syndromes have been epidemiologically associated with several antecedent bacterial and viral infections. To model this phenotype, we previously infected mice with the neurotropic flavivirus West Nile virus (WNV) and demonstrated intestinal transit defects. Here, we found that within 1 week of WNV infection, enteric neurons and glia became damaged, resulting in sustained reductions of neuronal cells and their networks of connecting fibers. Using cell-depleting antibodies, adoptive transfer experiments, and mice lacking specific immune cells or immune functions, we show that infiltrating WNV-specific CD4+ and CD8+ T cells damaged the enteric nervous system (ENS) and glia, which led to intestinal dysmotility; these T cells used multiple and redundant effector molecules including perforin and Fas ligand. In comparison, WNV-triggered ENS injury and intestinal dysmotility appeared to not require infiltrating monocytes, and damage may have been limited by resident muscularis macrophages. Overall, our experiments support a model in which antigen-specific T cell subsets and their effector molecules responding to WNV infection direct immune pathology against enteric neurons and supporting glia that results in intestinal dysmotility.
Collapse
Affiliation(s)
- Hana Janova
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Fang R. Zhao
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Pritesh Desai
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Matthias Mack
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Larissa B. Thackray
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| | | | - Michael S. Diamond
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
- Department of Pathology and Immunology
- Department of Molecular Microbiology, and
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
11
|
Momen YS, Mishra J, Kumar N. Brain-Gut and Microbiota-Gut-Brain Communication in Type-2 Diabetes Linked Alzheimer's Disease. Nutrients 2024; 16:2558. [PMID: 39125436 PMCID: PMC11313915 DOI: 10.3390/nu16152558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 08/12/2024] Open
Abstract
The gastrointestinal (GI) tract, home to the largest microbial population in the human body, plays a crucial role in overall health through various mechanisms. Recent advancements in research have revealed the potential implications of gut-brain and vice-versa communication mediated by gut-microbiota and their microbial products in various diseases including type-2 diabetes and Alzheimer's disease (AD). AD is the most common type of dementia where most of cases are sporadic with no clearly identified cause. However, multiple factors are implicated in the progression of sporadic AD which can be classified as non-modifiable (e.g., genetic) and modifiable (e.g. Type-2 diabetes, diet etc.). Present review focusses on key players particularly the modifiable factors such as Type-2 diabetes (T2D) and diet and their implications in microbiota-gut-brain (MGB) and brain-gut (BG) communication and cognitive functions of healthy brain and their dysfunction in Alzheimer's Disease. Special emphasis has been given on elucidation of the mechanistic aspects of the impact of diet on gut-microbiota and the implications of some of the gut-microbial products in T2D and AD pathology. For example, mechanistically, HFD induces gut dysbiosis with driven metabolites that in turn cause loss of integrity of intestinal barrier with concomitant colonic and systemic chronic low-grade inflammation, associated with obesity and T2D. HFD-induced obesity and T2D parallel neuroinflammation, deposition of Amyloid β (Aβ), and ultimately cognitive impairment. The review also provides a new perspective of the impact of diet on brain-gut and microbiota-gut-brain communication in terms of transcription factors as a commonly spoken language that may facilitates the interaction between gut and brain of obese diabetic patients who are at a higher risk of developing cognitive impairment and AD. Other commonality such as tyrosine kinase expression and functions maintaining intestinal integrity on one hand and the phagocytic clarence by migratory microglial functions in brain are also discussed. Lastly, the characterization of the key players future research that might shed lights on novel potential pharmacological target to impede AD progression are also discussed.
Collapse
Affiliation(s)
| | | | - Narendra Kumar
- Department of Pharmaceutical Sciences, ILR College of Pharmacy, Texas A&M Health Science Center, Kingsville, TX 78363, USA
| |
Collapse
|
12
|
Zhang S, Li Q, Huang Z, Wang G, Zheng X, Liu J. Exploring community succession and metabolic changes in corn gluten meal-bran mixed wastes during fermentation. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2024; 366:121684. [PMID: 38981273 DOI: 10.1016/j.jenvman.2024.121684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/11/2024]
Abstract
Addressing the challenge of sustainable agricultural processing waste management is crucial. Protein sources are essential for livestock farming, and one viable solution is the microbial fermentation of agricultural by-products. In this study, the microorganisms utilized for fermentation were Pichia fermentans PFZS and Limmosilactobacillus fermentum LFZS. The results demonstrated that the fermented corn gluten meal-bran mixture (FCBM) effectively degraded high molecular weight proteins, resulting in increases of approximately 23.3%, 367.6%, and 159.3% in crude protein (CP), trichloroacetic acid-soluble protein (TCA-SP), and free amino acid (FAA), respectively. Additionally, there was a significant enhancement in the content of beneficial metabolites, including total phenols, carotenoids, and microorganisms. FCBM also effectively reduced anti-nutritional factors while boosting antioxidant and anti-inflammatory substances, such as dipeptides and tripeptides. The fermentation process was marked by an increase in beneficial endophytes, which was closely correlated with the enhancement of beneficial metabolites. Overall, FCBM provides a theoretical basis for substituting traditional protein resources in animal husbandry.
Collapse
Affiliation(s)
- Shuai Zhang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Qining Li
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Zhaoxin Huang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Guoxia Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Xin Zheng
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China.
| | - Jingsheng Liu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin, 130118, China; National Engineering Research Center for Wheat and Corn Deep Processing, Changchun, Jilin, 130118, China.
| |
Collapse
|
13
|
Wang X, Wen X, Yuan S, Zhang J. Gut-brain axis in the pathogenesis of sepsis-associated encephalopathy. Neurobiol Dis 2024; 195:106499. [PMID: 38588753 DOI: 10.1016/j.nbd.2024.106499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 03/31/2024] [Accepted: 04/04/2024] [Indexed: 04/10/2024] Open
Abstract
The gut-brain axis is a bidirectional communication network linking the gut and the brain, overseeing digestive functions, emotional responses, body immunity, brain development, and overall health. Substantial research highlights a connection between disruptions of the gut-brain axis and various psychiatric and neurological conditions, including depression and Alzheimer's disease. Given the impact of the gut-brain axis on behavior, cognition, and brain diseases, some studies have started to pay attention to the role of the axis in sepsis-associated encephalopathy (SAE), where cognitive impairment is the primary manifestation. SAE emerges as the primary and earliest form of organ dysfunction following sepsis, potentially leading to acute cognitive impairment and long-term cognitive decline in patients. Notably, the neuronal damage in SAE does not stem directly from the central nervous system (CNS) infection but rather from an infection occurring outside the brain. The gut-brain axis is posited as a pivotal factor in this process. This review will delve into the gut-brain axis, exploring four crucial pathways through which inflammatory signals are transmitted and elevate the incidence of SAE. These pathways encompass the vagus nerve pathway, the neuroendocrine pathway involving the hypothalamic-pituitary-adrenal (HPA) axis and serotonin (5-HT) regulation, the neuroimmune pathway, and the microbial regulation. These pathways can operate independently or collaboratively on the CNS to modulate brain activity. Understanding how the gut affects and regulates the CNS could offer the potential to identify novel targets for preventing and treating this condition, ultimately enhancing the prognosis for individuals with SAE.
