1
|
Tümmler B, Pallenberg ST, Dittrich AM, Graeber SY, Naehrlich L, Sommerburg O, Mall MA. Progress of personalized medicine of cystic fibrosis in the times of efficient CFTR modulators. Mol Cell Pediatr 2025; 12:6. [PMID: 40320452 PMCID: PMC12050259 DOI: 10.1186/s40348-025-00194-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 04/03/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Cystic fibrosis (CF) is a systemic disorder of exocrine glands that is caused by mutations in the CFTR gene. MAIN BODY The basic defect in people with CF (pwCF) leads to impaired epithelial transport of chloride and bicarbonate that can be assessed by CFTR biomarkers, i.e. the β-adrenergic sweat rate and sweat chloride concentration (SCC), chloride conductance of the nasal respiratory epithelium (NPD), urine secretion of bicarbonate, intestinal current measurements (ICM) of chloride secretory responses in rectal biopsies and in bioassays of chloride transport in organoids or cell cultures. CFTR modulators are a novel class of drugs that improve defective posttranslational processing, trafficking and function of mutant CFTR. By April 2025, triple combination therapy with the CFTR potentiator ivacaftor (IVA) and the CFTR correctors elexacaftor (ELX) and tezacaftor (TEZ) has been approved in Europe for the treatment of all pwCF who do not carry two minimal function CFTR mutations. Previous phase 3 and post-approval phase 4 studies in pwCF who harbour one or two alleles of the major mutation F508del consistently reported significant improvements of lung function and anthropometry upon initiation of ELX/TEZ/IVA compared to baseline. Normalization of SCC, NPD and ICM correlated with clinical outcomes on the population level, but the restoration of CFTR function was diverse and not predictive for clinical outcome in the individual patient. Theratyping of non-F508del CF genotypes in patient-derived organoids and cell cultures revealed for most cases clinically meaningful increases of CFTR activity upon exposure to ELX/TEZ/IVA. Likewise, every second CF patient with non-F508del genotypes improved in SCC and clinical outcome upon exposure to ELX/TEZ/IVA indicating that triple CFTR modulator therapy is potentially beneficial for all pwCF who do not carry two minimal function CFTR mutations. This group who is not eligible for CFTR modulators may opt for gene addition therapy in the future, as the first-in-human trial with a recombinant lentiviral vector is underway. FUTURE DIRECTIONS The upcoming generation of pwCF will probably experience a rather normal life in childhood and adolescence. To classify the upcoming personal signatures of CF disease in the times of efficient modulators, we need more sensitive CFTR biomarkers that address the long-term course of airway and gut microbiome, host defense, epithelial homeostasis and multiorgan metabolism.
Collapse
Affiliation(s)
- Burkhard Tümmler
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover, 30625, Germany.
- German Center for Lung Research, Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Hannover Medical School, Hannover, Germany.
| | - Sophia Theres Pallenberg
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover, 30625, Germany
| | - Anna-Maria Dittrich
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover, 30625, Germany
- German Center for Lung Research, Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Hannover Medical School, Hannover, Germany
| | - Simon Y Graeber
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, and Cystic Fibrosis Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Center for Lung Research (DZL), Associated Partner Site, Berlin, Germany
| | - Lutz Naehrlich
- Department of Pediatrics, Justus Liebig University Giessen, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Giessen, Germany
| | - Olaf Sommerburg
- Division of Pediatric Pneumology and Allergy, and Cystic Fibrosis Center, Department of Pediatrics, University of Heidelberg, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL),, University of Heidelberg, Heidelberg, Germany
| | - Marcus A Mall
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, and Cystic Fibrosis Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Center for Lung Research (DZL), Associated Partner Site, Berlin, Germany
| |
Collapse
|
2
|
Wang D, Spoelstra WK, Lin L, Akkerman N, Krueger D, Dayton T, van Zon JS, Tans SJ, van Es JH, Clevers H. Interferon-responsive intestinal BEST4/CA7 + cells are targets of bacterial diarrheal toxins. Cell Stem Cell 2025; 32:598-612.e5. [PMID: 40010349 DOI: 10.1016/j.stem.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 09/25/2024] [Accepted: 02/04/2025] [Indexed: 02/28/2025]
Abstract
BEST4/CA7+ cells of the human intestine were recently identified by single-cell RNA sequencing. While their gene expression profile predicts a role in electrolyte balance, BEST4/CA7+ cell function has not been explored experimentally owing to the absence of BEST4/CA7+ cells in mice and the paucity of human in vitro models. Here, we establish a protocol that allows the emergence of BEST4/CA7+ cells in human intestinal organoids. Differentiation of BEST4/CA7+ cells requires activation of Notch signaling and the transcription factor SPIB. BEST4/CA7+ cell numbers strongly increase in response to the cytokine interferon-γ, supporting a role in immunity. Indeed, we demonstrate that BEST4/CA7+ cells generate robust CFTR-mediated fluid efflux when stimulated with bacterial diarrhea-causing toxins and find the norepinephrine-ADRA2A axis as a potential mechanism in blocking BEST4/CA7+ cell-mediated fluid secretion. Our observations identify a central role of BEST4/CA7+ cells in fluid homeostasis in response to bacterial infections.
Collapse
Affiliation(s)
- Daisong Wang
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht 3584 CT, the Netherlands; Oncode Institute, Hubrecht Institute, Utrecht 3584 CT, the Netherlands
| | | | - Lin Lin
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht 3584 CT, the Netherlands; Oncode Institute, Hubrecht Institute, Utrecht 3584 CT, the Netherlands; The Princess Máxima Center for Pediatric Oncology, Utrecht 3584 CS, the Netherlands
| | - Ninouk Akkerman
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht 3584 CT, the Netherlands; Oncode Institute, Hubrecht Institute, Utrecht 3584 CT, the Netherlands
| | - Daniel Krueger
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht 3584 CT, the Netherlands; Oncode Institute, Hubrecht Institute, Utrecht 3584 CT, the Netherlands
| | - Talya Dayton
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht 3584 CT, the Netherlands; Oncode Institute, Hubrecht Institute, Utrecht 3584 CT, the Netherlands
| | | | - Sander J Tans
- AMOLF, Amsterdam 1009 DB, the Netherlands; Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, Delft 2629 HZ, the Netherlands
| | - Johan H van Es
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht 3584 CT, the Netherlands; Oncode Institute, Hubrecht Institute, Utrecht 3584 CT, the Netherlands
| | - Hans Clevers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht 3584 CT, the Netherlands; Oncode Institute, Hubrecht Institute, Utrecht 3584 CT, the Netherlands; The Princess Máxima Center for Pediatric Oncology, Utrecht 3584 CS, the Netherlands.
