1
|
Nwako JG, McCauley HA. Enteroendocrine cells regulate intestinal homeostasis and epithelial function. Mol Cell Endocrinol 2024; 593:112339. [PMID: 39111616 PMCID: PMC11401774 DOI: 10.1016/j.mce.2024.112339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/23/2024] [Accepted: 08/04/2024] [Indexed: 08/11/2024]
Abstract
Enteroendocrine cells (EECs) are well-known for their systemic hormonal effects, especially in the regulation of appetite and glycemia. Much less is known about how the products made by EECs regulate their local environment within the intestine. Here, we focus on paracrine interactions between EECs and other intestinal cells as they regulate three essential aspects of intestinal homeostasis and physiology: 1) intestinal stem cell function and proliferation; 2) nutrient absorption; and 3) mucosal barrier function. We also discuss the ability of EECs to express multiple hormones, describe in vitro and in vivo models to study EECs, and consider how EECs are altered in GI disease.
Collapse
Affiliation(s)
- Jennifer G Nwako
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill School of Medicine, 111 Mason Farm Road, Molecular Biology Research Building 5341C, Chapel Hill, NC 27599, USA
| | - Heather A McCauley
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill School of Medicine, 111 Mason Farm Road, Molecular Biology Research Building 5341C, Chapel Hill, NC 27599, USA.
| |
Collapse
|
2
|
Johnson CJ, Razy-Krajka F, Zeng F, Piekarz KM, Biliya S, Rothbächer U, Stolfi A. Specification of distinct cell types in a sensory-adhesive organ important for metamorphosis in tunicate larvae. PLoS Biol 2024; 22:e3002555. [PMID: 38478577 PMCID: PMC10962819 DOI: 10.1371/journal.pbio.3002555] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 03/25/2024] [Accepted: 02/21/2024] [Indexed: 03/22/2024] Open
Abstract
The papillae of tunicate larvae contribute sensory, adhesive, and metamorphosis-regulating functions that are crucial for the biphasic lifestyle of these marine, non-vertebrate chordates. We have identified additional molecular markers for at least 5 distinct cell types in the papillae of the model tunicate Ciona, allowing us to further study the development of these organs. Using tissue-specific CRISPR/Cas9-mediated mutagenesis and other molecular perturbations, we reveal the roles of key transcription factors and signaling pathways that are important for patterning the papilla territory into a highly organized array of different cell types and shapes. We further test the contributions of different transcription factors and cell types to the production of the adhesive glue that allows for larval attachment during settlement, and to the processes of tail retraction and body rotation during metamorphosis. With this study, we continue working towards connecting gene regulation to cellular functions that control the developmental transition between the motile larva and sessile adult of Ciona.
Collapse
Affiliation(s)
- Christopher J Johnson
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Florian Razy-Krajka
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Fan Zeng
- Department of Zoology, University of Innsbruck, Innsbruck, Austria
| | - Katarzyna M Piekarz
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Shweta Biliya
- Molecular Evolution Core, Petit H. Parker Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Ute Rothbächer
- Department of Zoology, University of Innsbruck, Innsbruck, Austria
| | - Alberto Stolfi
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| |
Collapse
|
3
|
Uribe RA. Genetic regulation of enteric nervous system development in zebrafish. Biochem Soc Trans 2024; 52:177-190. [PMID: 38174765 PMCID: PMC10903509 DOI: 10.1042/bst20230343] [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: 08/29/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 01/05/2024]
Abstract
The enteric nervous system (ENS) is a complex series of interconnected neurons and glia that reside within and along the entire length of the gastrointestinal tract. ENS functions are vital to gut homeostasis and digestion, including local control of peristalsis, water balance, and intestinal cell barrier function. How the ENS develops during embryological development is a topic of great concern, as defects in ENS development can result in various diseases, the most common being Hirschsprung disease, in which variable regions of the infant gut lack ENS, with the distal colon most affected. Deciphering how the ENS forms from its progenitor cells, enteric neural crest cells, is an active area of research across various animal models. The vertebrate animal model, zebrafish, has been increasingly leveraged to understand early ENS formation, and over the past 20 years has contributed to our knowledge of the genetic regulation that underlies enteric development. In this review, I summarize our knowledge regarding the genetic regulation of zebrafish enteric neuronal development, and based on the most current literature, present a gene regulatory network inferred to underlie its construction. I also provide perspectives on areas for future zebrafish ENS research.
Collapse
Affiliation(s)
- Rosa A. Uribe
- Biosciences Department, Rice University, Houston, TX 77005, U.S.A
- Laboratory of Neural Crest and Enteric Nervous System Development, Rice University, Houston, TX 77005, U.S.A
| |
Collapse
|
4
|
Jones LO, Willms RJ, Xu X, Graham RDV, Eklund M, Shin M, Foley E. Single-cell resolution of the adult zebrafish intestine under conventional conditions and in response to an acute Vibrio cholerae infection. Cell Rep 2023; 42:113407. [PMID: 37948182 DOI: 10.1016/j.celrep.2023.113407] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 10/08/2023] [Accepted: 10/24/2023] [Indexed: 11/12/2023] Open
Abstract
Vibrio cholerae is an aquatic bacterium that causes severe and potentially deadly diarrheal disease. Despite the impact on global health, our understanding of host mucosal responses to Vibrio remains limited, highlighting a knowledge gap critical for the development of effective prevention and treatment strategies. Using a natural infection model, we combine physiological and single-cell transcriptomic studies to characterize conventionally reared adult zebrafish guts and guts challenged with Vibrio. We demonstrate that Vibrio causes a mild mucosal immune response characterized by T cell activation and enhanced antigen capture; Vibrio suppresses host interferon signaling; and ectopic activation of interferon alters the course of infection. We show that the adult zebrafish gut shares similarities with mammalian counterparts, including the presence of Best4+ cells, tuft cells, and a population of basal cycling cells. These findings provide important insights into host-pathogen interactions and emphasize the utility of zebrafish as a natural model of Vibrio infection.
Collapse
Affiliation(s)
- Lena O Jones
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Reegan J Willms
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Xinyue Xu
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Ralph Derrick V Graham
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Mckenna Eklund
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Minjeong Shin
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Edan Foley
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada; Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
5
|
Hibdon ES, Keeley TM, Merchant JL, Samuelson LC. The bHLH transcription factor ASCL1 promotes differentiation of endocrine cells in the stomach and is regulated by Notch signaling. Am J Physiol Gastrointest Liver Physiol 2023; 325:G458-G470. [PMID: 37698169 PMCID: PMC10887855 DOI: 10.1152/ajpgi.00043.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 09/05/2023] [Accepted: 09/05/2023] [Indexed: 09/13/2023]
Abstract
Notch signaling regulates gastrointestinal stem cell proliferation and differentiation yet Notch-regulated transcriptional effectors of gastric epithelial cell differentiation are poorly understood. Here we tested the role of the bHLH transcription factor Achaete-Scute homolog 1 (ASCL1) in gastric epithelial cell differentiation, and its regulation by Notch. Newborn Ascl1 null mice showed a loss of expression of markers of neurogenin-3-dependent enteroendocrine cells, with normal expression of enterochromaffin-like cells, mucous cells, chief cells, and parietal cells. In adult mice, Ascl1 gene expression was observed in the stomach, but not the intestine, with higher expression in antral than corpus epithelium. Lineage tracing in Ascl1-CreERT2; Rosa26-LSL-tdTomato mice revealed single, scattered ASCL1+ cells in the gastric epithelium, demonstrating expression in antral gastrin- and serotonin-producing endocrine cells. ASCL1-expressing endocrine cells persisted for several weeks posttamoxifen labeling with a half-life of approximately 2 months. Lineage tracing in Gastrin-CreERT2 mice demonstrated a similar lifespan for gastrin-producing cells, confirming that gastric endocrine cells are long-lived. Finally, treatment of Ascl1-CreERT2; Rosa26-LSL-tdTomato mice with the pan-Notch inhibitor dibenzazepine increased the number of lineage-labeled cells in the gastric antrum, suggesting that Notch signaling normally inhibits Ascl1 expression. Notch regulation of Ascl1 was also demonstrated in a genetic mouse model of Notch activation, as well as Notch-manipulated antral organoid cultures, thus suggesting that ASCL1 is a key downstream Notch pathway effector promoting endocrine cell differentiation in the gastric epithelium.NEW & NOTEWORTHY Although Notch signaling is known to regulate cellular differentiation in the stomach, downstream effectors are poorly described. Here we demonstrate that the bHLH transcription factor ASCL1 is expressed in endocrine cells in the stomach and is required for formation of neurogenin-3-dependent enteroendocrine cells but not enterochromaffin-like cells. We also demonstrate that Ascl1 expression is inhibited by Notch signaling, suggesting that ASCL1 is a Notch-regulated transcriptional effector directing enteroendocrine cell fate in the mouse stomach.
Collapse
Affiliation(s)
- Elise S Hibdon
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States
| | - Theresa M Keeley
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States
| | - Juanita L Merchant
- Department of Medicine, University of Arizona, Tucson, Arizona, United States
| | - Linda C Samuelson
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States
| |
Collapse
|
6
|
Sree Kumar H, Wisner AS, Refsnider JM, Martyniuk CJ, Zubcevic J. Small fish, big discoveries: zebrafish shed light on microbial biomarkers for neuro-immune-cardiovascular health. Front Physiol 2023; 14:1186645. [PMID: 37324381 PMCID: PMC10267477 DOI: 10.3389/fphys.2023.1186645] [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: 03/15/2023] [Accepted: 05/22/2023] [Indexed: 06/17/2023] Open
Abstract
Zebrafish (Danio rerio) have emerged as a powerful model to study the gut microbiome in the context of human conditions, including hypertension, cardiovascular disease, neurological disorders, and immune dysfunction. Here, we highlight zebrafish as a tool to bridge the gap in knowledge in linking the gut microbiome and physiological homeostasis of cardiovascular, neural, and immune systems, both independently and as an integrated axis. Drawing on zebrafish studies to date, we discuss challenges in microbiota transplant techniques and gnotobiotic husbandry practices. We present advantages and current limitations in zebrafish microbiome research and discuss the use of zebrafish in identification of microbial enterotypes in health and disease. We also highlight the versatility of zebrafish studies to further explore the function of human conditions relevant to gut dysbiosis and reveal novel therapeutic targets.