Collapse
Affiliation(s)
- Xin Wang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China
| | - Xiaoyue Wen
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China
| | - Shiying Yuan
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China.
| | - Jiancheng Zhang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China.
| |
Collapse
|
14
|
Atanga R, Appell LL, Thompson MN, Lauer FT, Brearley A, Campen MJ, Castillo EF, In JG. Single Cell Analysis of Human Colonoids Exposed to Uranium-Bearing Dust. ENVIRONMENTAL HEALTH PERSPECTIVES 2024; 132:57006. [PMID: 38771937 PMCID: PMC11108582 DOI: 10.1289/ehp13855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 04/23/2024] [Accepted: 04/30/2024] [Indexed: 05/23/2024]
Abstract
BACKGROUND Uranium exposure remains an important environmental legacy and physiological health concern, with hundreds of abandoned uranium mines located in the Southwestern United States largely impacting underserved indigenous communities. The negative effects of heavy metals on barrier permeability and inhibition of intestinal epithelial healing have been described; however, transcriptomic changes within the intestinal epithelial cells and impacts on lineage differentiation are largely unknown. OBJECTIVES Herein, we sought to determine the molecular and cellular changes that occur in the colon in response to uranium bearing dust (UBD) exposure. METHODS Human colonoids from three biologically distinct donors were acutely exposed to UBD then digested for single cell RNA sequencing to define the molecular changes that occur to specific identities of colonic epithelial cells. Validation in colonoids was assessed using morphological and imaging techniques. RESULTS Human colonoids acutely exposed to UBD exhibited disrupted proliferation and hyperplastic differentiation of the secretory lineage cell, enteroendocrine cells (EEC). Single-cell RNA sequencing also showed more EEC subtypes present in UBD-exposed colonoids. DISCUSSION These findings highlight the significance of crypt-based proliferative cells and secretory cell differentiation using human colonoids to model major colonic responses to uranium-bearing particulate dust exposure. https://doi.org/10.1289/EHP13855.
Collapse
Affiliation(s)
- Roger Atanga
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Lidia L. Appell
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Myranda N. Thompson
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Fredine T. Lauer
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Adrian Brearley
- Department of Earth and Planetary Sciences, College of Arts and Sciences, University of New Mexico, Albuquerque, New Mexico, USA
| | - Matthew J. Campen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Eliseo F. Castillo
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
- Autophagy, Inflammation and Metabolism Center of Biomedical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Julie G. In
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
- Autophagy, Inflammation and Metabolism Center of Biomedical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| |
Collapse
|
15
|
Norrby K. On Connective Tissue Mast Cells as Protectors of Life, Reproduction, and Progeny. Int J Mol Sci 2024; 25:4499. [PMID: 38674083 PMCID: PMC11050338 DOI: 10.3390/ijms25084499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/10/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
The connective tissue mast cell (MC), a sentinel tissue-residing secretory immune cell, has been preserved in all vertebrate classes since approximately 500 million years. No physiological role of the MC has yet been established. Considering the power of natural selection of cells during evolution, it is likely that the MCs exert essential yet unidentified life-promoting actions. All vertebrates feature a circulatory system, and the MCs interact readily with the vasculature. It is notable that embryonic MC progenitors are generated from endothelial cells. The MC hosts many surface receptors, enabling its activation via a vast variety of potentially harmful exogenous and endogenous molecules and via reproductive hormones in the female sex organs. Activated MCs release a unique composition of preformed and newly synthesized bioactive molecules, like heparin, histamine, serotonin, proteolytic enzymes, cytokines, chemokines, and growth factors. MCs play important roles in immune responses, tissue remodeling, cell proliferation, angiogenesis, inflammation, wound healing, tissue homeostasis, health, and reproduction. As recently suggested, MCs enable perpetuation of the vertebrates because of key effects-spanning generations-in ovulation and pregnancy, as in life-preserving activities in inflammation and wound healing from birth till reproductive age, thus creating a permanent life-sustaining loop. Here, we present recent advances that further indicate that the MC is a specific life-supporting and progeny-safeguarding cell.
Collapse
Affiliation(s)
- Klas Norrby
- Department of Pathology, Institute of Medical Biology, Sahlgren Academy, University of Gothenburg, 7 Ostindiefararen, SE-417 65 Gothenburg, Sweden
| |
Collapse
|
16
|
Sha Y, Liu X, He Y, Zhao S, Hu J, Wang J, Li W, Shao P, Wang F, Chen X, Yang W, Xie Z. Multi-omics revealed rumen microbiota metabolism and host immune regulation in Tibetan sheep of different ages. Front Microbiol 2024; 15:1339889. [PMID: 38414776 PMCID: PMC10896911 DOI: 10.3389/fmicb.2024.1339889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 01/17/2024] [Indexed: 02/29/2024] Open
Abstract
The rumen microbiota and metabolites play an important role in energy metabolism and immune regulation of the host. However, the regulatory mechanism of rumen microbiota and metabolite interactions with host on Tibetan sheep's plateau adaptability is still unclear. We analyzed the ruminal microbiome and metabolome, host transcriptome and serum metabolome characteristics of Tibetan sheep at different ages. Biomarkers Butyrivibrio, Lachnospiraceae_XPB1014_group, Prevotella, and Rikenellaceae_RC9_gut_group were found in 4 months, 1.5 years, 3.5 years, and 6 years Tibetan sheep, respectively. The rumen microbial metabolites were mainly enriched in galactose metabolism, unsaturated fatty acid biosynthesis and fatty acid degradation pathways, and had significant correlation with microbiota. These metabolites further interact with mRNA, and are co-enriched in arginine and proline metabolism, metabolism of xenobiotics by cytochrome P450, propanoate metabolism, starch and sucrose metabolism, gap junction pathway. Meanwhile, serum metabolites also have a similar function, such as chemical carcinogenesis - reactive oxygen species, limonene and pinene degradation, and cutin, suberine and wax biosynthesis, thus participating in the regulation of the body's immune and energy-related metabolic processes. This study systematically revealed that rumen microbiota, metabolites, mRNA and serum metabolites of Tibetan sheep were involved in the regulation of fermentation metabolic function and immune level of Tibetan sheep at different ages, which provided a new perspective for plateau adaptability research of Tibetan sheep at different ages.