| |
Collapse
|
3
|
Son GY, Zou A, Wahl A, Huang KT, Zorgit S, Vinu M, Zhou F, Wagner L, Idaghdour Y, Yule DI, Feske S, Lacruz RS. Loss of STIM1 and STIM2 in Salivary Glands Disrupts ANO1 Function but Does Not Induce Sjogren's Disease. FUNCTION 2025; 6:zqae047. [PMID: 39479800 PMCID: PMC11815586 DOI: 10.1093/function/zqae047] [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: 06/23/2024] [Revised: 10/02/2024] [Accepted: 10/28/2024] [Indexed: 11/06/2024] Open
Abstract
Ca2+ signaling via the store-operated Ca2+ entry (SOCE) mediated by STIM1 and STIM2 proteins and the ORAI1 Ca2+ channel is important in saliva fluid secretion and has been associated with Sjogren's disease (SjD). However, there are no studies addressing STIM1/2 dysfunction in salivary glands or SjD in animal models. We report that mice lacking Stim1 and Stim2 [Stim1/2K14Cre(+)] in salivary glands exhibited reduced Ca2+ levels and hyposalivate. SOCE was functionally required for the activation of the Ca2+ activated Cl- channel ANO1. Ageing Stim1/2K14Cre(+) mice showed no evidence of lymphocytic infiltration or increased levels of autoantibodies characteristic of SjD, possibly associated with a downregulation of toll-like receptor 8 (Tlr8) expression. Salivary gland biopsies of SjD patients showed increased expression of STIM1 and TLR7/8. Our study shows that SOCE activates ANO1 function and fluid secretion in salivary glands and highlights a potential link between SOCE and TLR signaling in SjD.
Collapse
Affiliation(s)
- Ga-Yeon Son
- Department of Molecular Pathobiology, New York University College of Dentistry, New York 10010, USA
| | - Anna Zou
- Department of Molecular Pathobiology, New York University College of Dentistry, New York 10010, USA
| | - Amanda Wahl
- Department of Pharmacology and Physiology, University of Rochester, Rochester, New York 14642, USA
| | - Kai Ting Huang
- Department of Pharmacology and Physiology, University of Rochester, Rochester, New York 14642, USA
| | - Saruul Zorgit
- Biology Program, Division of Science and Mathematics, New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates
| | - Manikandan Vinu
- Biology Program, Division of Science and Mathematics, New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates
| | - Fang Zhou
- Department of Pathology, New York University Grossman School of Medicine, New York, New York 10010, USA
| | - Larry Wagner
- Department of Pharmacology and Physiology, University of Rochester, Rochester, New York 14642, USA
| | - Youssef Idaghdour
- Biology Program, Division of Science and Mathematics, New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates
| | - David I Yule
- Department of Pharmacology and Physiology, University of Rochester, Rochester, New York 14642, USA
| | - Stefan Feske
- Department of Pathology, New York University Grossman School of Medicine, New York, New York 10010, USA
| | - Rodrigo S Lacruz
- Department of Molecular Pathobiology, New York University College of Dentistry, New York 10010, USA
| |
Collapse
|
4
|
Dmitriev AV, Linsenmeier RA. pH in the vertebrate retina and its naturally occurring and pathological changes. Prog Retin Eye Res 2025; 104:101321. [PMID: 39608565 PMCID: PMC11711014 DOI: 10.1016/j.preteyeres.2024.101321] [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: 08/23/2024] [Revised: 11/23/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
This review summarizes the existing information on the concentration of H+ (pH) in vertebrate retinae and its changes due to various reasons. Special features of H+ homeostasis that make it different from other ions will be discussed, particularly metabolic production of H+ and buffering. The transretinal distribution of extracellular H+ concentration ([H+]o) and its changes under illumination and other conditions will be described in detail, since [H+]o is more intensively investigated than intracellular pH. In vertebrate retinae, the highest [H+]o occurs in the inner part of the outer nuclear layer, and decreases toward the RPE, reaching the blood level on the apical side of the RPE. [H+]o falls toward the vitreous as well, but less, so that the inner retina is acidic to the vitreous. Light leads to complex changes with both electrogenic and metabolic origins, culminating in alkalinization. There is a rhythm of [H+]o with H+ being higher during circadian night. Extracellular pH can potentially be used as a signal in intercellular volume transmission, but evidence is against pH as a normal controller of fluid transport across the RPE or as a horizontal cell feedback signal. Pathological and experimentally created conditions (systemic metabolic acidosis, hypoxia and ischemia, vascular occlusion, excess glucose and diabetes, genetic disorders, and blockade of carbonic anhydrase) disturb H+ homeostasis, mostly producing retinal acidosis, with consequences for retinal blood flow, metabolism and function.
Collapse
Affiliation(s)
- Andrey V Dmitriev
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA.
| | - Robert A Linsenmeier
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA; Department of Neurobiology, Northwestern University, Evanston, IL, USA; Department of Ophthalmology, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
5
|
Di Palma M, Koh W, Lee CJ, Conti F. A quantitative analysis of bestrophin 1 cellular localization in mouse cerebral cortex. Acta Physiol (Oxf) 2025; 241:e14245. [PMID: 39466647 DOI: 10.1111/apha.14245] [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: 09/11/2024] [Revised: 10/01/2024] [Accepted: 10/08/2024] [Indexed: 10/30/2024]
Abstract
AIM Calcium-activated ligand-gated chloride channels, beyond their role in maintaining anion homeostasis, modulate neuronal excitability by facilitating nonvesicular neurotransmitter release. BEST1, a key member of this family, is permeable to γ-aminobutyric acid (GABA) and glutamate. While astrocytic BEST1 is well-studied and known to regulate neurotransmitter levels, its distribution and role in other brain cell types remain unclear. This study aimed to reassess the localization of BEST1 in the mouse cerebral cortex. METHODS We examined the localization and distribution of BEST1 in the mouse parietal cortex using light microscopy, confocal double-labeling with markers for astrocytes, neurons, microglia, and oligodendrocyte precursor cells, and 3D reconstruction techniques. RESULTS In the cerebral cortex, BEST1 is more broadly distributed than previously thought. Neurons are the second most abundant BEST1+ cell type in the cerebral cortex, following astrocytes. BEST1 is diffusely expressed in neuronal somatic and neuropilar domains and is present at glutamatergic and GABAergic terminals, with a prevalence at GABAergic terminals. We also confirmed that BEST1 is expressed in cortical microglia and identified it in oligodendrocyte precursor cells, albeit to a lesser extent. CONCLUSIONS Together, these findings suggest that BEST1's role in controlling neurotransmission may extend beyond astrocytes to include other brain cells. Understanding BEST1's function in these cells could offer new insights into the molecular mechanisms shaping cortical circuitry. Further research is needed to clarify the diverse roles of BEST1 in both normal and pathophysiological conditions.