Collapse
Affiliation(s)
- Hemaa Sree Kumar
- Department of Physiology and Pharmacology, University of Toledo, Toledo, OH, United States
- Department of Neuroscience and Neurological Disorders, University of Toledo, Toledo, OH, United States
| | - Alexander S. Wisner
- Department of Medicinal and Biological Chemistry, University of Toledo, Toledo, OH, United States
- Center for Drug Design and Development, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, United States
| | - Jeanine M. Refsnider
- Department of Environmental Sciences, University of Toledo, Toledo, OH, United States
| | - Christopher J. Martyniuk
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, OH, United States
| | - Jasenka Zubcevic
- Department of Physiology and Pharmacology, University of Toledo, Toledo, OH, United States
| |
Collapse
|
7
|
Reuter AS, Stern D, Bernard A, Goossens C, Lavergne A, Flasse L, Von Berg V, Manfroid I, Peers B, Voz ML. Identification of an evolutionarily conserved domain in Neurod1 favouring enteroendocrine versus goblet cell fate. PLoS Genet 2022; 18:e1010109. [PMID: 35286299 PMCID: PMC8959185 DOI: 10.1371/journal.pgen.1010109] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 03/28/2022] [Accepted: 02/17/2022] [Indexed: 12/16/2022] Open
Abstract
ARP/ASCL transcription factors are key determinants of cell fate specification in a wide variety of tissues, coordinating the acquisition of generic cell fates and of specific subtype identities. How these factors, recognizing highly similar DNA motifs, display specific activities, is not yet fully understood. To address this issue, we overexpressed different ARP/ASCL factors in zebrafish ascl1a-/- mutant embryos to determine which ones are able to rescue the intestinal secretory lineage. We found that Ascl1a/b, Atoh1a/b and Neurod1 factors are all able to trigger the first step of the secretory regulatory cascade but distinct secretory cells are induced by these factors. Indeed, Neurod1 rescues the enteroendocrine lineage while Ascl1a/b and Atoh1a/b rescue the goblet cells. Gain-of-function experiments with Ascl1a/Neurod1 chimeric proteins revealed that the functional divergence is encoded by a 19-aa ultra-conserved element (UCE), present in all Neurod members but absent in the other ARP/ASCL proteins. Importantly, inserting the UCE into the Ascl1a protein reverses the rescuing capacity of this Ascl1a chimeric protein that cannot rescue the goblet cells anymore but can efficiently rescue the enteroendocrine cells. This novel domain acts indeed as a goblet cell fate repressor that inhibits gfi1aa expression, known to be important for goblet cell differentiation. Deleting the UCE domain of the endogenous Neurod1 protein leads to an increase in the number of goblet cells concomitant with a reduction of the enteroendocrine cells, phenotype also observed in the neurod1 null mutant. This highlights the crucial function of the UCE domain for NeuroD1 activity in the intestine. As Gfi1 acts as a binary cell fate switch in several tissues where Neurod1 is also expressed, we can envision a similar role of the UCE in other tissues, allowing Neurod1 to repress Gfi1 to influence the balance between cell fates. It is not yet clear how highly related factors like the ARP/Ascl factors display specific activities even though they recognize the same consensus DNA motif. This specificity could be provided by their cellular environment or by intrinsic properties of the factors themselves. To distinguish between these two possibilities, we have expressed several ARP/Ascl factors in the ascl1a-/- mutant to determine which ones are able to rescue the intestinal secretory defects. We found that Ascl1a/b and Atoh1a/b are able to rescue the goblet cells while Neurod1 rescues the enteroendocrine lineage. Furthermore, we show that the specific Neurod1 activity is conferred by the presence of a 19-aa ultra-conserved element (UCE), present in all vertebrate Neurod members but absent in all the other ARP/ASCL proteins. This UCE domain, so far uncharacterized, acts as a goblet cell fate repressor and inhibits gfi1aa expression, known to be important for goblet cell differentiation. Inserting the UCE into Ascl1a protein reverses the rescuing capacity of this chimeric protein that cannot rescue the goblet cells anymore but can efficiently rescue the enteroendocrine cells. This study therefore highlights an unique intrinsic property of Neurod1 allowing it to repress Gfi1 to influence the balance between cell fates. As Gfi1 acts as a binary cell fate switch in several tissues where Neurod1 is also expressed, we can envision a similar role of the UCE in other tissues, allowing Neurod1 to repress Gfi1 to influence the balance between cell fates.
Collapse
Affiliation(s)
- Anne Sophie Reuter
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Liège, Belgium
| | - David Stern
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Liège, Belgium
| | - Alice Bernard
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Liège, Belgium
| | - Chiara Goossens
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Liège, Belgium
| | - Arnaud Lavergne
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Liège, Belgium
| | - Lydie Flasse
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Liège, Belgium
| | - Virginie Von Berg
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Liège, Belgium
| | - Isabelle Manfroid
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Liège, Belgium
| | - Bernard Peers
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Liège, Belgium
| | - Marianne L. Voz
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Liège, Belgium
- * E-mail:
| |
Collapse
|
8
|
A cell atlas of microbe-responsive processes in the zebrafish intestine. Cell Rep 2022; 38:110311. [PMID: 35108531 DOI: 10.1016/j.celrep.2022.110311] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 10/28/2021] [Accepted: 01/07/2022] [Indexed: 02/08/2023] Open
Abstract
Gut microbial products direct growth, differentiation, and development in animal hosts. However, we lack system-wide understanding of cell-specific responses to the microbiome. We profiled cell transcriptomes from the intestine, and associated tissue, of zebrafish larvae raised in the presence or absence of a microbiome. We uncovered extensive cellular heterogeneity in the conventional zebrafish intestinal epithelium, including previously undescribed cell types with known mammalian homologs. By comparing conventional to germ-free profiles, we mapped microbial impacts on transcriptional activity in each cell population. We revealed intricate degrees of cellular specificity in host responses to the microbiome that included regulatory effects on patterning and on metabolic and immune activity. For example, we showed that the absence of microbes hindered pro-angiogenic signals in the developing vasculature, causing impaired intestinal vascularization. Our work provides a high-resolution atlas of intestinal cellular composition in the developing fish gut and details the effects of the microbiome on each cell type.
Collapse
|
9
|
Interleukin-10 regulates goblet cell numbers through Notch signaling in the developing zebrafish intestine. Mucosal Immunol 2022; 15:940-951. [PMID: 35840681 PMCID: PMC9385495 DOI: 10.1038/s41385-022-00546-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 06/20/2022] [Accepted: 06/27/2022] [Indexed: 02/04/2023]
Abstract
Cytokines are immunomodulatory proteins that orchestrate cellular networks in health and disease. Among these, interleukin (IL)-10 is critical for the establishment of intestinal homeostasis, as mutations in components of the IL-10 signaling pathway result in spontaneous colitis. Whether IL-10 plays other than immunomodulatory roles in the intestines is poorly understood. Here, we report that il10, il10ra, and il10rb are expressed in the zebrafish developing intestine as early as 3 days post fertilization. CRISPR/Cas9-generated il10-deficient zebrafish larvae showed an increased expression of pro-inflammatory genes and an increased number of intestinal goblet cells compared to WT larvae. Mechanistically, Il10 promotes Notch signaling in zebrafish intestinal epithelial cells, which in turn restricts goblet cell expansion. Using murine organoids, we showed that IL-10 modulates goblet cell frequencies in mammals, suggesting conservation across species. This study demonstrates a previously unappreciated IL-10-Notch axis regulating goblet cell homeostasis in the developing zebrafish intestine and may help explain the disease severity of IL-10 deficiency in the intestines of mammals.
Collapse
|
10
|
Zhang C, Huang R, Ma X, Chen J, Han X, Li L, Luo L, Ruan H, Huang H. The Ribosome Biogenesis Factor Ltv1 Is Essential for Digestive Organ Development and Definitive Hematopoiesis in Zebrafish. Front Cell Dev Biol 2021; 9:704730. [PMID: 34692673 PMCID: PMC8528963 DOI: 10.3389/fcell.2021.704730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 09/13/2021] [Indexed: 11/13/2022] Open
Abstract
Ribosome biogenesis is a fundamental activity in cells. Ribosomal dysfunction underlies a category of diseases called ribosomopathies in humans. The symptomatic characteristics of ribosomopathies often include abnormalities in craniofacial skeletons, digestive organs, and hematopoiesis. Consistently, disruptions of ribosome biogenesis in animals are deleterious to embryonic development with hypoplasia of digestive organs and/or impaired hematopoiesis. In this study, ltv1, a gene involved in the small ribosomal subunit assembly, was knocked out in zebrafish by clustered regularly interspaced short palindromic repeats (CRISPRs)/CRISPR associated protein 9 (Cas9) technology. The recessive lethal mutation resulted in disrupted ribosome biogenesis, and ltv1 Δ14/Δ14 embryos displayed hypoplastic craniofacial cartilage, digestive organs, and hematopoiesis. In addition, we showed that the impaired cell proliferation, instead of apoptosis, led to the defects in exocrine pancreas and hematopoietic stem and progenitor cells (HSPCs) in ltv1 Δ14/Δ14 embryos. It was reported that loss of function of genes associated with ribosome biogenesis often caused phenotypes in a P53-dependent manner. In ltv1 Δ14/Δ14 embryos, both P53 protein level and the expression of p53 target genes, Δ113p53 and p21, were upregulated. However, knockdown of p53 failed to rescue the phenotypes in ltv1 Δ14/Δ14 larvae. Taken together, our data demonstrate that LTV1 ribosome biogenesis factor (Ltv1) plays an essential role in digestive organs and hematopoiesis development in zebrafish in a P53-independent manner.