Collapse
Affiliation(s)
- Yuzhu Sha
- College of Animal Science and Technology/Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou, China
| | - Xiu Liu
- College of Animal Science and Technology/Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou, China
| | - Yanyu He
- School of Fundamental Sciences, Massey University, Palmerston North, New Zealand
| | - Shengguo Zhao
- College of Animal Science and Technology/Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou, China
| | - Jiang Hu
- College of Animal Science and Technology/Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou, China
| | - Jiqing Wang
- College of Animal Science and Technology/Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou, China
| | - Wenhao Li
- Academy of Animal Science and Veterinary Medicine, Qinghai University, Xining, China
| | - Pengyang Shao
- College of Animal Science and Technology/Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou, China
| | - Fanxiong Wang
- College of Animal Science and Technology/Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou, China
| | - Xiaowei Chen
- College of Animal Science and Technology/Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou, China
| | - Wenxin Yang
- College of Animal Science and Technology/Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou, China
| | - Zhuanhui Xie
- College of Animal Science and Technology/Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou, China
| |
Collapse
|
17
|
Sung J, Rajendraprasad SS, Philbrick KL, Bauer BA, Gajic O, Shah A, Laudanski K, Bakken JS, Skalski J, Karnatovskaia LV. The human gut microbiome in critical illness: disruptions, consequences, and therapeutic frontiers. J Crit Care 2024; 79:154436. [PMID: 37769422 PMCID: PMC11034825 DOI: 10.1016/j.jcrc.2023.154436] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/23/2023] [Accepted: 09/18/2023] [Indexed: 09/30/2023]
Abstract
With approximately 39 trillion cells and over 20 million genes, the human gut microbiome plays an integral role in both health and disease. Modern living has brought a widespread use of processed food and beverages, antimicrobial and immunomodulatory drugs, and invasive procedures, all of which profoundly disrupt the delicate homeostasis between the host and its microbiome. Of particular interest is the human gut microbiome, which is progressively being recognized as an important contributing factor in many aspects of critical illness, from predisposition to recovery. Herein, we describe the current understanding of the adverse impacts of standard intensive care interventions on the human gut microbiome and delve into how these microbial alterations can influence patient outcomes. Additionally, we explore the potential association between the gut microbiome and post-intensive care syndrome, shedding light on a previously underappreciated avenue that may enhance patient recuperation following critical illness. There is an impending need for future epidemiological studies to encompass detailed phenotypic analyses of gut microbiome perturbations. Interventions aimed at restoring the gut microbiome represent a promising therapeutic frontier in the quest to prevent and treat critical illnesses.
Collapse
Affiliation(s)
- Jaeyun Sung
- Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | | | - Kemuel L Philbrick
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Brent A Bauer
- Department of General Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Ognjen Gajic
- Department of Pulmonary & Critical Care, Mayo Clinic, Rochester, MN, USA
| | - Aditya Shah
- Division of Public Health, Infectious Diseases and Occupational Medicine, Mayo Clinic, Rochester, MN, USA
| | - Krzysztof Laudanski
- Department of Anesthesiology and Perioperative Care, Mayo Clinic, Rochester, MN, USA
| | - Johan S Bakken
- Department of Infectious Diseases, St Luke's Hospital, Duluth, MN, United States of America
| | - Joseph Skalski
- Department of Pulmonary & Critical Care, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
18
|
Sacoor C, Marugg JD, Lima NR, Empadinhas N, Montezinho L. Gut-Brain Axis Impact on Canine Anxiety Disorders: New Challenges for Behavioral Veterinary Medicine. Vet Med Int 2024; 2024:2856759. [PMID: 38292207 PMCID: PMC10827376 DOI: 10.1155/2024/2856759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 08/28/2023] [Accepted: 10/25/2023] [Indexed: 02/01/2024] Open
Abstract
Anxiety disorders in dogs are ever-growing and represent an important concern in the veterinary behavior field. These disorders are often disregarded in veterinary clinical practice, negatively impacting the animal's and owner's quality of life. Moreover, these anxiety disorders can potentially result in the abandonment or euthanasia of dogs. Growing evidence shows that the gut microbiota is a central player in the gut-brain axis. A variety of microorganisms inhabit the intestines of dogs, which are essential in maintaining intestinal homeostasis. These microbes can impact mental health through several mechanisms, including metabolic, neural, endocrine, and immune-mediated pathways. The disruption of a balanced composition of resident commensal communities, or dysbiosis, is implicated in several pathological conditions, including mental disorders such as anxiety. Studies carried out in rodent models and humans demonstrate that the intestinal microbiota can influence mental health through these mechanisms, including anxiety disorders. Furthermore, novel therapeutic strategies using prebiotics and probiotics have been shown to ameliorate anxiety-related symptoms. However, regarding the canine veterinary behavior field, there is still a lack of insightful research on this topic. In this review, we explore the few but relevant studies performed on canine anxiety disorders. We agree that innovative bacterial therapeutical approaches for canine anxiety disorders will become a promising field of investigation and certainly pave the way for new approaches to these behavioral conditions.
Collapse
Affiliation(s)
- Carina Sacoor
- Vasco da Gama Research Center (CIVG), Vasco da Gama University School (EUVG), 3020–210 Coimbra, Portugal
- Department of Veterinary Sciences, School of Agrarian and Veterinary Sciences (ECAV), University of Trás-os-Montes e Alto Douro (UTAD), 5000–801 Vila Real, Portugal
| | - John D. Marugg
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004–504 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004–504 Coimbra, Portugal
| | - Nuno R. Lima
- Department of Veterinary Sciences, School of Agrarian and Veterinary Sciences (ECAV), University of Trás-os-Montes e Alto Douro (UTAD), 5000–801 Vila Real, Portugal
- Animal and Veterinary Research Centre (CECAV), UTAD, and Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 5000–801 Vila Real, Portugal
- Innovation in Health and Well-Being Research Unit (iHealth4Well-Being), Polytechnic Health Institute of North (IPSN-CESPU), 4585-116 Gandra, Portugal
| | - Nuno Empadinhas
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004–504 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004–504 Coimbra, Portugal
| | - Liliana Montezinho
- Vasco da Gama Research Center (CIVG), Vasco da Gama University School (EUVG), 3020–210 Coimbra, Portugal
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004–504 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004–504 Coimbra, Portugal
| |
Collapse
|
19
|
Ciernikova S, Sevcikova A, Mladosievicova B, Mego M. Microbiome in Cancer Development and Treatment. Microorganisms 2023; 12:24. [PMID: 38257851 PMCID: PMC10819529 DOI: 10.3390/microorganisms12010024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 12/13/2023] [Accepted: 12/20/2023] [Indexed: 01/24/2024] Open
Abstract
Targeting the microbiome, microbiota-derived metabolites, and related pathways represents a significant challenge in oncology. Microbiome analyses have confirmed the negative impact of cancer treatment on gut homeostasis, resulting in acute dysbiosis and severe complications, including massive inflammatory immune response, mucosal barrier disruption, and bacterial translocation across the gut epithelium. Moreover, recent studies revealed the relationship between an imbalance in the gut microbiome and treatment-related toxicity. In this review, we provide current insights into the role of the microbiome in tumor development and the impact of gut and tumor microbiomes on chemo- and immunotherapy efficacy, as well as treatment-induced late effects, including cognitive impairment and cardiotoxicity. As discussed, microbiota modulation via probiotic supplementation and fecal microbiota transplantation represents a new trend in cancer patient care, aiming to increase bacterial diversity, alleviate acute and long-term treatment-induced toxicity, and improve the response to various treatment modalities. However, a more detailed understanding of the complex relationship between the microbiome and host can significantly contribute to integrating a microbiome-based approach into clinical practice. Determination of causal correlations might lead to the identification of clinically relevant diagnostic and prognostic microbial biomarkers. Notably, restoration of intestinal homeostasis could contribute to optimizing treatment efficacy and improving cancer patient outcomes.