Collapse
Affiliation(s)
- Michael Di Palma
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle Marche, Ancona, Italy
| | - Wuhyun Koh
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, South Korea
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, South Korea
| | - Fiorenzo Conti
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Università Politecnica delle Marche, Ancona, Italy
- Center for Neurobiology of Aging, IRCCS INRCA, Ancona, Italy
| |
Collapse
|
6
|
Malonga T, Vialaneix N, Beaumont M. BEST4 + cells in the intestinal epithelium. Am J Physiol Cell Physiol 2024; 326:C1345-C1352. [PMID: 38557358 PMCID: PMC11371329 DOI: 10.1152/ajpcell.00042.2024] [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: 01/19/2024] [Revised: 03/13/2024] [Accepted: 03/21/2024] [Indexed: 04/04/2024]
Abstract
The recent development of single-cell transcriptomics highlighted the existence of a new lineage of mature absorptive cells in the human intestinal epithelium. This subpopulation is characterized by the specific expression of Bestrophin 4 (BEST4) and of other marker genes including OTOP2, CA7, GUCA2A, GUCA2B, and SPIB. BEST4+ cells appear early in development and are present in all regions of the small and large intestine at a low abundance (<5% of all epithelial cells). Location-specific gene expression profiles in BEST4+ cells suggest their functional specialization in each gut region, as exemplified by the small intestine-specific expression of the ion channel CFTR. The putative roles of BEST4+ cells include sensing and regulation of luminal pH, tuning of guanylyl cyclase-C signaling, transport of electrolytes, hydration of mucus, and secretion of antimicrobial peptides. However, most of these hypotheses lack functional validation, notably because BEST4+ cells are absent in mice. The presence of BEST4+ cells in human intestinal organoids indicates that this in vitro model should be suitable to study their role. Recent studies showed that BEST4+ cells are also present in the intestinal epithelium of macaque, pig, and zebrafish and, here, we report their presence in rabbits, which suggests that these species could be appropriate animal models to study BEST4+ cells during the development of diseases and their interactions with environmental factors such as diet or the microbiota. In this review, we summarize the existing literature regarding BEST4+ cells and emphasize the description of their predicted roles in the intestinal epithelium in health and disease.NEW & NOTEWORTHY BEST4+ cells are a novel subtype of mature absorptive cells in the human intestinal epithelium highlighted by single-cell transcriptomics. The gene expression profile of BEST4+ cells suggests their role in pH regulation, electrolyte secretion, mucus hydration, and innate immune defense. The absence of BEST4+ cells in mice requires the use of alternative animal models or organoids to decipher the role of this novel type of intestinal epithelial cells.
Collapse
Affiliation(s)
- Tania Malonga
- GenPhySE, Université de Toulouse, INRAE, ENVT, 31326, Castanet Tolosan, France
- Université de Toulouse, INRAE, UR MIAT, Castanet-Tolosan, France
| | - Nathalie Vialaneix
- Université de Toulouse, INRAE, UR MIAT, Castanet-Tolosan, France
- Université de Toulouse, INRAE, BioinfOmics, GenoToul Bioinformatics Facility, Castanet-Tolosan, France
| | - Martin Beaumont
- GenPhySE, Université de Toulouse, INRAE, ENVT, 31326, Castanet Tolosan, France
| |
Collapse
|
7
|
Son GY, Zou A, Wahl A, Huang KT, Vinu M, Zorgit S, Zhou F, Wagner L, Idaghdour Y, Yule DI, Feske S, Lacruz RS. Loss of STIM1 and STIM2 in salivary glands disrupts ANO1 function but does not induce Sjogren's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.08.574702. [PMID: 38260625 PMCID: PMC10802497 DOI: 10.1101/2024.01.08.574702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Sjogren's disease (SjD) is an autoimmune disease characterized by xerostomia (dry mouth), lymphocytic infiltration into salivary glands and the presence of SSA and SSB autoantibodies. Xerostomia is caused by hypofunction of the salivary glands and has been involved in the development of SjD. Saliva production is regulated by parasympathetic input into the glands initiating intracellular Ca 2+ signals that activate the store operated Ca 2+ entry (SOCE) pathway eliciting sustained Ca 2+ influx. SOCE is mediated by the STIM1 and STIM2 proteins and the ORAI1 Ca 2+ channel. However, there are no studies on the effects of lack of STIM1/2 function in salivary acini in animal models and its impact on SjD. Here we report that male and female mice lacking Stim1 and Stim2 ( Stim1/2 K14Cre ) in salivary glands showed reduced intracellular Ca 2+ levels via SOCE in parotid acini and hyposalivate upon pilocarpine stimulation. Bulk RNASeq of the parotid glands of Stim1/2 K14Cre mice showed a decrease in the expression of Stim1/2 but no other Ca 2+ associated genes mediating saliva fluid secretion. SOCE was however functionally required for the activation of the Ca 2+ activated chloride channel ANO1. Despite hyposalivation, ageing Stim1/2 K14Cre mice showed no evidence of lymphocytic infiltration in the glands or elevated levels of SSA or SSB autoantibodies in the serum, which may be linked to the downregulation of the toll-like receptor 8 ( Tlr8 ). By contrast, salivary gland biopsies of SjD patients showed increased STIM1 and TLR8 expression, and induction of SOCE in a salivary gland cell line increased the expression of TLR8 . Our data demonstrate that SOCE is an important activator of ANO1 function and saliva fluid secretion in salivary glands. They also provide a novel link between SOCE and TLR8 signaling which may explain why loss of SOCE does not result in SjD.