Collapse
Affiliation(s)
- Chong Zhang
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, State Key Laboratory Breeding Base of Eco-Environments and Bio-Resources of the Three Gorges Reservoir Region, School of Life Sciences, Southwest University, Chongqing, China
| | - Rui Huang
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, State Key Laboratory Breeding Base of Eco-Environments and Bio-Resources of the Three Gorges Reservoir Region, School of Life Sciences, Southwest University, Chongqing, China
| | - Xirui Ma
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, State Key Laboratory Breeding Base of Eco-Environments and Bio-Resources of the Three Gorges Reservoir Region, School of Life Sciences, Southwest University, Chongqing, China
| | - Jiehui Chen
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, State Key Laboratory Breeding Base of Eco-Environments and Bio-Resources of the Three Gorges Reservoir Region, School of Life Sciences, Southwest University, Chongqing, China
| | - Xinlu Han
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, State Key Laboratory Breeding Base of Eco-Environments and Bio-Resources of the Three Gorges Reservoir Region, School of Life Sciences, Southwest University, Chongqing, China
| | - Li Li
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, State Key Laboratory Breeding Base of Eco-Environments and Bio-Resources of the Three Gorges Reservoir Region, School of Life Sciences, Southwest University, Chongqing, China
| | - Lingfei Luo
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, State Key Laboratory Breeding Base of Eco-Environments and Bio-Resources of the Three Gorges Reservoir Region, School of Life Sciences, Southwest University, Chongqing, China
| | - Hua Ruan
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, State Key Laboratory Breeding Base of Eco-Environments and Bio-Resources of the Three Gorges Reservoir Region, School of Life Sciences, Southwest University, Chongqing, China
| | - Honghui Huang
- Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, State Key Laboratory Breeding Base of Eco-Environments and Bio-Resources of the Three Gorges Reservoir Region, School of Life Sciences, Southwest University, Chongqing, China
| |
Collapse
|
11
|
Differential Modulation of the Central and Peripheral Monoaminergic Neurochemicals by Deprenyl in Zebrafish Larvae. TOXICS 2021; 9:toxics9060116. [PMID: 34071101 PMCID: PMC8224676 DOI: 10.3390/toxics9060116] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/16/2021] [Accepted: 05/21/2021] [Indexed: 01/27/2023]
Abstract
Zebrafish embryos and larvae are vertebrate models increasingly used in translational neuroscience research. Behavioral impairment induced by the exposure to neuroactive or neurotoxic compounds is commonly linked to changes in modulatory neurotransmitters in the brain. Although different analytical methods for determining monoaminergic neurochemicals in zebrafish larvae have been developed, these methods have been used only on whole larvae, as the dissection of the brain of hundreds of larvae is not feasible. This raises a key question: Are the changes in the monoaminergic profile of the whole larvae predictive of the changes in the brain? In this study, the levels of ten monoaminergic neurotransmitters were determined in the head, trunk, and the whole body of zebrafish larvae in a control group and in those treated for 24 h with 5 M deprenyl, a prototypic monoamine-oxidase B inhibitor, eight days post-fertilization. In control larvae, most of the monoaminergic neurochemicals were found at higher levels in the head than in the trunk. Significant changes were found in the distribution of some neurochemicals after deprenyl-treatment, with serotonin and norepinephrine increasing in both the head and the trunk, whereas dopamine, L-DOPA, and homovanillic acid levels were only modulated in the head. In fact, the highly significant increase in dopamine levels observed in the head after deprenyl-treatment was not detected in the whole-body analysis. These results indicate that the analysis of neurotransmitters in the zebrafish larvae whole-body should not be used as a general surrogate of the brain.
Collapse
|
12
|
Fujii K, Nakajo K, Egashira Y, Yamamoto Y, Kitada K, Taniguchi K, Kawai M, Tomiyama H, Kawakami K, Uchiyama K, Ono F. Gastrointestinal Neurons Expressing HCN4 Regulate Retrograde Peristalsis. Cell Rep 2021; 30:2879-2888.e3. [PMID: 32130893 DOI: 10.1016/j.celrep.2020.02.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 09/30/2019] [Accepted: 02/06/2020] [Indexed: 12/31/2022] Open
Abstract
Peristalsis is indispensable for physiological function of the gut. The enteric nervous system (ENS) plays an important role in regulating peristalsis. While the neural network regulating anterograde peristalsis, which migrates from the oral end to the anal end, is characterized to some extent, retrograde peristalsis remains unresolved with regards to its neural regulation. Using forward genetics in zebrafish, we reveal that a population of neurons expressing a hyperpolarization-activated nucleotide-gated channel HCN4 specifically regulates retrograde peristalsis. When HCN4 channels are blocked by an HCN channel inhibitor or morpholinos blocking the protein expression, retrograde peristalsis is specifically attenuated. Conversely, when HCN4(+) neurons expressing channelrhodopsin are activated by illumination, retrograde peristalsis is enhanced while anterograde peristalsis remains unchanged. We propose that HCN4(+) neurons in the ENS forward activating signals toward the oral end and simultaneously stimulate local circuits regulating the circular muscle.
Collapse
Affiliation(s)
- Kensuke Fujii
- Department of General and Gastroenterological Surgery, Osaka Medical College, Takatsuki, Japan
| | - Koichi Nakajo
- Department of Physiology, Osaka Medical College, Takatsuki, Japan; Division of Integrative Physiology, Department of Physiology, Jichi Medical University, Shimotsuke, Japan
| | | | | | - Kazuya Kitada
- Department of General and Gastroenterological Surgery, Osaka Medical College, Takatsuki, Japan
| | - Kohei Taniguchi
- Department of General and Gastroenterological Surgery, Osaka Medical College, Takatsuki, Japan
| | - Masaru Kawai
- Department of General and Gastroenterological Surgery, Osaka Medical College, Takatsuki, Japan
| | - Hideki Tomiyama
- Department of General and Gastroenterological Surgery, Osaka Medical College, Takatsuki, Japan
| | - Koichi Kawakami
- Laboratory of Molecular and Developmental Biology, National Institute of Genetics and Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies), Hayama, Japan
| | - Kazuhisa Uchiyama
- Department of General and Gastroenterological Surgery, Osaka Medical College, Takatsuki, Japan
| | - Fumihito Ono
- Department of Physiology, Osaka Medical College, Takatsuki, Japan.
| |
Collapse
|
13
|
Ye L, Bae M, Cassilly CD, Jabba SV, Thorpe DW, Martin AM, Lu HY, Wang J, Thompson JD, Lickwar CR, Poss KD, Keating DJ, Jordt SE, Clardy J, Liddle RA, Rawls JF. Enteroendocrine cells sense bacterial tryptophan catabolites to activate enteric and vagal neuronal pathways. Cell Host Microbe 2020; 29:179-196.e9. [PMID: 33352109 DOI: 10.1016/j.chom.2020.11.011] [Citation(s) in RCA: 162] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/08/2020] [Accepted: 11/19/2020] [Indexed: 12/12/2022]
Abstract
The intestinal epithelium senses nutritional and microbial stimuli using epithelial sensory enteroendocrine cells (EEC). EECs communicate nutritional information to the nervous system, but whether they also relay signals from intestinal microbes remains unknown. Using in vivo real-time measurements of EEC and nervous system activity in zebrafish, we discovered that the bacteria Edwardsiella tarda activate EECs through the receptor transient receptor potential ankyrin A1 (Trpa1) and increase intestinal motility. Microbial, pharmacological, or optogenetic activation of Trpa1+EECs directly stimulates vagal sensory ganglia and activates cholinergic enteric neurons by secreting the neurotransmitter 5-hydroxytryptamine (5-HT). A subset of indole derivatives of tryptophan catabolism produced by E. tarda and other gut microbes activates zebrafish EEC Trpa1 signaling. These catabolites also directly stimulate human and mouse Trpa1 and intestinal 5-HT secretion. These results establish a molecular pathway by which EECs regulate enteric and vagal neuronal pathways in response to microbial signals.
Collapse
Affiliation(s)
- Lihua Ye
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, NC 27710, USA; Division of Gastroenterology, Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Munhyung Bae
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Chelsi D Cassilly
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Sairam V Jabba
- Department of Anesthesiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Daniel W Thorpe
- Flinders Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Alyce M Martin
- Flinders Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Hsiu-Yi Lu
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, NC 27710, USA
| | - Jinhu Wang
- Division of Cardiology, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - John D Thompson
- Department of Cell Biology, Regeneration Next, Duke University School of Medicine, Durham, NC 27710, USA
| | - Colin R Lickwar
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, NC 27710, USA; Division of Gastroenterology, Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kenneth D Poss
- Department of Cell Biology, Regeneration Next, Duke University School of Medicine, Durham, NC 27710, USA
| | - Damien J Keating
- Flinders Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Sven-Eric Jordt
- Department of Anesthesiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Jon Clardy
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Rodger A Liddle
- Division of Gastroenterology, Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA; Department of Veterans Affairs, Durham, NC 27705, USA
| | - John F Rawls
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, NC 27710, USA; Division of Gastroenterology, Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
14
|
Xu J, Yu H, Sun X. Less Is More: Rare Pulmonary Neuroendocrine Cells Function as Critical Sensors in Lung. Dev Cell 2020; 55:123-132. [PMID: 33108755 DOI: 10.1016/j.devcel.2020.09.024] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/04/2020] [Accepted: 09/23/2020] [Indexed: 12/14/2022]
Abstract
Pulmonary neuroendocrine cells (PNECs) are rare airway epithelial cells that also uniquely harbor neuronal and endocrine characteristics. In vitro data indicate that these cells respond to chemical or mechanical stimuli by releasing neuropeptides and neurotransmitters, implicating them as airway sensors. Emerging in vivo data corroborate this role and demonstrate that PNECs are important for lung response to signals, such as allergens. With close proximity to steady-state immune cells and innervating nerves, PNECs, as prototype tissue-resident neuroendocrine cells, are at the center of a neuro-immune module that enables the fundamental ability of an organ to sense and respond to the environment.