Collapse
Affiliation(s)
- Sona Ciernikova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia;
| | - Aneta Sevcikova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of the Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia;
| | - Beata Mladosievicova
- Institute of Pathological Physiology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia;
| | - Michal Mego
- 2nd Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, 833 10 Bratislava, Slovakia;
| |
Collapse
|
20
|
Penkova N, Atanasova P, Penkov R, Hrischev P, Peychev L, Peychev Z. Serotonin production of the developing gastrointestinal tract of human embryos in 6th gestation week. PHARMACIA 2023; 70:1499-1509. [DOI: 10.3897/pharmacia.70.e114080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
Background: Local regulation of gastrointestinal tract digestion is performed by a large number of hormones produced by the mucosal enteroendocrine cells. Some of the earliest differentiating cells in the gastrointestinal tract are enteroendocrine cells. Serotonin-producing cells - EC cells are found mostly in the stomach and duodenum.
Aim: The aim of our study is to establish the presence and to make morphological and morphofunctional characteristic of ЕС cells in the developing gastrointestinal tract of a human.
Materials and methods: Our study was performed with biopsy specimen from human stomach and duodenum and fragments of gastrointestinal tract of human embryos 6th gestation week, studied by immunohistochemical, electron microscopy and morphometric methods.
Results: EC cells have already been differentiated in the 6th gestation week. Embryonic EC cells had identical characteristics with those of adults. They were in two morphofunctional conditions: stage of increased synthesis and stage of relative secretory rest.
Conclusion: In the early embryonic period - 6th gestation week EC cells have already been differentiated. The occurrence of EC cells with hormonal production prior to the definitive differentiation of tissues presupposes participation of serotonin in the digestive tube histogenetic processes.
Collapse
|
21
|
Poplaski V, Bomidi C, Kambal A, Nguyen-Phuc H, Di Rienzi SC, Danhof HA, Zeng XL, Feagins LA, Deng N, Vilar E, McAllister F, Coarfa C, Min S, Kim HJ, Shukla R, Britton R, Estes MK, Blutt SE. Human intestinal organoids from Cronkhite-Canada syndrome patients reveal link between serotonin and proliferation. J Clin Invest 2023; 133:e166884. [PMID: 37909332 PMCID: PMC10617781 DOI: 10.1172/jci166884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 08/29/2023] [Indexed: 11/03/2023] Open
Abstract
Cronkhite-Canada Syndrome (CCS) is a rare, noninherited polyposis syndrome affecting 1 in every million individuals. Despite over 50 years of CCS cases, the etiopathogenesis and optimal treatment for CCS remains unknown due to the rarity of the disease and lack of model systems. To better understand the etiology of CCS, we generated human intestinal organoids (HIOs) from intestinal stem cells isolated from 2 patients. We discovered that CCS HIOs are highly proliferative and have increased numbers of enteroendocrine cells producing serotonin (also known as 5-hydroxytryptamine or 5HT). These features were also confirmed in patient tissue biopsies. Recombinant 5HT increased proliferation of non-CCS donor HIOs and inhibition of 5HT production in the CCS HIOs resulted in decreased proliferation, suggesting a link between local epithelial 5HT production and control of epithelial stem cell proliferation. This link was confirmed in genetically engineered HIOs with an increased number of enteroendocrine cells. This work provides a new mechanism to explain the pathogenesis of CCS and illustrates the important contribution of HIO cultures to understanding disease etiology and in the identification of novel therapies. Our work demonstrates the principle of using organoids for personalized medicine and sheds light on how intestinal hormones can play a role in intestinal epithelial proliferation.
Collapse
Affiliation(s)
- Victoria Poplaski
- Program in Translational Biology and Molecular Medicine
- Department of Molecular Virology and Microbiology, and
| | | | - Amal Kambal
- Department of Molecular Virology and Microbiology, and
| | | | - Sara C. Di Rienzi
- Department of Molecular Virology and Microbiology, and
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, Texas, USA
| | - Heather A. Danhof
- Department of Molecular Virology and Microbiology, and
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, Texas, USA
| | - Xi-Lei Zeng
- Department of Molecular Virology and Microbiology, and
| | - Linda A. Feagins
- Department of Internal Medicine, Center for Inflammatory Bowl Diseases, The University of Texas at Austin Dell Medical School, Austin, Texas, USA
| | - Nan Deng
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston Texas, USA
| | - Eduardo Vilar
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston Texas, USA
| | - Florencia McAllister
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston Texas, USA
| | - Cristian Coarfa
- Dan L Duncan Comprehensive Cancer Center and
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Soyoun Min
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Hyun Jung Kim
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Richa Shukla
- Department of Medicine, Section of Gasteroenterology and Hepatology, Baylor College of Medicine, Houston, Texas, USA
| | - Robert Britton
- Department of Molecular Virology and Microbiology, and
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, Texas, USA
| | - Mary K. Estes
- Department of Molecular Virology and Microbiology, and
- Department of Medicine, Section of Gasteroenterology and Hepatology, Baylor College of Medicine, Houston, Texas, USA
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston Texas, USA
| | | |
Collapse
|
22
|
Wang Z, Tang H, Liu G, Gong H, Li Y, Chen Y, Yang Y. Compound probiotics producing cellulase could replace cellulase preparations during solid-state fermentation of millet bran. BIORESOURCE TECHNOLOGY 2023; 385:129457. [PMID: 37422095 DOI: 10.1016/j.biortech.2023.129457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/02/2023] [Accepted: 07/03/2023] [Indexed: 07/10/2023]
Abstract
Low-value agricultural by-products can be converted into high-value biological products by fermentation with probiotic strains or by enzymatic hydrolysis. However, the high costs of enzyme preparations significantly limit their applications in fermentation. In this study, the solid-state fermentation of millet bran was performed using a cellulase preparation and compound probiotics producing cellulase (CPPC), respectively. The results showed that both factors effectively destroyed the fiber structure, reduced the crude fiber content by 23.78% and 28.32%, respectively, and significantly increased the contents of beneficial metabolites and microorganisms. Moreover, CPPC could more effectively reduce the anti-nutrient factors and increase the content of anti-inflammatory metabolites. The correlation analysis revealed that Lactiplantibacillus and Issatchenkia had synergistic growth during fermentation. Overall, these results suggested that CPPC could replace cellulase preparation and improve antioxidant properties while reducing anti-nutrient factors of millet bran, thus providing a theoretical reference for the efficient utilization of agricultural by-products.