Collapse
|
8
|
Ning H, Liu J, Tan J, Yi M, Lin X. The role of the Notch signalling pathway in the pathogenesis of ulcerative colitis: from the perspective of intestinal mucosal barrier. Front Med (Lausanne) 2024; 10:1333531. [PMID: 38249980 PMCID: PMC10796567 DOI: 10.3389/fmed.2023.1333531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 12/12/2023] [Indexed: 01/23/2024] Open
Abstract
Ulcerative colitis is a common digestive disorder worldwide, with increasing incidence in recent years. It is an urgent problem to be solved, as it seriously affects and threatens the health and life of the global population. Studies have shown that dysfunction of the intestinal mucosal barrier is a critical pathogenic factor and molecular basis of ulcerative colitis, and some scholars have described it as a "barrier organ disease." While the Notch signalling pathway affects a series of cellular processes, including proliferation, differentiation, development, migration, and apoptosis. Therefore, it can regulate intestinal stem cells, CD4+ T cells, innate lymphoid cells, macrophages, and intestinal microbiota and intervene in the chemical, physical, immune, and biological mucosal barriers in cases of ulcerative colitis. The Notch signalling pathway associated with the pathogenesis of ulcerative colitis has distinct characteristics, with good regulatory effects on the mucosal barrier. However, research on ulcerative colitis has mainly focused on immune regulation, anti-inflammatory activity, and antioxidant stress; therefore, the study of the Notch signalling pathway suggests the possibility of understanding the pathogenesis of ulcerative colitis from another perspective. In this article we explore the role and mechanism of the Notch signalling pathway in the pathogenesis of ulcerative colitis from the perspective of the intestinal mucosal barrier to provide new targets and theoretical support for further research on the pathogenesis and clinical treatment of ulcerative colitis.
Collapse
Affiliation(s)
- Hang Ning
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Jiemin Liu
- Guizhou Provincial People’s Hospital, Guiyang, China
| | - Jiaqian Tan
- Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Mengni Yi
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Xiaoyuan Lin
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
9
|
Musco N, Pascon G, Addeo NF, Zarantoniello M, Lanzieri M, Olivotto I, Tulli F, Iervolino V, Amato R, Lombardi P, Bovera F. Cannabidiol can affect morphology, morphometry, enzymatic and microbial activity of rabbit digestive system. J Anim Sci 2024; 102:skae376. [PMID: 39661328 PMCID: PMC11683832 DOI: 10.1093/jas/skae376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 12/10/2024] [Indexed: 12/12/2024] Open
Abstract
The present research aimed to evaluate the effects of the continuative dietary administration of a hemp oil extract containing cannabinoids (cannabidiol, CBD) on the macroscopic morphology, morphometry, and enzymatic activity of different intestinal tracts as well as on the production of short-chain fatty acids (SCFAs) in the cecum of growing rabbits. The research was performed on 16 rabbits randomly selected from 2 experimental groups (8 per group). In detail, 42 sixty-day-old New Zealand White × California rabbits (sex ratio 1:1, average weight 1621.3 ± 46.2 g) were homogeneously divided into 2 groups (21 animals/group), namely control and CBD. Both groups were fed the same commercial diet, but the CBD one was supplemented with 0.1 mL of hemp extract in coconut-based oil corresponding to 10 mg of CBD/animal/d. Up to 92 d of age (for 27 d), individual live weight and feed intake were measured weekly. At 92 d of age, 8 rabbits/group (sex ratio 1:1) were moved to a specialized slaughterhouse, and the gastrointestinal tract was separated from the carcass. Samples from 8 rabbits per dietary treatment were used for the histomorphological analysis of small and large intestines. In addition, duodenum, jejunum, ileum, and cecum were processed for enzymatic analysis. The caecal contents were used for the SCFAs determination. The administration of CBD did not affect feed intake and the final rabbits' whole body weight (P > 0.05), but some changes were detected in the gastrointestinal tract of the animals. CBD seemed to interfere with protein digestion, with a significantly lower activity of the enzymes related to peptides in the small intestine and a consequent increase of the fermentative activity of caecal microbiota. This effect, in combination with a general decrease of fermentative activity in the caecal content of rabbits submitted to CBD treatment, was responsible for a change in the SCFA proportion mainly regarding the reduction of butyrate production (P < 0.01) that resulted significant higher in CTR group compared to CBD. This last result is very important for intestinal health. Such fermentation activity modification was coupled with changes in the relative abundance of goblet cells in the colon. Overall, our findings suggest that a relatively long-term administration of CBD may affect digestion in rabbits, in particular at enzymatic and fermentative levels.
Collapse
Affiliation(s)
- Nadia Musco
- Department of Veterinary Medicine and Animal Production, University of Napoli Federico II, Napoli, Italy
| | - Giulia Pascon
- Department of Agricultural, Food, Environmental and Animal Sciences, University of Udine, Udine, Italy
| | - Nicola Francesco Addeo
- Department of Veterinary Medicine and Animal Production, University of Napoli Federico II, Napoli, Italy
| | - Matteo Zarantoniello
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Mariarosaria Lanzieri
- Department of Veterinary Medicine and Animal Production, University of Napoli Federico II, Napoli, Italy
| | - Ike Olivotto
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Francesca Tulli
- Department of Agricultural, Food, Environmental and Animal Sciences, University of Udine, Udine, Italy
| | - Valeria Iervolino
- Department of Veterinary Medicine and Animal Production, University of Napoli Federico II, Napoli, Italy
| | - Ruggero Amato
- Department of Veterinary Medicine and Animal Production, University of Napoli Federico II, Napoli, Italy
| | - Pietro Lombardi
- Department of Veterinary Medicine and Animal Production, University of Napoli Federico II, Napoli, Italy
| | - Fulvia Bovera
- Department of Veterinary Medicine and Animal Production, University of Napoli Federico II, Napoli, Italy
| |
Collapse
|
10
|
Liu W, Wang Q, Bai Y, Xiao H, Li Z, Wang Y, Wang Q, Yang J, Sun H. Potential Application of Intestinal Organoids in Intestinal Diseases. Stem Cell Rev Rep 2024; 20:124-137. [PMID: 37938407 DOI: 10.1007/s12015-023-10651-w] [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] [Accepted: 10/30/2023] [Indexed: 11/09/2023]
Abstract
To accurately reveal the scenario and mecahnism of gastrointestinal diseases, the establishment of in vitro models of intestinal diseases and drug screening platforms have become the focus of attention. Over the past few decades, animal models and immortalized cell lines have provided valuable but limited insights into gastrointestinal research. In recent years, the development of intestinal organoid culture system has revolutionized in vitro studies of intestinal diseases. Intestinal organoids are derived from self-renewal and self-organization intestinal stem cells (ISCs), which can replicate the genetic characteristics, functions, and structures of the original tissues. Consequently, they provide new stragety for studying various intestinal diseases in vitro. In the review, we will discuss the culture techniques of intestinal organoids and describe the use of intestinal organoids as research tools for intestinal diseases. The role of intestinal epithelial cells (IECs) played in the pathogenesis of inflammatory bowel diseases (IBD) and the treatment of intestinal epithelial dysfunction will be highlighted. Besides, we review the current knowledge on using intestinal organoids as models to study the pathogenesis of IBD caused by epithelial dysfunction and to develop new therapeutic approaches. Finally, we shed light on the current challenges of using intestinal organoids as in vitro models.