Collapse
Affiliation(s)
- Jinhao Xu
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA; Department of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Haoze Yu
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Xin Sun
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA; Department of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
15
|
Lavergne A, Tarifeño-Saldivia E, Pirson J, Reuter AS, Flasse L, Manfroid I, Voz ML, Peers B. Pancreatic and intestinal endocrine cells in zebrafish share common transcriptomic signatures and regulatory programmes. BMC Biol 2020; 18:109. [PMID: 32867764 PMCID: PMC7457809 DOI: 10.1186/s12915-020-00840-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 08/04/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Endocrine cells of the zebrafish digestive system play an important role in regulating metabolism and include pancreatic endocrine cells (PECs) clustered in the islets of Langerhans and the enteroendocrine cells (EECs) scattered in the intestinal epithelium. Despite EECs and PECs are being located in distinct organs, their differentiation involves shared molecular mechanisms and transcription factors. However, their degree of relatedness remains unexplored. In this study, we investigated comprehensively the similarity of EECs and PECs by defining their transcriptomic landscape and comparing the regulatory programmes controlled by Pax6b, a key player in both EEC and PEC differentiations. RESULTS RNA sequencing was performed on EECs and PECs isolated from wild-type and pax6b mutant zebrafish. Data mining of wild-type zebrafish EEC data confirmed the expression of orthologues for most known mammalian EEC hormones, but also revealed the expression of three additional neuropeptide hormones (Proenkephalin-a, Calcitonin-a and Adcyap1a) not previously reported to be expressed by EECs in any species. Comparison of transcriptomes from EECs, PECs and other zebrafish tissues highlights a very close similarity between EECs and PECs, with more than 70% of genes being expressed in both endocrine cell types. Comparison of Pax6b-regulated genes in EECs and PECs revealed a significant overlap. pax6b loss-of-function does not affect the total number of EECs and PECs but instead disrupts the balance between endocrine cell subtypes, leading to an increase of ghrelin- and motilin-like-expressing cells in both the intestine and pancreas at the expense of other endocrine cells such as beta and delta cells in the pancreas and pyyb-expressing cells in the intestine. Finally, we show that the homeodomain of Pax6b is dispensable for its action in both EECs and PECs. CONCLUSION We have analysed the transcriptomic landscape of wild-type and pax6b mutant zebrafish EECs and PECs. Our study highlights the close relatedness of EECs and PECs at the transcriptomic and regulatory levels, supporting the hypothesis of a common phylogenetic origin and underscoring the potential implication of EECs in metabolic diseases such as type 2 diabetes.
Collapse
Affiliation(s)
- Arnaud Lavergne
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Avenue de l’Hôpital 1, B34, Sart Tilman, 4000 Liège, Belgium
| | - Estefania Tarifeño-Saldivia
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Avenue de l’Hôpital 1, B34, Sart Tilman, 4000 Liège, Belgium
- Present Address: Gene Expression and Regulation Laboratory, Department of Biochemistry and Molecular Biology, University of Concepción, Concepción, Chile
| | - Justine Pirson
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Avenue de l’Hôpital 1, B34, Sart Tilman, 4000 Liège, Belgium
| | - Anne-Sophie Reuter
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Avenue de l’Hôpital 1, B34, Sart Tilman, 4000 Liège, Belgium
| | - Lydie Flasse
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Avenue de l’Hôpital 1, B34, Sart Tilman, 4000 Liège, Belgium
| | - Isabelle Manfroid
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Avenue de l’Hôpital 1, B34, Sart Tilman, 4000 Liège, Belgium
| | - Marianne L. Voz
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Avenue de l’Hôpital 1, B34, Sart Tilman, 4000 Liège, Belgium
| | - Bernard Peers
- Laboratory of Zebrafish Development and Disease Models (ZDDM), GIGA, University of Liège, Avenue de l’Hôpital 1, B34, Sart Tilman, 4000 Liège, Belgium
| |
Collapse
|
16
|
Kikuchi K, Noh H, Numayama-Tsuruta K, Ishikawa T. Mechanical roles of anterograde and retrograde intestinal peristalses after feeding in a larval fish ( Danio rerio). Am J Physiol Gastrointest Liver Physiol 2020; 318:G1013-G1021. [PMID: 32281395 DOI: 10.1152/ajpgi.00165.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Transport in gut is important, not only for digestion, metabolism, and nutrient uptake, but also for microbiotic circumstance in the digestive tract; however, the effects of mixing and pumping in the intestine have not been fully clarified. Therefore, in this study, we quantitatively explored intestinal mixing and pumping, represented using a dispersion coefficient and pressure rise in zebrafish larvae, which is a model organism for vertebrate digestive studies, over time by measuring transport phenomena after feeding. Here we provide the first quantitative evidence of the roles of anterograde and retrograde intestinal peristalses in the larval fish of Danio rerio after feeding in terms of digestive pumping and mixing functions by an in vivo imaging of intestinal propagation waves in the larval intestine. Peristaltic velocities in the anterior and posterior intestines change considerably after feeding for 5 h, while the intervals and amplitudes remain almost constant. The intestinal transport is successively visualized after feeding to elimination. Moreover, the particle tracking velocimetry in the chyme leads our quantitative understanding of outstanding mixing and pumping functions in the anterior and posterior intestines by adopting physical parameters of diffusivity and pressure rise, respectively. From scaling analysis, we found that the anterior intestine maintains mixing for 5 h from feeding, whereas the posterior intestine activates gradually pumping up. These results suggest that time change of pumping and mixing functions of intestinal peristalsis could considerably influence the nutrient uptake and microbiotic circumstance in the larval fish intestine.NEW & NOTEWORTHY Transport in gut is important, not only for digestion, metabolism, and nutrient uptake, but also for microbiotic circumstance; however, hydrodynamic effects in the intestine have not been fully clarified. We provide the first quantitative evidence of the mechanical roles of anterograde and retrograde intestinal peristalses in the larval fish of Danio rerio by adopting physical parameters of diffusivity and pressure rise. The intestine transitionally regulates mixing and pumping functions by peristaltic propagations after feeding.
Collapse
Affiliation(s)
- Kenji Kikuchi
- Department of Finemechanics, Graduate School of Engineering, Tohoku University, Sendai, Japan.,Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan
| | - Hyeongtak Noh
- Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan
| | | | - Takuji Ishikawa
- Department of Finemechanics, Graduate School of Engineering, Tohoku University, Sendai, Japan.,Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan
| |
Collapse
|
17
|
Li J, Dedloff MR, Stevens K, Maney L, Prochaska M, Hongay CF, Wallace KN. A novel group of secretory cells regulates development of the immature intestinal stem cell niche through repression of the main signaling pathways driving proliferation. Dev Biol 2019; 456:47-62. [PMID: 31398318 DOI: 10.1016/j.ydbio.2019.08.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 07/23/2019] [Accepted: 08/05/2019] [Indexed: 12/12/2022]
Abstract
The intestinal epithelium has constant turnover throughout the life of the organ, with apoptosis of cells at the tips of folds or villi releasing cells into the lumen. Due to constant turnover, epithelial cells need to be constantly replaced. Epithelial cells are supplied by stem cell niches that form at the base of the interfold space (zebrafish) and crypts (birds and mammals). Within the adult stem cell niche of mammals, secretory cells such as Paneth and goblet cells play a role in modulation of proliferation and stem cell activity, producing asymmetric divisions. Progeny of asymmetric divisions move up the fold or villi, giving rise to all of the epithelial cell types. Although much is known about function and organization of the adult intestinal stem cell niche, less is understood about regulation within the immature stem cell compartment. Following smooth muscle formation, the intestinal epithelium folds and proliferation becomes restricted to the interfold base. Symmetric divisions continue in the developing interfold niche until stem cell progeny begin asymmetric divisions, producing progeny that migrate up the developing folds. Proliferative progeny from the developing stem cell niche begin migrating out of the niche during the third week post-embryogenesis (zebrafish) or during the postnatal period (mammals). Regulation and organization of epithelial proliferation in the immature stem cell niche may be regulated by signals comparable to the adult niche. Here we identify a novel subset of secretory cells associated with the developing stem cell niche that receive Notch signaling (referred to as NRSCs). Inhibition of the embryonic NRSCs between 74 hpf to 120 hpf increases epithelial proliferation as well as EGF and IGF signaling. Inhibition of post-embryonic NRSCs (6 hpf to 12 dpf) also increases epithelial proliferation and expression level of Wnt target genes. We conclude that NRSCs play a role in modulation of epithelial proliferation through repression of signaling pathways that drive proliferation during both embryogenesis and the post embryonic period.