Collapse
Affiliation(s)
- Zhiwei Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Haoran Tang
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Gongwei Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Hanxuan Gong
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Yangguang Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Yulin Chen
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Yuxin Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, China.
| |
Collapse
|
23
|
Goel KK, Thapliyal S, Kharb R, Joshi G, Negi A, Kumar B. Imidazoles as Serotonin Receptor Modulators for Treatment of Depression: Structural Insights and Structure-Activity Relationship Studies. Pharmaceutics 2023; 15:2208. [PMID: 37765177 PMCID: PMC10535231 DOI: 10.3390/pharmaceutics15092208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/13/2023] [Accepted: 08/19/2023] [Indexed: 09/29/2023] Open
Abstract
Serotoninergic signaling is identified as a crucial player in psychiatric disorders (notably depression), presenting it as a significant therapeutic target for treating such conditions. Inhibitors of serotoninergic signaling (especially selective serotonin reuptake inhibitors (SSRI) or serotonin and norepinephrine reuptake inhibitors (SNRI)) are prominently selected as first-line therapy for the treatment of depression, which benefits via increasing low serotonin levels and norepinephrine by blocking serotonin/norepinephrine reuptake and thereby increasing activity. While developing newer heterocyclic scaffolds to target/modulate the serotonergic systems, imidazole-bearing pharmacophores have emerged. The imidazole-derived pharmacophore already demonstrated unique structural characteristics and an electron-rich environment, ultimately resulting in a diverse range of bioactivities. Therefore, the current manuscript discloses such a specific modification and structural activity relationship (SAR) of attempted derivatization in terms of the serotonergic efficacy of the resultant inhibitor. We also featured a landscape of imidazole-based development, focusing on SAR studies against the serotoninergic system to target depression. This study covers the recent advancements in synthetic methodologies for imidazole derivatives and the development of new molecules having antidepressant activity via modulating serotonergic systems, along with their SAR studies. The focus of the study is to provide structural insights into imidazole-based derivatives as serotonergic system modulators for the treatment of depression.
Collapse
Affiliation(s)
- Kapil Kumar Goel
- Department of Pharmaceutical Sciences, Gurukul Kangri (Deemed to Be University), Haridwar 249404, Uttarakhand, India
| | - Somesh Thapliyal
- Department of Pharmaceutical Sciences, HNB Garhwal University, Chauras Campus, Srinagar Garhwal 246174, Uttarakhand, India (G.J.)
| | - Rajeev Kharb
- Amity Institute of Pharmacy, Amity University, Noida 201313, Uttar Pradesh, India
| | - Gaurav Joshi
- Department of Pharmaceutical Sciences, HNB Garhwal University, Chauras Campus, Srinagar Garhwal 246174, Uttarakhand, India (G.J.)
| | - Arvind Negi
- Department of Bioproduct and Biosystems, Aalto University, 02150 Espoo, Finland
| | - Bhupinder Kumar
- Department of Pharmaceutical Sciences, HNB Garhwal University, Chauras Campus, Srinagar Garhwal 246174, Uttarakhand, India (G.J.)
- Department of Chemistry, Graphic Era (Deemed to Be University), Dehradun 248002, Uttarakhand, India
| |
Collapse
|
24
|
Yoo J, Han J, Lim MH. Transition metal ions and neurotransmitters: coordination chemistry and implications for neurodegeneration. RSC Chem Biol 2023; 4:548-563. [PMID: 37547459 PMCID: PMC10398360 DOI: 10.1039/d3cb00052d] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 06/26/2023] [Indexed: 08/08/2023] Open
Abstract
Neurodegeneration is characterized by a disturbance in neurotransmitter-mediated signaling pathways. Recent studies have highlighted the significant role of transition metal ions, including Cu(i/ii), Zn(ii), and Fe(ii/iii), in neurotransmission, thereby making the coordination chemistry of neurotransmitters a growing field of interest in understanding signal dysfunction. This review outlines the physiological functions of transition metal ions and neurotransmitters, with the metal-binding properties of small molecule-based neurotransmitters and neuropeptides. Additionally, we discuss the structural and conformational changes of neurotransmitters induced by redox-active metal ions, such as Cu(i/ii) and Fe(ii/iii), and briefly describe the outcomes arising from their oxidation, polymerization, and aggregation. These observations have important implications for neurodegeneration and emphasize the need for further research to develop potential therapeutic strategies.
Collapse
Affiliation(s)
- Jeasang Yoo
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Jiyeon Han
- Department of Applied Chemistry, University of Seoul Seoul 02504 Republic of Korea
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| |
Collapse
|
25
|
Shaw C, Hess M, Weimer BC. Microbial-Derived Tryptophan Metabolites and Their Role in Neurological Disease: Anthranilic Acid and Anthranilic Acid Derivatives. Microorganisms 2023; 11:1825. [PMID: 37512997 PMCID: PMC10384668 DOI: 10.3390/microorganisms11071825] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
The gut microbiome provides the host access to otherwise indigestible nutrients, which are often further metabolized by the microbiome into bioactive components. The gut microbiome can also shift the balance of host-produced compounds, which may alter host health. One precursor to bioactive metabolites is the essential aromatic amino acid tryptophan. Tryptophan is mostly shunted into the kynurenine pathway but is also the primary metabolite for serotonin production and the bacterial indole pathway. Balance between tryptophan-derived bioactive metabolites is crucial for neurological homeostasis and metabolic imbalance can trigger or exacerbate neurological diseases. Alzheimer's, depression, and schizophrenia have been linked to diverging levels of tryptophan-derived anthranilic, kynurenic, and quinolinic acid. Anthranilic acid from collective microbiome metabolism plays a complex but important role in systemic host health. Although anthranilic acid and its metabolic products are of great importance for host-microbe interaction in neurological health, literature examining the mechanistic relationships between microbial production, host regulation, and neurological diseases is scarce and at times conflicting. This narrative review provides an overview of the current understanding of anthranilic acid's role in neurological health and disease, with particular focus on the contribution of the gut microbiome, the gut-brain axis, and the involvement of the three major tryptophan pathways.