Collapse
Affiliation(s)
- Wenxiu Liu
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
- Lanzhou Huazhitiancheng Biotechnologies Co., Ltd, Lanzhou, 730000, Gansu, China
| | - Qian Wang
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Yanrui Bai
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Han Xiao
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Zhunduo Li
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Yan Wang
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China
| | - Qi Wang
- Lanzhou Huazhitiancheng Biotechnologies Co., Ltd, Lanzhou, 730000, Gansu, China.
| | - Jing Yang
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China.
| | - Hui Sun
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, China.
| |
Collapse
|
11
|
Elverson K, Freeman S, Manson F, Warwicker J. Computational Investigation of Mechanisms for pH Modulation of Human Chloride Channels. Molecules 2023; 28:5753. [PMID: 37570721 PMCID: PMC10420675 DOI: 10.3390/molecules28155753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Many transmembrane proteins are modulated by intracellular or extracellular pH. Investigation of pH dependence generally proceeds by mutagenesis of a wide set of amino acids, guided by properties such as amino-acid conservation and structure. Prediction of pKas can streamline this process, allowing rapid and effective identification of amino acids of interest with respect to pH dependence. Commencing with the calcium-activated chloride channel bestrophin 1, the carboxylate ligand structure around calcium sites relaxes in the absence of calcium, consistent with a measured lack of pH dependence. By contrast, less relaxation in the absence of calcium in TMEM16A, and maintenance of elevated carboxylate sidechain pKas, is suggested to give rise to pH-dependent chloride channel activity. This hypothesis, modulation of calcium/proton coupling and pH-dependent activity through the extent of structural relaxation, is shown to apply to the well-characterised cytosolic proteins calmodulin (pH-independent) and calbindin D9k (pH-dependent). Further application of destabilised, ionisable charge sites, or electrostatic frustration, is made to other human chloride channels (that are not calcium-activated), ClC-2, GABAA, and GlyR. Experimentally determined sites of pH modulation are readily identified. Structure-based tools for pKa prediction are freely available, allowing users to focus on mutagenesis studies, construct hypothetical proton pathways, and derive hypotheses such as the model for control of pH-dependent calcium activation through structural flexibility. Predicting altered pH dependence for mutations in ion channel disorders can support experimentation and, ultimately, clinical intervention.
Collapse
Affiliation(s)
- Kathleen Elverson
- Division of Evolution, Infection and Genomics, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Sally Freeman
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Forbes Manson
- Division of Evolution, Infection and Genomics, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Jim Warwicker
- Division of Molecular and Cellular Function, Faculty of Biology, Medicine and Health, Manchester Institute of Biotechnology, The University of Manchester, Manchester M1 7DN, UK
| |
Collapse
|
12
|
Liu Y, Yu Z, Zhu L, Ma S, Luo Y, Liang H, Liu Q, Chen J, Guli S, Chen X. Orchestration of MUC2 - The key regulatory target of gut barrier and homeostasis: A review. Int J Biol Macromol 2023; 236:123862. [PMID: 36870625 DOI: 10.1016/j.ijbiomac.2023.123862] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 03/06/2023]
Abstract
The gut mucosa of human is covered by mucus, functioning as a crucial defense line for the intestine against external stimuli and pathogens. Mucin2 (MUC2) is a subtype of secretory mucins generated by goblet cells and is the major macromolecular component of mucus. Currently, there is an increasing interest on the investigations of MUC2, noting that its function is far beyond a maintainer of the mucus barrier. Moreover, numerous gut diseases are associated with dysregulated MUC2 production. Appropriate production level of MUC2 and mucus contributes to gut barrier function and homeostasis. The production of MUC2 is regulated by a series of physiological processes, which are orchestrated by various bioactive molecules, signaling pathways and gut microbiota, etc., forming a complex regulatory network. Incorporating the latest findings, this review provided a comprehensive summary of MUC2, including its structure, significance and secretory process. Furthermore, we also summarized the molecular mechanisms of the regulation of MUC2 production aiming to provide developmental directions for future researches on MUC2, which can act as a potential prognostic indicator and targeted therapeutic manipulation for diseases. Collectively, we elucidated the micro-level mechanisms underlying MUC2-related phenotypes, hoping to offer some constructive guidance for intestinal and overall health of mankind.
Collapse
Affiliation(s)
- Yaxin Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Zihan Yu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Lanping Zhu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Shuang Ma
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Yang Luo
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Huixi Liang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Qinlingfei Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Jihua Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Sitan Guli
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China
| | - Xin Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China; Tianjin Institute of Digestive Disease, Tianjin Medical University General Hospital, Anshan Road 154, Heping District, Tianjin 300052, China.
| |
Collapse
|
13
|
Owji AP, Yu K, Kittredge A, Wang J, Zhang Y, Yang T. Bestrophin-2 and glutamine synthetase form a complex for glutamate release. Nature 2022; 611:180-187. [PMID: 36289327 PMCID: PMC9873481 DOI: 10.1038/s41586-022-05373-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 09/21/2022] [Indexed: 01/26/2023]
Abstract
Bestrophin-2 (BEST2) is a member of the bestrophin family of calcium-activated anion channels that has a critical role in ocular physiology1-4. Here we uncover a directional permeability of BEST2 to glutamate that heavily favours glutamate exit, identify glutamine synthetase (GS) as a binding partner of BEST2 in the ciliary body of the eye, and solve the structure of the BEST2-GS complex. BEST2 reduces cytosolic GS activity by tethering GS to the cell membrane. GS extends the ion conducting pathway of BEST2 through its central cavity and inhibits BEST2 channel function in the absence of intracellular glutamate, but sensitizes BEST2 to intracellular glutamate, which promotes the opening of BEST2 and thus relieves the inhibitory effect of GS. We demonstrate the physiological role of BEST2 in conducting chloride and glutamate and the influence of GS in non-pigmented ciliary epithelial cells. Together, our results reveal a novel mechanism of glutamate release through BEST2-GS.