Collapse
Affiliation(s)
- Jianlong Li
- Department of Biology, Clarkson University, Potsdam, NY, USA
| | | | - Katrina Stevens
- Department of Biology, Clarkson University, Potsdam, NY, USA
| | - Lea Maney
- Department of Biology, Clarkson University, Potsdam, NY, USA
| | | | - Cintia F Hongay
- Department of Biology, Clarkson University, Potsdam, NY, USA
| | | |
Collapse
|
18
|
Sims KC, Schwendinger KL, Szymkowicz DB, Swetenburg JR, Bain LJ. Embryonic arsenic exposure reduces intestinal cell proliferation and alters hepatic IGF mRNA expression in killifish (Fundulus heteroclitus). JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2019; 82:142-156. [PMID: 30729860 PMCID: PMC6397093 DOI: 10.1080/15287394.2019.1571465] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Arsenic (As) is a toxicant found in food and water throughout the world, and studies suggested that exposure early in life reduces growth. Thus, the goal of this study was to examine mechanisms by which As impacted organismal growth. Killifish (Fundulus heteroclitus) were exposed to 0, 10, 50, or 200 ppb As as embryos and, after hatching, were reared in clean water for up to 40 weeks. Metabolism studies revealed that killifish biotransform As such that monomethylated and dimethylated arsenicals account for 15-17% and 45-61%, respectively, of the total metal. Growth, as measured by condition factor (CF), was significantly and dose-dependently reduced at 8 weeks of age but was similar to controls by 40 weeks. To determine mechanisms underlying the observed initial decrease, intestinal proliferation and morphology were examined. Arsenic-exposed fish exhibited significant 1.3- to 1.5-fold reduction in intestinal villus height and 1.4- to 1.6-fold decrease in proliferating cell nuclear antigen (PCNA+) intestinal cells at all weeks examined. In addition, there were significant correlations between CF, PCNA+ cells, and intestinal villus height. Upon examining whether fish might compensate for the intestinal changes, it was found that hepatic mRNA expression of insulin-like growth factor 1 (IGF-1) and its binding protein (IGFBP-1) were dose-dependently increased. These results indicate that embryonic exposure initially diminished growth, and while intestinal cell proliferation remained reduced, fish appear to compensate by enhancing transcript levels of hepatic IGF-1 and IGFBP-1.
Collapse
Affiliation(s)
- Kaleigh C. Sims
- Environmental Toxicology Graduate Program, Clemson University, Clemson, SC, USA
| | | | - Dana B. Szymkowicz
- Environmental Toxicology Graduate Program, Clemson University, Clemson, SC, USA
| | | | - Lisa J. Bain
- Environmental Toxicology Graduate Program, Clemson University, Clemson, SC, USA
- Department of Biological Sciences, Clemson University, Clemson, SC, USA
| |
Collapse
|
19
|
James DM, Kozol RA, Kajiwara Y, Wahl AL, Storrs EC, Buxbaum JD, Klein M, Moshiree B, Dallman JE. Intestinal dysmotility in a zebrafish ( Danio rerio) shank3a;shank3b mutant model of autism. Mol Autism 2019; 10:3. [PMID: 30733854 PMCID: PMC6357389 DOI: 10.1186/s13229-018-0250-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 11/26/2018] [Indexed: 02/06/2023] Open
Abstract
Background and aims Autism spectrum disorder (ASD) is currently estimated to affect more than 1% of the world population. For people with ASD, gastrointestinal (GI) distress is a commonly reported but a poorly understood co-occurring symptom. Here, we investigate the physiological basis for GI distress in ASD by studying gut function in a zebrafish model of Phelan-McDermid syndrome (PMS), a condition caused by mutations in the SHANK3 gene. Methods To generate a zebrafish model of PMS, we used CRISPR/Cas9 to introduce clinically related C-terminal frameshift mutations in shank3a and shank3b zebrafish paralogues (shank3abΔC). Because PMS is caused by SHANK3 haploinsufficiency, we assessed the digestive tract (DT) structure and function in zebrafish shank3abΔC+/− heterozygotes. Human SHANK3 mRNA was then used to rescue DT phenotypes in larval zebrafish. Results Significantly slower rates of DT peristaltic contractions (p < 0.001) with correspondingly prolonged passage time (p < 0.004) occurred in shank3abΔC+/− mutants. Rescue injections of mRNA encoding the longest human SHANK3 isoform into shank3abΔC+/− mutants produced larvae with intestinal bulb emptying similar to wild type (WT), but still deficits in posterior intestinal motility. Serotonin-positive enteroendocrine cells (EECs) were significantly reduced in both shank3abΔC+/− and shank3abΔC−/− mutants (p < 0.05) while enteric neuron counts and overall structure of the DT epithelium, including goblet cell number, were unaffected in shank3abΔC+/− larvae. Conclusions Our data and rescue experiments support mutations in SHANK3 as causal for GI transit and motility abnormalities. Reductions in serotonin-positive EECs and serotonin-filled ENS boutons suggest an endocrine/neural component to this dysmotility. This is the first study to date demonstrating DT dysmotility in a zebrafish single gene mutant model of ASD. Electronic supplementary material The online version of this article (10.1186/s13229-018-0250-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- David M James
- 1Department of Biology, University of Miami, Coral Gables, FL USA
| | - Robert A Kozol
- 1Department of Biology, University of Miami, Coral Gables, FL USA
| | - Yuji Kajiwara
- 2Seaver Autism Center for Research and Treatment, Department of Psychiatry, Friedman Brain Institute and Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA.,5Denali Therapeutics, South San Francisco, CA USA
| | - Adam L Wahl
- 1Department of Biology, University of Miami, Coral Gables, FL USA
| | - Emily C Storrs
- 1Department of Biology, University of Miami, Coral Gables, FL USA
| | - Joseph D Buxbaum
- 2Seaver Autism Center for Research and Treatment, Department of Psychiatry, Friedman Brain Institute and Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Mason Klein
- 3Department of Physics, University of Miami, Coral Gables, FL USA
| | - Baharak Moshiree
- Division of Gastroenterology, Atrium Health, University of North Carolina, Charlotte, NC USA
| | - Julia E Dallman
- 1Department of Biology, University of Miami, Coral Gables, FL USA
| |
Collapse
|
20
|
Verri T, Werner A. Type II Na +-phosphate Cotransporters and Phosphate Balance in Teleost Fish. Pflugers Arch 2018; 471:193-212. [PMID: 30542786 DOI: 10.1007/s00424-018-2239-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/15/2018] [Accepted: 11/18/2018] [Indexed: 12/27/2022]
Abstract
Teleost fish are excellent models to study the phylogeny of the slc34 gene family, Slc34-mediated phosphate (Pi) transport and how Slc34 transporters contribute Pi homeostasis. Fish need to accumulate Pi from the diet to sustain growth. Much alike in mammals, intestinal uptake in fish is partly a paracellular and partly a Slc34-mediated transcellular process. Acute regulation of Pi balance is achieved in the kidney via a combination of Slc34-mediated secretion and/or reabsorption. A great plasticity is observed in how various species perform and combine the different processes of secretion and reabsorption. A reason for this diversity is found in one or two whole genome duplication events followed by potential gene loss; consequently, teleosts exhibit distinctly different repertoires of Slc34 transporters. Moreover, due to habitats with vastly different salinity, teleosts face the challenge of either preserving water in a hyperosmotic environment (seawater) or excreting water in hypoosmotic freshwater. An additional challenge in understanding teleost Pi homeostasis are the genome duplication and retention events that diversified peptide hormones such as parathyroid hormone and stanniocalcin. Dietary Pi and non-coding RNAs also regulate the expression of piscine Slc34 transporters. The adaptive responses of teleost Slc34 transporters to e.g. Pi diets and vitamin D are informative in the context of comparative physiology, but also relevant in applied physiology and aquaculture. In fact, Pi is essential for teleost fish growth but it also exerts significant adverse consequences if over-supplied. Thus, investigating Slc34 transporters helps tuning the physiology of commercially valuable teleost fish in a confined environment.
Collapse
Affiliation(s)
- Tiziano Verri
- Laboratory of General Physiology, Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy.
| | - Andreas Werner
- Epithelial Research Group, Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
21
|
Yang J, Shimogonya Y, Ishikawa T. What causes the spatial heterogeneity of bacterial flora in the intestine of zebrafish larvae? J Theor Biol 2018. [PMID: 29526663 DOI: 10.1016/j.jtbi.2018.03.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Microbial flora in the intestine has been thoroughly investigated, as it plays an important role in the health of the host. Jemielita et al. (2014) showed experimentally that Aeromonas bacteria in the intestine of zebrafish larvae have a heterogeneous spatial distribution. Although bacterial aggregation is important biologically and clinically, there is no mathematical model describing the phenomenon and its mechanism remains largely unknown. In this study, we developed a computational model to describe the heterogeneous distribution of bacteria in the intestine of zebrafish larvae. The results showed that biological taxis could cause the bacterial aggregation. Intestinal peristalsis had the effect of reducing bacterial aggregation through mixing function. Using a scaling argument, we showed that the taxis velocity of bacteria must be larger than the sum of the diffusive velocity and background bulk flow velocity to induce bacterial aggregation. Our model and findings will be useful to further the scientific understanding of intestinal microbial flora.