Collapse
Affiliation(s)
- Claire Shaw
- Department of Population Health and Reproduction, 100K Pathogen Genome Project, University of California Davis, Davis, CA 95616, USA
- Department of Animal Science, College of Agricultural and Environmental Sciences, University of California Davis, Davis, CA 95616, USA
| | - Matthias Hess
- Department of Animal Science, College of Agricultural and Environmental Sciences, University of California Davis, Davis, CA 95616, USA
| | - Bart C Weimer
- Department of Population Health and Reproduction, 100K Pathogen Genome Project, University of California Davis, Davis, CA 95616, USA
| |
Collapse
|
26
|
Ramírez-Perdomo A, Márquez-Barrios G, Gutiérrez-Castañeda LD, Parra-Medina R. NEUROENDOCRINE PEPTIDES IN THE PATHOGENESIS OF COLORECTAL CARCINOMA. Exp Oncol 2023; 45:3-16. [PMID: 37417286 DOI: 10.15407/exp-oncology.2023.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Indexed: 07/08/2023]
Abstract
Colorectal carcinoma (CRC) is the third most frequent neoplasm worldwide and the second leading cause of mortality. Neuroendocrine peptides such as glucagon, bombesin, somatostatin, cholecystokinin, and gastrin as well as growth factors such as platelet-derived growth factor, epidermal growth factor, insulin-like growth factor, and fibroblast growth factor have been postulated as being involved in carcinogenesis. The fact that these neuroendocrine peptides are involved in the development of CRC through the activation of growth factors that stimulate a series of molecular pathways that activate oncogenic signaling mechanisms is emphasized in this review. Peptides such as CCK1, serotonin, and bombesin have been found to be over-expressed in human tumor tissues. Meanwhile, the expression of peptides such as GLP2 has been seen mainly in murine models. The information contained in this review provides a better understanding of the role these peptides play in the pathogenesis of CRC for basic and clinical science studies.
Collapse
Affiliation(s)
- A Ramírez-Perdomo
- Pathology, University Foundation of Health Sciences, Bogota Calle 10 #18-75, ColombiaPathology, University Foundation of Health Sciences, Bogota Calle 10 #18-75, Colombia
| | - G Márquez-Barrios
- Pathology, University Foundation of Health Sciences, Bogota Calle 10 #18-75, Colombia
| | - L D Gutiérrez-Castañeda
- Basic Health Sciences Group, University Foundation of Health Sciences, Bogota, Colombia
- Research Institute, University Foundation of Health Sciences (FUCS), Bogotá, Colombia
| | - R Parra-Medina
- Pathology Department, University Foundation of Health Sciences (FUCS), Bogota Calle 10 #18-75, Colombia
- Research Institute, University Foundation of Health Sciences, Bogota, Colombia
| |
Collapse
|
27
|
The intestinal barrier in disorders of the central nervous system. Lancet Gastroenterol Hepatol 2023; 8:66-80. [PMID: 36334596 DOI: 10.1016/s2468-1253(22)00241-2] [Citation(s) in RCA: 93] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/14/2022] [Accepted: 07/14/2022] [Indexed: 12/12/2022]
Abstract
The intestinal barrier, which primarily consists of a mucus layer, an epithelial barrier, and a gut vascular barrier, has a crucial role in health and disease by facilitating nutrient absorption and preventing the entry of pathogens. The intestinal barrier is in close contact with gut microbiota on its luminal side and with enteric neurons and glial cells on its tissue side. Mounting evidence now suggests that the intestinal barrier is compromised not only in digestive disorders, but also in disorders of the central nervous system (CNS), such as Parkinson's disease, autism spectrum disorder, depression, multiple sclerosis, and Alzheimer's disease. After providing an overview of the structure and functions of the intestinal barrier, we review existing preclinical and clinical studies supporting the notion that intestinal barrier dysfunction is present in neurological, neurodevelopmental, and psychiatric disorders. On the basis of this evidence, we discuss the mechanisms that possibly link gut barrier dysfunction and CNS disorders and the potential impact that evaluating enteric barriers in brain disorders could have on clinical practice, in terms of novel diagnostic and therapeutic strategies, in the near future.
Collapse
|
28
|
Li RQ, Zhao XH, Zhu Q, Liu T, Hondermarck H, Thorne RF, Zhang XD, Gao JN. Exploring neurotransmitters and their receptors for breast cancer prevention and treatment. Theranostics 2023; 13:1109-1129. [PMID: 36793869 PMCID: PMC9925324 DOI: 10.7150/thno.81403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/18/2023] [Indexed: 02/04/2023] Open
Abstract
While psychological factors have long been linked to breast cancer pathogenesis and outcomes, accumulating evidence is revealing how the nervous system contributes to breast cancer development, progression, and treatment resistance. Central to the psychological-neurological nexus are interactions between neurotransmitters and their receptors expressed on breast cancer cells and other types of cells in the tumor microenvironment, which activate various intracellular signaling pathways. Importantly, the manipulation of these interactions is emerging as a potential avenue for breast cancer prevention and treatment. However, an important caveat is that the same neurotransmitter can exert multiple and sometimes opposing effects. In addition, certain neurotransmitters can be produced and secreted by non-neuronal cells including breast cancer cells that similarly activate intracellular signaling upon binding to their receptors. In this review we dissect the evidence for the emerging paradigm linking neurotransmitters and their receptors with breast cancer. Foremost, we explore the intricacies of such neurotransmitter-receptor interactions, including those that impinge on other cellular components of the tumor microenvironment, such as endothelial cells and immune cells. Moreover, we discuss findings where clinical agents used to treat neurological and/or psychological disorders have exhibited preventive/therapeutic effects against breast cancer in either associative or pre-clinical studies. Further, we elaborate on the current progress to identify druggable components of the psychological-neurological nexus that can be exploited for the prevention and treatment of breast cancer as well as other tumor types. We also provide our perspectives regarding future challenges in this field where multidisciplinary cooperation is a paramount requirement.