Collapse
Affiliation(s)
- Aaron P Owji
- Department of Ophthalmology, Columbia University, New York, NY, USA
- Department of Pharmacology, Columbia University, New York, NY, USA
| | - Kuai Yu
- Department of Cell Biology, Emory University, School of Medicine, Atlanta, GA, USA
| | - Alec Kittredge
- Department of Ophthalmology, Columbia University, New York, NY, USA
- Department of Pharmacology, Columbia University, New York, NY, USA
| | - Jiali Wang
- Department of Ophthalmology, Columbia University, New York, NY, USA
| | - Yu Zhang
- Department of Ophthalmology, Columbia University, New York, NY, USA.
| | - Tingting Yang
- Department of Ophthalmology, Columbia University, New York, NY, USA.
| |
Collapse
|
14
|
Owji AP, Wang J, Kittredge A, Clark Z, Zhang Y, Hendrickson WA, Yang T. Structures and gating mechanisms of human bestrophin anion channels. Nat Commun 2022; 13:3836. [PMID: 35789156 PMCID: PMC9253114 DOI: 10.1038/s41467-022-31437-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 06/17/2022] [Indexed: 01/16/2023] Open
Abstract
Bestrophin-1 (Best1) and bestrophin-2 (Best2) are two members of the bestrophin family of calcium (Ca2+)-activated chloride (Cl−) channels with critical involvement in ocular physiology and direct pathological relevance. Here, we report cryo-EM structures of wild-type human Best1 and Best2 in various states at up to 1.8 Å resolution. Ca2+-bound Best1 structures illustrate partially open conformations at the two Ca2+-dependent gates of the channels, in contrast to the fully open conformations observed in Ca2+-bound Best2, which is in accord with the significantly smaller currents conducted by Best1 in electrophysiological recordings. Comparison of the closed and open states reveals a C-terminal auto-inhibitory segment (AS), which constricts the channel concentrically by wrapping around the channel periphery in an inter-protomer manner and must be released to allow channel opening. Our results demonstrate that removing the AS from Best1 and Best2 results in truncation mutants with similar activities, while swapping the AS between Best1 and Best2 results in chimeric mutants with swapped activities, underlying a key role of the AS in determining paralog specificity among bestrophins. Bestrophin channels are critical for physiology of the eye. Here, authors report cryo-EM structures of human bestrophins in various states at up to 1.8 Å resolution, revealing paralog-specific features that underlie molecular mechanisms of permeation.
Collapse
Affiliation(s)
- Aaron P Owji
- Department of Ophthalmology, Columbia University, New York, NY, USA.,Department of Pharmacology, Columbia University, New York, NY, USA
| | - Jiali Wang
- Department of Ophthalmology, Columbia University, New York, NY, USA
| | - Alec Kittredge
- Department of Ophthalmology, Columbia University, New York, NY, USA.,Department of Pharmacology, Columbia University, New York, NY, USA
| | - Zada Clark
- Department of Ophthalmology, Columbia University, New York, NY, USA
| | - Yu Zhang
- Department of Ophthalmology, Columbia University, New York, NY, USA.
| | - Wayne A Hendrickson
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA. .,Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA. .,New York Structural Biology Center, New York, NY, USA.
| | - Tingting Yang
- Department of Ophthalmology, Columbia University, New York, NY, USA.
| |
Collapse
|
15
|
Du P, Chai Y, Zong S, Yue J, Xiao H. Identification of a Prognostic Model Based on Fatty Acid Metabolism-Related Genes of Head and Neck Squamous Cell Carcinoma. Front Genet 2022; 13:888764. [PMID: 35846149 PMCID: PMC9280184 DOI: 10.3389/fgene.2022.888764] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 06/09/2022] [Indexed: 01/12/2023] Open
Abstract
The fatty acid metabolism (FAM) is known to impact tumorigenesis, tumor progression and treatment resistance via enhancing lipid synthesis, storage and catabolism. However, the role of FAM in head and neck squamous cell carcinoma (HNSCC) has remained elusive. In the present study, we obtained a total of 69 differentially expressed FAM-related genes between 502 HNSCC samples and 44 normal samples from The Cancer Genome Atlas (TCGA) database. The HNSCC samples were divided into 2 clusters according to 69 differentially expressed genes (DEGs) via cluster analysis. Then DEGs in the two clusters were found, and 137 prognostic DEGs were identified by univariate analysis. Subsequently, combined with the clinical information of 546 HNSCC patients from TCGA database, a 12-gene prognostic risk model was established (FEPHX3, SPINK7, FCRLA, MASP1, ZNF541, CD5, BEST2 and ZAP70 were down-regulation, ADPRHL1, DYNC1I1, KCNG1 and LINC00460 were up-regulation) using multivariate Cox regression and LASSO regression analysis. The risk scores of 546 HNSCC samples were calculated. According to the median risk score, 546 HNSCC patients were divided into the high- and low-risk (high- and low score) groups. The Kaplan-Meier survival analysis showed that the survival time of HNSCC patients was significantly shorter in the high-risk group than that in the low-risk group (p < 0.001). The same conclusion was obtained in the Gene Expression Omnibus (GEO) dataset. After that, the multivariate Cox regression analysis indicated that the risk score was an independent factor for patients with HNSCC in the TCGA cohort. In addition, single-sample gene set enrichment analysis (ssGSEA) indicated that the level of infiltrating immune cells was relatively low in the high-risk group compared with the low-risk group. In summary, FAM-related gene expression-based risk signature could predict the prognosis of HNSCC independently.
Collapse
Affiliation(s)
- Peiyu Du
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Chai
- Department of Medical Oncology, National Cancer Cente, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shimin Zong
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianxin Yue
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Jianxin Yue, ; Hongjun Xiao,
| | - Hongjun Xiao
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Jianxin Yue, ; Hongjun Xiao,
| |
Collapse
|
16
|
Self-organization and surface properties of hBest1 in models of biological membranes. Adv Colloid Interface Sci 2022; 302:102619. [PMID: 35276535 DOI: 10.1016/j.cis.2022.102619] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/19/2022] [Accepted: 02/20/2022] [Indexed: 11/22/2022]
Abstract
The transmembrane Ca2+ - activated Cl- channel - human bestrophin-1 (hBest1) is expressed in retinal pigment epithelium and mutations of BEST1 gene cause ocular degenerative diseases colectivelly referred to as "bestrophinopathies". A large number of genetical, biochemical, biophysical and molecular biological studies have been performed to understand the relationship between structure and function of the hBest1 protein and its pathophysiological significance. Here, we review the current understanding of hBest1 surface organization, interactions with membrane lipids in model membranes, and its association with microdomains of cellular membranes. These highlights are significant for modulation of channel activity in cells.