Collapse
Affiliation(s)
- Jinyou Yang
- Department of Biomedical Engineering, Graduate School of Biomedical Engineering, Tohoku University, 6-6-01 Aoba, Sendai 980-8579, Japan; School of Fundamental Sciences, China Medical University, Shenyang 110122, China.
| | - Yuji Shimogonya
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, 6-3 Aoba, Sendai 980-8578, Japan
| | - Takuji Ishikawa
- Department of Biomedical Engineering, Graduate School of Biomedical Engineering, Tohoku University, 6-6-01 Aoba, Sendai 980-8579, Japan; Department of Finemechanics, Graduate School of Engineering, Tohoku University, 6-6-01 Aoba, Sendai 980-8579, Japan
| |
Collapse
|
22
|
Dumitrescu E, Wallace KN, Andreescu S. Real time electrochemical investigation of the release, distribution and modulation of nitric oxide in the intestine of individual zebrafish embryos. Nitric Oxide 2018; 74:32-38. [DOI: 10.1016/j.niox.2018.01.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/03/2018] [Accepted: 01/10/2018] [Indexed: 12/16/2022]
|
23
|
Lin B, Coleman JH, Peterson JN, Zunitch MJ, Jang W, Herrick DB, Schwob JE. Injury Induces Endogenous Reprogramming and Dedifferentiation of Neuronal Progenitors to Multipotency. Cell Stem Cell 2017; 21:761-774.e5. [PMID: 29174332 DOI: 10.1016/j.stem.2017.09.008] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 07/12/2017] [Accepted: 09/12/2017] [Indexed: 02/08/2023]
Abstract
Adult neurogenesis in the olfactory epithelium is often depicted as a unidirectional pathway during homeostasis and repair. We challenge the unidirectionality of this model by showing that epithelial injury unlocks the potential for Ascl1+ progenitors and Neurog1+ specified neuronal precursors to dedifferentiate into multipotent stem/progenitor cells that contribute significantly to tissue regeneration in the murine olfactory epithelium (OE). We characterize these dedifferentiating cells using several lineage-tracing strains and single-cell mRNA-seq, and we show that Sox2 is required for initiating dedifferentiation and that inhibition of Ezh2 promotes multipotent progenitor expansion. These results suggest that the apparent hierarchy of neuronal differentiation is not irreversible and that lineage commitment can be overridden following severe tissue injury. We elucidate a previously unappreciated pathway for endogenous tissue repair by a highly regenerative neuroepithelium and introduce a system to study the mechanisms underlying plasticity in the OE that can be adapted for other tissues.
Collapse
Affiliation(s)
- Brian Lin
- Department of Developmental, Molecular and Chemical Biology, School of Medicine, Tufts University, Boston, MA 02111, USA; Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA 02111, USA
| | - Julie H Coleman
- Department of Developmental, Molecular and Chemical Biology, School of Medicine, Tufts University, Boston, MA 02111, USA; Program in Neuroscience, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA 02111, USA
| | - Jesse N Peterson
- Department of Developmental, Molecular and Chemical Biology, School of Medicine, Tufts University, Boston, MA 02111, USA; Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA 02111, USA
| | - Matthew J Zunitch
- Department of Developmental, Molecular and Chemical Biology, School of Medicine, Tufts University, Boston, MA 02111, USA; Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA 02111, USA
| | - Woochan Jang
- Department of Developmental, Molecular and Chemical Biology, School of Medicine, Tufts University, Boston, MA 02111, USA
| | - Daniel B Herrick
- Department of Developmental, Molecular and Chemical Biology, School of Medicine, Tufts University, Boston, MA 02111, USA; Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA 02111, USA
| | - James E Schwob
- Department of Developmental, Molecular and Chemical Biology, School of Medicine, Tufts University, Boston, MA 02111, USA.
| |
Collapse
|
24
|
Hartenstein V, Takashima S, Hartenstein P, Asanad S, Asanad K. bHLH proneural genes as cell fate determinants of entero-endocrine cells, an evolutionarily conserved lineage sharing a common root with sensory neurons. Dev Biol 2017; 431:36-47. [PMID: 28751238 DOI: 10.1016/j.ydbio.2017.07.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 07/14/2017] [Accepted: 07/23/2017] [Indexed: 01/02/2023]
Abstract
Entero-endocrine cells involved in the regulation of digestive function form a large and diverse cell population within the intestinal epithelium of all animals. Together with absorptive enterocytes and secretory gland cells, entero-endocrine cells are generated by the embryonic endoderm and, in the mature animal, from a pool of endoderm derived, self-renewing stem cells. Entero-endocrine cells share many structural/functional and developmental properties with sensory neurons, which hints at the possibility of an ancient evolutionary relationship between these two cell types. We will survey in this article recent findings that emphasize the similarities between entero-endocrine cells and sensory neurons in vertebrates and insects, for which a substantial volume of data pertaining to the entero-endocrine system has been compiled. We will then report new findings that shed light on the specification and morphogenesis of entero-endocrine cells in Drosophila. In this system, presumptive intestinal stem cells (pISCs), generated during early metamorphosis, undergo several rounds of mitosis that produce the endocrine cells and stem cells (ISCs) with which the fly is born. Clonal analysis demonstrated that individual pISCs can give rise to endocrine cells expressing different types of peptides. Immature endocrine cells start out as unpolarized cells located basally of the gut epithelium; they each extend an apical process into the epithelium which establishes a junctional complex and apical membrane specializations contacting the lumen of the gut. Finally, we show that the Drosophila homolog of ngn3, a bHLH gene that defines the entero-endocrine lineage in mammals, is expressed and required for the differentiation of this cell type in the fly gut.
Collapse
Affiliation(s)
- Volker Hartenstein
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095-1606, USA.
| | - Shigeo Takashima
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095-1606, USA
| | - Parvana Hartenstein
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095-1606, USA
| | - Samuel Asanad
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095-1606, USA
| | - Kian Asanad
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095-1606, USA
| |
Collapse
|
25
|
Ma H, Du X, Zhang S, Wang Q, Yin Y, Qiu X, Da P, Yue H, Wu H, Xu F. Achaete-scute complex homologue-1 promotes development of laryngocarcinoma via facilitating the epithelial-mesenchymal transformation. Tumour Biol 2017; 39:1010428317705752. [PMID: 28618959 DOI: 10.1177/1010428317705752] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Laryngeal cancer is one of the most common fatal cancers among head and neck carcinomas, whose mechanism, however, remains unclear. The proneural basic-helix-loop-helix protein achaete-scute complex homologue-1, a member of the basic helix-loop-helix family, plays a very important role in many cancers. This study aims to explore the clinical value and mechanism of achaete-scute complex homologue-1 in laryngeal cancer. Methods including Cell Counting Kit-8, flow cytometry, Transwell invasion assays, and scratch assay were adopted to further explore the bio-function of achaete-scute complex homologue-1, whose expression was examined in fresh and paraffin chip of laryngeal carcinoma tissues by means of western blot and immunohistochemistry, after the interference of achaete-scute complex homologue-1; achaete-scute complex homologue-1, an overexpression in laryngeal carcinoma whose carcinogenicity potential was confirmed via western blot, was correlative with T classification (p = 0.002), histological differentiation (p = 0.000), lymph node metastasis (p = 0.000), and poor survival (p = 0.000). Multivariate analysis shows that achaete-scute complex homologue-1 overexpression is an independent prognostic factor unfavorable to laryngeal carcinoma patients (p = 0.000). Moreover, knocking down achaete-scute complex homologue-1 expression could significantly suppress the proliferation, migration, and invasion of laryngeal carcinoma cell in vitro and disorder epithelial-mesenchymal transformation-associated protein expression. Achaete-scute complex homologue-1 plays an important role in the genesis and progression of laryngeal carcinoma and may act as a potential biomarker for therapeutic target and prognostic prediction.
Collapse
Affiliation(s)
- Huaci Ma
- 1 Department of Otorhinolaryngology, Affiliated Hospital of Nantong University, Nantong, China
| | - Xiaodong Du
- 2 Department of Otolaryngology/Head and Neck Surgery, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Shu Zhang
- 1 Department of Otorhinolaryngology, Affiliated Hospital of Nantong University, Nantong, China
| | - Qiang Wang
- 1 Department of Otorhinolaryngology, Affiliated Hospital of Nantong University, Nantong, China
| | - Yong Yin
- 1 Department of Otorhinolaryngology, Affiliated Hospital of Nantong University, Nantong, China
| | - Xiaoxia Qiu
- 1 Department of Otorhinolaryngology, Affiliated Hospital of Nantong University, Nantong, China
| | - Peng Da
- 1 Department of Otorhinolaryngology, Affiliated Hospital of Nantong University, Nantong, China
| | - Huijun Yue
- 1 Department of Otorhinolaryngology, Affiliated Hospital of Nantong University, Nantong, China
| | - Hao Wu
- 1 Department of Otorhinolaryngology, Affiliated Hospital of Nantong University, Nantong, China
| | - Fenglei Xu
- 3 Department of Otolaryngology/Head and Neck Surgery, The Second Hospital of Shandong University, Jinan, China
| |
Collapse
|
26
|
Mixing and pumping functions of the intestine of zebrafish larvae. J Theor Biol 2017; 419:152-158. [DOI: 10.1016/j.jtbi.2017.02.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 01/30/2017] [Accepted: 02/04/2017] [Indexed: 01/18/2023]
|
27
|
Alcaino C, Farrugia G, Beyder A. Mechanosensitive Piezo Channels in the Gastrointestinal Tract. CURRENT TOPICS IN MEMBRANES 2017; 79:219-244. [PMID: 28728818 PMCID: PMC5606247 DOI: 10.1016/bs.ctm.2016.11.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Sensation of mechanical forces is critical for normal function of the gastrointestinal (GI) tract and abnormalities in mechanosensation are linked to GI pathologies. In the GI tract there are several mechanosensitive cell types-epithelial enterochromaffin cells, intrinsic and extrinsic enteric neurons, smooth muscle cells and interstitial cells of Cajal. These cells use mechanosensitive ion channels that respond to mechanical forces by altering transmembrane ionic currents in a process called mechanoelectrical coupling. Several mechanosensitive ionic conductances have been identified in the mechanosensory GI cells, ranging from mechanosensitive voltage-gated sodium and calcium channels to the mechanogated ion channels, such as the two-pore domain potassium channels K2P (TREK-1) and nonselective cation channels from the transient receptor potential family. The recently discovered Piezo channels are increasingly recognized as significant contributors to cellular mechanosensitivity. Piezo1 and Piezo2 are nonselective cationic ion channels that are directly activated by mechanical forces and have well-defined biophysical and pharmacologic properties. The role of Piezo channels in the GI epithelium is currently under investigation and their role in the smooth muscle syncytium and enteric neurons is still not known. In this review, we outline the current state of knowledge on mechanosensitive ion channels in the GI tract, with a focus on the known and potential functions of the Piezo channels.