Collapse
Affiliation(s)
- Ruo Qi Li
- General Surgery Department, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, China.,These authors contributed equally to this work
| | - Xiao Hong Zhao
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, New South Wales, Australia.,These authors contributed equally to this work
| | - Qin Zhu
- General Surgery Department, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, China
| | - Tao Liu
- Children's Cancer Institute Australia for Medical Research, The University of New South Wales, Sydney, NSW, Australia
| | - Hubert Hondermarck
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, New South Wales, Australia
| | - Rick F Thorne
- Translational Research Institute, Henan Provincial and Zhengzhou City Key laboratory of Non-coding RNA and Cancer Metabolism, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Academy of Medical Sciences, Zhengzhou University, Henan, China
| | - Xu Dong Zhang
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, New South Wales, Australia.,Translational Research Institute, Henan Provincial and Zhengzhou City Key laboratory of Non-coding RNA and Cancer Metabolism, Henan International Joint Laboratory of Non-coding RNA and Metabolism in Cancer, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, Academy of Medical Sciences, Zhengzhou University, Henan, China
| | - Jin Nan Gao
- General Surgery Department, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, China
| |
Collapse
|
29
|
Yan T, Shi L, Liu T, Zhang X, Yang M, Peng W, Sun X, Yan L, Dai X, Yang X. Diet-rich in wheat bran modulates tryptophan metabolism and AhR/IL-22 signalling mediated metabolic health and gut dysbacteriosis: A novel prebiotic-like activity of wheat bran. Food Res Int 2023; 163:112179. [PMID: 36596122 DOI: 10.1016/j.foodres.2022.112179] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 11/01/2022] [Accepted: 11/15/2022] [Indexed: 11/21/2022]
Abstract
Tryptophan metabolism has shown to involve in pathogenesis of various metabolic diseases. Gut microbiota-orientated diets hold great potentials to improve metabolic health via regulating tryptophan metabolism. The present study showed that the 6-week high fat diet (HFD) disturbed tryptophan metabolism accompanied with gut dysbacteriosis, also influenced the dietary tryptophan induced changes in cecum microbiome and serum metabolome in mice. The colonic expressions of aryl hydrocarbon receptor (AhR) and interleukin-22 (IL-22) were significantly reduced in mice fed on HFD. Notably, a diet- rich in wheat bran effectively inhibited transformation of tryptophan to kynurenine-pathway metabolites, while increased melatonin and microbial catabolites, i.e. indole-3-propionic acid, indole-3-acetaldehyde and 5-hydroxy-indole-3-acetic acid. Such regulatory effects were accompanied with reduced fasting glucose and total triglycerides, and promoted AhR and IL-22 levels in HFD mice. Wheat bran increased the abundance of health promoting bacteria (e.g., Akkermansia and Lactobacillus), which were significantly correlated with tryptophan derived indolic metabolites. Additionally, beneficial modulatory effects of wheat bran on indolic metabolites in associations with gut dysbacteriosis from type 2 diabetes patients were confirmed in vitro fecal fermentation experiment. Our study proves the detrimental effects of HFD induced gut dysbacteriosis on tryptophan metabolism that may influence immune modulation, and provides novel insights in the mechanisms by which wheat bran could induce health benefits.
Collapse
Affiliation(s)
- Tao Yan
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Lin Shi
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710119, China; Division of Food and Nutrition Science, Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg SE-412 96, Sweden.
| | - Tianqi Liu
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Xiangnan Zhang
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Minmin Yang
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Wen Peng
- Nutrition and Health Promotion Center, Department of Public Health, Medical College, Qinghai University, Xining, Qinghai 810016, China
| | - Xiaomin Sun
- Global Health Institute, Department of Nutrition and Food Safety, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Lijing Yan
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Xiaoshuang Dai
- BGI Institute of Applied Agriculture, BGI-Agro, Shenzhen, Guangdong 518083, China.
| | - Xinbing Yang
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710119, China.
| |
Collapse
|
30
|
Shaw C, Hess M, Weimer BC. Two-component systems regulate bacterial virulence in response to the host gastrointestinal environment and metabolic cues. Virulence 2022; 13:1666-1680. [PMID: 36128741 PMCID: PMC9518994 DOI: 10.1080/21505594.2022.2127196] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Two-component systems are ubiquitous signaling mechanisms in bacteria that enable intracellular changes from extracellular cues. These bacterial regulatory systems couple external stimuli to control genetic expression via an autophosphorylation cascade that transduces membrane signals to intracellular locations, thereby allowing bacteria to rapidly adapt to the changing environmental conditions. Well known to control basic cellular processes, it is evident that two-component systems also exercise control over virulence traits, such as motility, secretion systems, and stress responses that impact the complex cascade of networks that alter virulence traits. In the gastrointestinal system, cues for activation of virulence-related two-component systems include metal ions, host-derived metabolites, and gut conditions. The diversity and origin of these cues suggest that the host can exert control over enteric pathogenicity via regulation in the gastrointestinal system. With the rise in multi-drug resistant pathogens, the potential control of pathogenicity with host cues via two-component systems presents a potential alternative to antimicrobials. Though the signaling mechanism itself is well studied, to date there is no systematic review compiling the host-associated cues of two-component systems and virulence traits. This review highlights the direct link between the host gastrointestinal environment and pathogenicity by focusing on two-component systems that are associated with the genetic expression of virulence traits, and that are activated by host-derived cues. The direct link between the host gastrointestinal environment, metabolites, and pathogenicity established in this review both underscores the importance of host-derived cues on bacterial activity and presents an enticing therapeutic target in the fight against antimicrobial resistant pathogens.
Collapse
Affiliation(s)
- Claire Shaw
- Department of Animal Science, Systems Microbiology & Natural Products Laboratory, University of California, Davis, USA
| | - Matthias Hess
- Department of Animal Science, Systems Microbiology & Natural Products Laboratory, University of California, Davis, USA
| | - Bart C Weimer
- Department of Population Health and Reproduction, 100K Pathogen Genome Project, University of California, Davis, CA, USA
| |
Collapse
|
31
|
Monoamine Neurotransmitters Control Basic Emotions and Affect Major Depressive Disorders. Pharmaceuticals (Basel) 2022; 15:ph15101203. [PMID: 36297314 PMCID: PMC9611768 DOI: 10.3390/ph15101203] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/16/2022] [Accepted: 09/20/2022] [Indexed: 11/17/2022] Open
Abstract
Major depressive disorder (MDD) is a common and complex mental disorder, that adversely impacts an individual’s quality of life, but its diagnosis and treatment are not accurately executed and a symptom-based approach is utilized in most cases, due to the lack of precise knowledge regarding the pathophysiology. So far, the first-line treatments are still based on monoamine neurotransmitters. Even though there is a lot of progress in this field, the mechanisms seem to get more and more confusing, and the treatment is also getting more and more controversial. In this study, we try to review the broad advances of monoamine neurotransmitters in the field of MDD, and update its effects in many advanced neuroscience studies. We still propose the monoamine hypothesis but paid special attention to their effects on the new pathways for MDD, such as inflammation, oxidative stress, neurotrophins, and neurogenesis, especially in the glial cells, which have recently been found to play an important role in many neurodegenerative disorders, including MDD. In addition, we will extend the monoamine hypothesis to basic emotions; as suggested in our previous reports, the three monoamine neurotransmitters play different roles in emotions: dopamine—joy, norepinephrine—fear (anger), serotonins—disgust (sadness). Above all, this paper tries to give a full picture of the relationship between the MDD and the monoamine neurotransmitters such as DA, NE, and 5-HT, as well as their contributions to the Three Primary Color Model of Basic Emotions (joy, fear, and disgust). This is done by explaining the contribution of the monoamine from many sides for MDD, such the digestive tract, astrocytes, microglial, and others, and very briefly addressing the potential of monoamine neurotransmitters as a therapeutic approach for MDD patients and also the reasons for its limited clinical efficacy, side effects, and delayed onset of action. We hope this review might offer new pharmacological management of MDD.