Collapse
|
17
|
Gonzalez-Perez V, Zhou Y, Ciorba MA, Lingle CJ. The LRRC family of BK channel regulatory subunits: potential roles in health and disease. J Physiol 2022; 600:1357-1371. [PMID: 35014034 PMCID: PMC8930516 DOI: 10.1113/jp281952] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 01/04/2022] [Indexed: 11/08/2022] Open
Abstract
Large conductance K+ channels, termed BK channels, regulate a variety of cellular and physiological functions. Although universally activated by changes in voltage or [Ca2+ ]i , the threshold for BK channel activation varies among loci of expression, often arising from cell-specific regulatory subunits including a family of leucine rich repeat-containing (LRRC) γ subunits (LRRC26, LRRC52, LRRC55 and LRRC38). The 'founding' member of this family, LRRC26, was originally identified as a tumour suppressor in various cancers. An LRRC26 knockout (KO) mouse model recently revealed that LRRC26 is also highly expressed in secretory epithelial cells and partners with BK channels in the salivary gland and colonic goblet cells to promote sustained K+ fluxes likely essential for normal secretory function. To accomplish this, LRRC26 negatively shifts the range of BK channel activation such that channels contribute to K+ flux near typical epithelial cell resting conditions. In colon, the absence of LRRC26 increases vulnerability to colitis. LRRC26-containing BK channels are also likely important regulators of epithelial function in other loci, including airways, female reproductive tract and mammary gland. Based on an LRRC52 KO mouse model, LRRC52 regulation of large conductance K+ channels plays a role both in sperm function and in cochlear inner hair cells. Although our understanding of LRRC-containing BK channels remains rudimentary, KO mouse models may help define other organs in which LRRC-containing channels support normal function. A key topic for future work concerns identification of endogenous mechanisms, whether post-translational or via gene regulation, that may impact LRRC-dependent pathologies.
Collapse
Affiliation(s)
- Vivian Gonzalez-Perez
- Department of Anesthesiology, Washington University School of Medicine, St Louis, MO, USA
| | - Yu Zhou
- Department of Anesthesiology, Washington University School of Medicine, St Louis, MO, USA
| | - Matthew A Ciorba
- Department of Internal Medicine, Division of Gastroenterology, Washington University School of Medicine, St Louis, MO, USA
| | - Christopher J Lingle
- Department of Anesthesiology, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
18
|
He XS, Ye WL, Zhang YJ, Yang XQ, Liu F, Wang JR, Ding XL, Yang Y, Zhang RN, Zhao YY, Bi HX, Guo LC, Gan WJ, Wu H. Oncogenic potential of BEST4 in colorectal cancer via activation of PI3K/Akt signaling. Oncogene 2022; 41:1166-1177. [PMID: 35058597 DOI: 10.1038/s41388-021-02160-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 12/02/2021] [Accepted: 12/14/2021] [Indexed: 12/11/2022]
Abstract
BEST4 is a member of the bestrophin protein family that plays a critical role in human intestinal epithelial cells. However, its role and mechanism in colorectal cancer (CRC) remain largely elusive. Here, we investigated the role and clinical significance of BEST4 in CRC. Our results demonstrate that BEST4 expression is upregulated in clinical CRC samples and its high-level expression correlates with advanced TNM (tumor, lymph nodes, distant metastasis) stage, LNM (lymph node metastasis), and poor survival. Functional studies revealed that ectopic expression of BEST4 promoted CRC cell proliferation and metastasis, whereas the depletion of BEST4 had the opposite effect both in vitro and in vivo. Mechanistically, BEST4 binds to the p85α regulatory subunit of phosphatidylinositol-3-kinase (PI3K) and promotes p110 kinase activity; this leads to activation of Akt signaling and expression of MYC and CCND1, which are critical regulators of cell proliferation and metastasis. In clinical samples, the expression of BEST4 is positively associated with the expression of phosphorylated Akt, MYC and CCND1. Pharmacological inhibition of Akt activity markedly repressed BEST4-mediated Akt signaling and proliferation and metastasis of CRC cells. Importantly, the interaction between BEST4 and p85α was also enhanced by epidermal growth factor (EGF) in CRC cells. Therapeutically, BEST4 suppression effectively sensitized CRC cells to gefitinib treatment in vivo. Taken together, our findings indicate the oncogenic potential of BEST4 in colorectal carcinogenesis and metastasis by modulating BEST4/PI3K/Akt signaling, highlighting a potential strategy for CRC therapy.
Collapse
Affiliation(s)
- Xiao-Shun He
- Department of Pathology, Medical College of Soochow University & The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China
| | - Wen-Long Ye
- Department of Pathology, Medical College of Soochow University & The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China
| | - Yu-Juan Zhang
- Department of Pathology, Medical College of Soochow University & The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China
| | - Xiao-Qin Yang
- Department of Bioinformatics, Medical College of Soochow University, Soochow University, Suzhou, 215123, China
| | - Feng Liu
- Department of General Surgery, Canglang Hospital of Suzhou, Suzhou, 215009, China
| | - Jing-Ru Wang
- Department of Pathology, Medical College of Soochow University & The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China
| | - Xiao-Lu Ding
- Department of Pathology, Medical College of Soochow University & The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China
| | - Yun Yang
- Department of Pathology, Medical College of Soochow University & The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China
| | - Ruo-Nan Zhang
- Department of Pathology, Medical College of Soochow University & The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China
| | - Yuan-Yuan Zhao
- Department of Pathology, Medical College of Soochow University & The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China
| | - Hai-Xia Bi
- Department of Pathology, Medical College of Soochow University & The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China
| | - Ling-Chuan Guo
- Department of Pathology, Medical College of Soochow University & The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China.
| | - Wen-Juan Gan
- Department of Pathology, Dushu Lake Hospital Affiliated to Soochow University, Soochow University, Suzhou, 215124, China.
- Department of Pathology, Medical Center of Soochow University, Soochow University, Suzhou, 215124, China.
| | - Hua Wu
- Department of Pathology, Medical College of Soochow University & The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China.