Collapse
Affiliation(s)
- C Alcaino
- Mayo Clinic College of Medicine, Rochester, MN, United States
| | - G Farrugia
- Mayo Clinic College of Medicine, Rochester, MN, United States
| | - A Beyder
- Mayo Clinic College of Medicine, Rochester, MN, United States
| |
Collapse
|
28
|
Linan-Rico A, Ochoa-Cortes F, Beyder A, Soghomonyan S, Zuleta-Alarcon A, Coppola V, Christofi FL. Mechanosensory Signaling in Enterochromaffin Cells and 5-HT Release: Potential Implications for Gut Inflammation. Front Neurosci 2016; 10:564. [PMID: 28066160 PMCID: PMC5165017 DOI: 10.3389/fnins.2016.00564] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 11/22/2016] [Indexed: 12/12/2022] Open
Abstract
Enterochromaffin (EC) cells synthesize 95% of the body 5-HT and release 5-HT in response to mechanical or chemical stimulation. EC cell 5-HT has physiological effects on gut motility, secretion and visceral sensation. Abnormal regulation of 5-HT occurs in gastrointestinal disorders and Inflammatory Bowel Diseases (IBD) where 5-HT may represent a key player in the pathogenesis of intestinal inflammation. The focus of this review is on mechanism(s) involved in EC cell "mechanosensation" and critical gaps in our knowledge for future research. Much of our knowledge and concepts are from a human BON cell model of EC, although more recent work has included other cell lines, native EC cells from mouse and human and intact mucosa. EC cells are "mechanosensors" that respond to physical forces generated during peristaltic activity by translating the mechanical stimulus (MS) into an intracellular biochemical response leading to 5-HT and ATP release. The emerging picture of mechanosensation includes Piezo 2 channels, caveolin-rich microdomains, and tight regulation of 5-HT release by purines. The "purinergic hypothesis" is that MS releases purines to act in an autocrine/paracrine manner to activate excitatory (P2Y1, P2Y4, P2Y6, and A2A/A2B) or inhibitory (P2Y12, A1, and A3) receptors to regulate 5-HT release. MS activates a P2Y1/Gαq/PLC/IP3-IP3R/SERCA Ca2+signaling pathway, an A2A/A2B-Gs/AC/cAMP-PKA signaling pathway, an ATP-gated P2X3 channel, and an inhibitory P2Y12-Gi/o/AC-cAMP pathway. In human IBD, P2X3 is down regulated and A2B is up regulated in EC cells, but the pathophysiological consequences of abnormal mechanosensory or purinergic 5-HT signaling remain unknown. EC cell mechanosensation remains poorly understood.
Collapse
Affiliation(s)
- Andromeda Linan-Rico
- Department of Anesthesiology, Wexner Medical Center at Ohio State UniversityColumbus, OH, USA; CONACYT-Centro Universitario de Investigaciones Biomedicas, University of ColimaColima, Mexico
| | - Fernando Ochoa-Cortes
- Department of Anesthesiology, Wexner Medical Center at Ohio State University Columbus, OH, USA
| | - Arthur Beyder
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic Rochester, MN, USA
| | - Suren Soghomonyan
- Department of Anesthesiology, Wexner Medical Center at Ohio State University Columbus, OH, USA
| | - Alix Zuleta-Alarcon
- Department of Anesthesiology, Wexner Medical Center at Ohio State University Columbus, OH, USA
| | - Vincenzo Coppola
- SBS-Cancer Biology and Genetics, Ohio State University Columbus, OH, USA
| | - Fievos L Christofi
- Department of Anesthesiology, Wexner Medical Center at Ohio State University Columbus, OH, USA
| |
Collapse
|
29
|
Wang F, Knutson K, Alcaino C, Linden DR, Gibbons SJ, Kashyap P, Grover M, Oeckler R, Gottlieb PA, Li HJ, Leiter AB, Farrugia G, Beyder A. Mechanosensitive ion channel Piezo2 is important for enterochromaffin cell response to mechanical forces. J Physiol 2016; 595:79-91. [PMID: 27392819 DOI: 10.1113/jp272718] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 06/29/2016] [Indexed: 12/31/2022] Open
Abstract
KEY POINTS The gastrointestinal epithelial enterochromaffin (EC) cell synthesizes the vast majority of the body's serotonin. As a specialized mechanosensor, the EC cell releases this serotonin in response to mechanical forces. However, the molecular mechanism of EC cell mechanotransduction is unknown. In the present study, we show, for the first time, that the mechanosensitive ion channel Piezo2 is specifically expressed by the human and mouse EC cells. Activation of Piezo2 by mechanical forces results in a characteristic ionic current, the release of serotonin and stimulation of gastrointestinal secretion. Piezo2 inhibition by drugs or molecular knockdown decreases mechanosensitive currents, serotonin release and downstream physiological effects. The results of the present study suggest that the mechanosensitive ion channel Piezo2 is specifically expressed by the EC cells of the human and mouse small bowel and that it is important for EC cell mechanotransduction. ABSTRACT The enterochromaffin (EC) cell in the gastrointestinal (GI) epithelium is the source of nearly all systemic serotonin (5-hydroxytryptamine; 5-HT), which is an important neurotransmitter and endocrine, autocrine and paracrine hormone. The EC cell is a specialized mechanosensor, and it is well known that it releases 5-HT in response to mechanical forces. However, the EC cell mechanotransduction mechanism is unknown. The present study aimed to determine whether Piezo2 is involved in EC cell mechanosensation. Piezo2 mRNA was expressed in human jejunum and mouse mucosa from all segments of the small bowel. Piezo2 immunoreactivity localized specifically within EC cells of human and mouse small bowel epithelium. The EC cell model released 5-HT in response to stretch, and had Piezo2 mRNA and protein, as well as a mechanically-sensitive inward non-selective cation current characteristic of Piezo2. Both inward currents and 5-HT release were inhibited by Piezo2 small interfering RNA and antagonists (Gd3+ and D-GsMTx4). Jejunum mucosal pressure increased 5-HT release and short-circuit current via submucosal 5-HT3 and 5-HT4 receptors. Pressure-induced secretion was inhibited by the mechanosensitive ion channel antagonists gadolinium, ruthenium red and D-GsMTx4. We conclude that the EC cells in the human and mouse small bowel GI epithelium selectively express the mechanosensitive ion channel Piezo2, and also that activation of Piezo2 by force leads to inward currents, 5-HT release and an increase in mucosal secretion. Therefore, Piezo2 is critical to EC cell mechanosensitivity and downstream physiological effects.
Collapse
Affiliation(s)
- Fan Wang
- Enteric Neuroscience Program, Division of Gastroenterology & Hepatology, Departments of Medicine and Physiology & Biomedical Engineering, Mayo Clinic, 200 First Street SW, Rochester, MN, USA.,Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 300 Yanchang Middle Road, Shanghai, PR China
| | - Kaitlyn Knutson
- Enteric Neuroscience Program, Division of Gastroenterology & Hepatology, Departments of Medicine and Physiology & Biomedical Engineering, Mayo Clinic, 200 First Street SW, Rochester, MN, USA
| | - Constanza Alcaino
- Enteric Neuroscience Program, Division of Gastroenterology & Hepatology, Departments of Medicine and Physiology & Biomedical Engineering, Mayo Clinic, 200 First Street SW, Rochester, MN, USA
| | - David R Linden
- Enteric Neuroscience Program, Division of Gastroenterology & Hepatology, Departments of Medicine and Physiology & Biomedical Engineering, Mayo Clinic, 200 First Street SW, Rochester, MN, USA
| | - Simon J Gibbons
- Enteric Neuroscience Program, Division of Gastroenterology & Hepatology, Departments of Medicine and Physiology & Biomedical Engineering, Mayo Clinic, 200 First Street SW, Rochester, MN, USA
| | - Purna Kashyap
- Enteric Neuroscience Program, Division of Gastroenterology & Hepatology, Departments of Medicine and Physiology & Biomedical Engineering, Mayo Clinic, 200 First Street SW, Rochester, MN, USA
| | - Madhusudan Grover
- Enteric Neuroscience Program, Division of Gastroenterology & Hepatology, Departments of Medicine and Physiology & Biomedical Engineering, Mayo Clinic, 200 First Street SW, Rochester, MN, USA
| | - Richard Oeckler
- Division of Pulmonary and Critical Care, Departments of Medicine and Physiology & Biomedical Engineering, Mayo Clinic, 200 First Street SW, Rochester, MN, USA
| | - Philip A Gottlieb
- Department of Physiology and Biophysics, State University of New York at Buffalo, 3435 Main Street, Buffalo, NY, USA
| | - Hui Joyce Li
- Department of Medicine, Division of Gastroenterology, University of Massachusetts Medical School, 55 N Lake Ave, Worcester, MA, USA
| | - Andrew B Leiter
- Department of Medicine, Division of Gastroenterology, University of Massachusetts Medical School, 55 N Lake Ave, Worcester, MA, USA
| | - Gianrico Farrugia
- Enteric Neuroscience Program, Division of Gastroenterology & Hepatology, Departments of Medicine and Physiology & Biomedical Engineering, Mayo Clinic, 200 First Street SW, Rochester, MN, USA
| | - Arthur Beyder
- Enteric Neuroscience Program, Division of Gastroenterology & Hepatology, Departments of Medicine and Physiology & Biomedical Engineering, Mayo Clinic, 200 First Street SW, Rochester, MN, USA
| |
Collapse
|
30
|
Zebrafish as a model for understanding enteric nervous system interactions in the developing intestinal tract. Methods Cell Biol 2016; 134:139-64. [PMID: 27312493 DOI: 10.1016/bs.mcb.2016.02.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The enteric nervous system (ENS) forms intimate connections with many other intestinal cell types, including immune cells and bacterial consortia resident in the intestinal lumen. In this review, we highlight contributions of the zebrafish model to understanding interactions among these cells. Zebrafish is a powerful model for forward genetic screens, several of which have uncovered genes previously unknown to be important for ENS development. More recently, zebrafish has emerged as a model for testing functions of genes identified in human patients or large-scale human susceptibility screens. In several cases, zebrafish studies have revealed mechanisms connecting intestinal symptoms with other, seemingly unrelated disease phenotypes. Importantly, chemical library screens in zebrafish have provided startling new insights into potential effects of common drugs on ENS development. A key feature of the zebrafish model is the ability to rear large numbers of animals germ free or in association with only specific bacterial species. Studies utilizing these approaches have demonstrated the importance of bacterial signals for normal intestinal development. These types of studies also show how luminal bacteria and the immune system can contribute to inflammatory processes that can feedback to influence ENS development. The excellent optical properties of zebrafish embryos and larvae, coupled with the ease of generating genetically marked cells of both the host and its resident bacteria, allow visualization of multiple intestinal cell types in living larvae and should promote a more in-depth understanding of intestinal cell interactions, especially interactions between other intestinal cell types and the ENS.