Collapse
|
32
|
Casagrande L, Pastre MJ, Trevizan AR, Cuman RKN, Bersani-Amado CA, Garcia JL, Gois MB, de Mello Gonçales Sant'Ana D, Nogueira-Melo GDA. Moderate intestinal immunopathology after acute oral infection with Toxoplasma gondii oocysts is associated with expressive levels of serotonin. Life Sci 2022; 309:120985. [PMID: 36150462 DOI: 10.1016/j.lfs.2022.120985] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/08/2022] [Accepted: 09/17/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Invasion of the intestinal mucosa by T. gondii elicits a local immune response of variable intensity. These reactions can be lethal in C57BL/6 mice. The tissue damage caused by inflammation and the functional effects depend on the host immunity, strain, and developmental form of the parasite. We investigated the effects of acute oral infection with T. gondii on histoarchitecture, enteric nervous system (ENS), and inflammatory markers in the jejunum and ileum of mice. METHODS Female C57BL/6 mice were divided into a control group and a group orally infected with 1000 sporulated T. gondii oocysts (ME-49 strain). After 5 days, jejunum and ileum were collected and processed for analyzes (e.g., histological and histopathological examinations, ENS, cytokine dosage, myeloperoxidase, nitric oxide activity). MAIN RESULTS In infected mice, we observed a significant increase in serotonin-immunoreactive cells (5-HT IR) in the intestinal mucosa, as well as cellular infiltrates in the lamina propria, periganglionitis, and ganglionitis in the myenteric plexus. We also noted decreased neuron density in the jejunum, increased population of enteric glial cells in the ileum, histomorphometric changes in the intestinal wall, villi, and epithelial cells, remodeling of collagen fibers, and increased myeloperoxidase activity, cytokines, and nitric oxide in the intestine. CONCLUSIONS AND INFERENCES Acute infection of female mice with T. gondii oocysts resulted in changes in ENS and a marked increase in 5-HT. These changes are consistent with its modulatory role in the development of moderate acute inflammation. The use of this experimental model may lend itself to studies aimed at understanding the pathophysiological mechanisms of intestinal inflammation in humans involving ENS.
Collapse
Affiliation(s)
- Lucas Casagrande
- Biosciences and Pathophysiology Postgraduate Program, State University of Maringá, Brazil
| | - Maria José Pastre
- Biosciences and Pathophysiology Postgraduate Program, State University of Maringá, Brazil
| | - Aline Rosa Trevizan
- Biosciences and Pathophysiology Postgraduate Program, State University of Maringá, Brazil
| | | | | | | | - Marcelo Biondaro Gois
- Faculty of Health Sciences, Federal University of Rondonópolis, Brazil; Institute of Health Sciences, Federal University of Bahia, Brazil
| | | | | |
Collapse
|
33
|
Chang WY, Yang YT, She MP, Tu CH, Lee TC, Wu MS, Sun CH, Hsin LW, Yu LCH. 5-HT 7 receptor-dependent intestinal neurite outgrowth contributes to visceral hypersensitivity in irritable bowel syndrome. J Transl Med 2022; 102:1023-1037. [PMID: 36775417 PMCID: PMC9420680 DOI: 10.1038/s41374-022-00800-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 04/22/2022] [Accepted: 04/22/2022] [Indexed: 12/19/2022] Open
Abstract
Irritable bowel syndrome (IBS) is characterized by visceral hypersensitivity (VH) associated with abnormal serotonin/5-hydroxytryptamine (5-HT) metabolism and neurotrophin-dependent mucosal neurite outgrowth. The underlying mechanisms of VH remain poorly understood. We investigated the role of 5-HT7 receptor in mucosal innervation and intestinal hyperalgesia. A high density of mucosal nerve fibres stained for 5-HT7 was observed in colonoscopic biopsy specimens from IBS patients compared with those from healthy controls. Staining of 5-HT3 and 5-HT4 receptors was observed mainly in colonic epithelia with comparable levels between IBS and controls. Visceromotor responses to colorectal distension were evaluated in two mouse models, one postinfectious with Giardia and subjected to water avoidance stress (GW) and the other postinflammatory with trinitrobenzene sulfonic acid-induced colitis (PT). Increased VH was associated with higher mucosal density of 5-HT7-expressing nerve fibres and elevated neurotrophin and neurotrophin receptor levels in the GW and PT mice. The increased VH was inhibited by intraperitoneal injection of SB-269970 (a selective 5-HT7 antagonist). Peroral multiple doses of CYY1005 (a novel 5-HT7 ligand) decreased VH and reduced mucosal density of 5-HT7-expressing nerve fibres in mouse colon. Human neuroblastoma SH-SY5Y cells incubated with bacteria-free mouse colonic supernatant, 5-HT, nerve growth factor, or brain-derived neurotrophic factor exhibited nerve fibre elongation, which was inhibited by 5-HT7 antagonists. Gene silencing of HTR7 also reduced the nerve fibre length. Activation of 5-HT7 upregulated NGF and BDNF gene expression, while stimulation with neurotrophins increased the levels of tryptophan hydroxylase 2 and 5-HT7 in neurons. A positive-feedback loop was observed between serotonin and neurotrophin pathways via 5-HT7 activation to aggravate fibre elongation, whereby 5-HT3 and 5-HT4 had no roles. In conclusion, 5-HT7-dependent mucosal neurite outgrowth contributed to VH. A novel 5-HT7 antagonist could be used as peroral analgesics for IBS-related pain.
Collapse
Affiliation(s)
- Wen-Ying Chang
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan ROC
| | - Yi-Ting Yang
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan ROC
| | - Meng-Ping She
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan ROC
| | - Chia-Hung Tu
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan ROC
| | - Tsung-Chun Lee
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan ROC
| | - Ming-Shiang Wu
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan ROC
| | - Chin-Hung Sun
- Department of Tropical Medicine and Parasitology, National Taiwan University College of Medicine, Taipei, Taiwan ROC
| | - Ling-Wei Hsin
- Graduate Institute of Pharmacy, National Taiwan University School of Pharmacy, Taipei, Taiwan ROC.
- Center for Innovative Therapeutics Discovery, National Taiwan University, Taipei, Taiwan ROC.
| | - Linda Chia-Hui Yu
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan ROC.
| |
Collapse
|