Collapse
|
31
|
Lobert VH, Mouradov D, Heath JK. Focusing the Spotlight on the Zebrafish Intestine to Illuminate Mechanisms of Colorectal Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 916:411-37. [PMID: 27165364 DOI: 10.1007/978-3-319-30654-4_18] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Colorectal cancer, encompassing colon and rectal cancer, arises from the epithelial lining of the large bowel. It is most prevalent in Westernised societies and is increasing in frequency as the world becomes more industrialised. Unfortunately, metastatic colorectal cancer is not cured by chemotherapy and the annual number of deaths caused by colorectal cancer, currently 700,000, is expected to rise. Our understanding of the contribution that genetic mutations make to colorectal cancer, although incomplete, is reasonably well advanced. However, it has only recently become widely appreciated that in addition to the ongoing accumulation of genetic mutations, chronic inflammation also plays a critical role in the initiation and progression of this disease. While a robust and tractable genetic model of colorectal cancer in zebrafish, suitable for pre-clinical studies, is not yet available, the identification of genes required for the rapid proliferation of zebrafish intestinal epithelial cells during development has highlighted a number of essential genes that could be targeted to disable colorectal cancer cells. Moreover, appreciation of the utility of zebrafish to study intestinal inflammation is on the rise. In particular, zebrafish provide unique opportunities to investigate the impact of genetic and environmental factors on the integrity of intestinal epithelial barrier function. With currently available tools, the interplay between epigenetic regulators, intestinal injury, microbiota composition and innate immune cell mobilisation can be analysed in exquisite detail. This provides excellent opportunities to define critical events that could potentially be targeted therapeutically. Further into the future, the use of zebrafish larvae as hosts for xenografts of human colorectal cancer tissue, while still in its infancy, holds great promise that zebrafish could one day provide a practical, preclinical personalized medicine platform for the rapid assessment of the metastatic potential and drug-sensitivity of patient-derived cancers.
Collapse
Affiliation(s)
- Viola H Lobert
- Development and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia.,Department of Biochemistry, Institute for Cancer Research, Oslo University Hospital, Montebello, N-0379, Oslo, Norway
| | - Dmitri Mouradov
- Systems Biology and Personalised Medicine Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
| | - Joan K Heath
- Development and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia. .,Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia.
| |
Collapse
|
32
|
Özel RE, Hayat A, Andreescu S. RECENT DEVELOPMENTS IN ELECTROCHEMICAL SENSORS FOR THE DETECTION OF NEUROTRANSMITTERS FOR APPLICATIONS IN BIOMEDICINE. ANAL LETT 2015; 48:1044-1069. [PMID: 26973348 PMCID: PMC4787221 DOI: 10.1080/00032719.2014.976867] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neurotransmitters are important biological molecules that are essential to many neurophysiological processes including memory, cognition, and behavioral states. The development of analytical methodologies to accurately detect neurotransmitters is of great importance in neurological and biological research. Specifically designed microelectrodes or microbiosensors have demonstrated potential for rapid, real-time measurements with high spatial resolution. Such devices can facilitate study of the role and mechanism of action of neurotransmitters and can find potential uses in biomedicine. This paper reviews the current status and recent advances in the development and application of electrochemical sensors for the detection of small-molecule neurotransmitters. Measurement challenges and opportunities of electroanalytical methods to advance study and understanding of neurotransmitters in various biological models and disease conditions are discussed.
Collapse
Affiliation(s)
- Rıfat Emrah Özel
- Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY, USA. Fax: 3152686610; Tel: 3152682394
| | - Akhtar Hayat
- Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY, USA. Fax: 3152686610; Tel: 3152682394
- Interdisciplinary Research Centre in Biomedical Materials (IRCBM), COMSATS Institute of Information Technology (CIIT), Lahore, Pakistan
| | - Silvana Andreescu
- Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY, USA. Fax: 3152686610; Tel: 3152682394
| |
Collapse
|
33
|
Can the ‘neuron theory’ be complemented by a universal mechanism for generic neuronal differentiation. Cell Tissue Res 2014; 359:343-84. [DOI: 10.1007/s00441-014-2049-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 10/23/2014] [Indexed: 12/19/2022]
|
34
|
Baral R, Ngounou Wetie AG, Darie CC, Wallace KN. Mass spectrometry for proteomics-based investigation using the zebrafish vertebrate model system. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 806:331-40. [PMID: 24952190 DOI: 10.1007/978-3-319-06068-2_15] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The zebrafish (Danio rerio) is frequently being used to investigate the genetics of human diseases as well as resulting pathologies. Ease of both forward and reverse genetic manipulation along with conservation of vertebrate organ systems and disease causing genes has made this system a popular model. Many techniques have been developed to manipulate the genome of zebrafish producing mutants in a vast array of genes. While genetic manipulation of zebrafish has progressed, proteomics have been under-utilized. This review highlights studies that have already been performed using proteomic techniques and as well as our initial proteomic work comparing changes to the proteome between the ascl1a-/- and WT intestine.
Collapse
Affiliation(s)
- Reshica Baral
- Department of Biology, Clarkson University, 8 Clarkson Avenue, Potsdam, NY, 13699-5810, USA
| | | | | | | |
Collapse
|
35
|
Abstract
INTRODUCTION Off-target effects represent one of the major concerns in the development of new pharmaceuticals, requiring large-scale animal toxicity testing. Faster, cheaper and more reliable assays based on zebrafish embryos (ZE) are being developed as major tools for assessing toxicity of chemicals during the drug-discovery process. AREAS COVERED This paper reviews techniques aimed to the analysis of in vivo sublethal toxic effects of drugs on major physiological functions, including the cardiovascular, nervous, neuromuscular, gastrointestinal and thyroid systems among others. Particular emphasis is placed on high-throughput screening techniques (HTS), including robotics, imaging technologies and image-analysis software. EXPERT OPINION The analysis of off-target effects of candidate drugs requires systemic analyses, as they often involve the complete organism rather than specific, tissue- or cell-specific targets. The unique physical and physiological characteristics of ZE make this system an essential tool for drug discovery and toxicity assessment. Different HTS methodologies applicable to ZE allow the screening of large numbers of different chemicals for many diverse and relevant toxic endpoints.
Collapse
Affiliation(s)
- Demetrio Raldúa
- IDAEA-CSIC, Environmental Chemistry , Jordi Girona 18, 08034 Barcelona , Spain +34 93400 6157 ; +34 93204 5904 ;
| | | |
Collapse
|
36
|
Özel RE, Wallace KN, Andreescu S. Alterations of intestinal serotonin following nanoparticle exposure in embryonic zebrafish. ENVIRONMENTAL SCIENCE. NANO 2014; 2014:27-36. [PMID: 24639893 PMCID: PMC3951830 DOI: 10.1039/c3en00001j] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
The increased use of engineered nanoparticles (NPs) in manufacturing and consumer products raises concerns about the potential environmental and health implications on the ecosystem and living organisms. Organs initially and more heavily affected by environmental NPs exposure in whole organisms are the skin and digestive system. We investigate the toxic effect of two types of NPs, nickel (Ni) and copper oxide (CuO), on the physiology of the intestine of a living aquatic system, zebrafish embryos. Embryos were exposed to a range of Ni and CuO NP concentrations at different stages of embryonic development. We use changes in the physiological serotonin (5HT) concentrations, determined electrochemically with carbon fiber microelectrodes inserted in the live embryo, to assess this organ dysfunction due to NP exposure. We find that exposure to both Ni and CuO NPs induces changes in the physiological 5HT concentration that varies with the type, exposure period and concentration of NPs, as well as with the developmental stage during which the embryo is exposed. These data suggest that exposure to NPs might alter development and physiological processes in living organisms and provide evidence of the effect of NPs on the physiology of the intestine.
Collapse
Affiliation(s)
- Rıfat Emrah Özel
- Department of Chemistry and Biomolecular Science, Clarkson University, 8 Clarkson Ave. Potsdam, NY, 13699-5810, USA
| | - Kenneth N. Wallace
- Department of Biology, Clarkson University, 8 Clarkson Ave. Potsdam, NY, 13699-5805, USA
| | - Silvana Andreescu
- Department of Chemistry and Biomolecular Science, Clarkson University, 8 Clarkson Ave. Potsdam, NY, 13699-5810, USA
| |
Collapse
|