- Department of Pathology, Dushu Lake Hospital Affiliated to Soochow University, Soochow University, Suzhou, 215124, China.
| |
Collapse
|
19
|
James KR, Elmentaite R, Teichmann SA, Hold GL. Redefining intestinal immunity with single-cell transcriptomics. Mucosal Immunol 2022; 15:531-541. [PMID: 34848830 PMCID: PMC8630196 DOI: 10.1038/s41385-021-00470-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/27/2021] [Accepted: 11/03/2021] [Indexed: 02/04/2023]
Abstract
The intestinal immune system represents the largest collection of immune cells in the body and is continually exposed to antigens from food and the microbiota. Here we discuss the contribution of single-cell transcriptomics in shaping our understanding of this complex system. We consider the impact on resolving early intestine development, engagement with the neighbouring microbiota, diversity of intestinal immune cells, compartmentalisation within the intestines and interactions with non-immune cells. Finally, we offer a perspective on open questions about gut immunity that evolving single-cell technologies are well placed to address.
Collapse
Affiliation(s)
- Kylie Renee James
- grid.415306.50000 0000 9983 6924Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, Sydney, NSW 2010 Australia ,grid.1005.40000 0004 4902 0432School of Medical Sciences, University of New South Wales, Sydney, NSW 2006 Australia
| | - Rasa Elmentaite
- grid.10306.340000 0004 0606 5382Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA UK
| | - Sarah Amalia Teichmann
- grid.10306.340000 0004 0606 5382Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA UK ,grid.5335.00000000121885934Theory of Condensed Matter Group, Cavendish Laboratory/Department of Physics, University of Cambridge, Cambridge, NSW CB3 0HE UK
| | - Georgina Louise Hold
- grid.1005.40000 0004 4902 0432University of New South Wales Microbiome Research Centre, Sydney, NSW 2217 Australia
| |
Collapse
|
20
|
Wang L, Zhu H, Sun W, Liang L, Li H, Han C, Huang W, Zhao B, Peng P, Qin M, Shi L, Mo Y, Huang J. Low expression of bestrophin-2 is associated with poor prognosis in colon cancer. Gene 2021; 813:146117. [PMID: 34902511 DOI: 10.1016/j.gene.2021.146117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/10/2021] [Accepted: 12/06/2021] [Indexed: 12/25/2022]
Abstract
OBJECTIVES The purpose of this research was to confirm the prognostic value of bestrophin-2 (BEST2), one of the hub genes in colon cancer, via bioinformatics analysis and validation in public databases and immunohistochemistry detection. METHODS The GEO2R online tool and Venn diagram software were utilized to identify differentially expressed genes (DEGs) from expression profiles, including GSE20916, GSE44861 and GSE74602, from the Gene Expression Omnibus (GEO). The overall survival (OS) and disease-free survival (DFS) of colon cancer patients from The Cancer Genome Atlas (TCGA) were analyzed through Kaplan-Meier survival curves. Verification of the significance of BEST2 in colon cancer was based on TCGA, Genotype Tissue Expression (GTEx) and 10 datasets from GEO. BEST2 expression was detected with immunohistochemistry (IHC) in 330 colon tissue samples on microarrays including 165 colon cancerand 165 adjacent normal tissues. For further validation, comprehensive analysis from tissue microarrays and multiple datasets was performed by the summarizing of receiver operating characteristic (SROC) curves and the standard mean differences (SMDs). BEST2 expression in various kinds of colon cancer tissues and cell lines in the context of pancancer was obtained from the Expression Atlas database. The CBioPortal database was queried to identify BEST2 gene alterations and mutation status in colon cancer. Correlated genes (CEGs) with BEST2 and DEGs from public database data were assembled for functional and pathway enrichment analysis. RESULTS We identified 85 DEGs from the three datasets and screened out BEST2 as a prognostic predictor via the TCGA database. Colon cancer patients with high expression of BEST2 had better survival than patients with low BEST2 (HR = 0.5, P = 0.006) as shown in Kaplan-Meier survival curves in GEPIA. In all, 1463 colon cancer tissues and 1023 colon normal tissues were gathered via public databases as well as in-house tissue microarrays. The comprehensiveexpression analysis suggested low-expression of BEST2 in colon cancer (SMD = -2.48, 95% CI [-3.15- -1.80]) and the notable efficacy of BEST2 expression in differentiating colon cancer from noncancer samples (AUC = 0.97). Gene alteration status of BEST2 occurred in 5% of colon cancer cases, mostly missense mutations and deep deletions. Genes positively correlated with BEST2 and DEGs primarily aggregated in pathways such as anion absorption, digestive juice secretion, cAMP signaling and so on (P < 0.05). CONCLUSION Ampleevidencesupportsthe role of BEST2 in distinguishing colon cancer from normal tissues in this research. Low expression of BEST2 is correlated with a shorter OS, which implies that BEST2 can be employed as a potential biomarker and therapeutictarget in colon cancer.
Collapse
Affiliation(s)
- Li Wang
- Department of Medical Oncology, The Second Affiliated Hospital of Guangxi Medical University, 166 Daxuedong Road, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| | - Huawei Zhu
- Department of Medical Oncology, The Second Affiliated Hospital of Guangxi Medical University, 166 Daxuedong Road, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| | - Weiliang Sun
- Department of Medical Oncology, The Second Affiliated Hospital of Guangxi Medical University, 166 Daxuedong Road, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| | - Li Liang
- Department of Medical Oncology, The Second Affiliated Hospital of Guangxi Medical University, 166 Daxuedong Road, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| | - Hui Li
- Department of Medical Oncology, The Second Affiliated Hospital of Guangxi Medical University, 166 Daxuedong Road, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| | - Chenglong Han
- Department of Medical Oncology, The Second Affiliated Hospital of Guangxi Medical University, 166 Daxuedong Road, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| | - Wenfeng Huang
- Department of Medical Oncology, The Second Affiliated Hospital of Guangxi Medical University, 166 Daxuedong Road, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| | - Bi Zhao
- Department of Medical Oncology, The Second Affiliated Hospital of Guangxi Medical University, 166 Daxuedong Road, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| | - Peng Peng
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, 166 Daxuedong Road, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| | - Mengbin Qin
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, 166 Daxuedong Road, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| | - Ling Shi
- Department of Pathology, The Second Affiliated Hospital of Guangxi Medical University, 166 Daxuedong Road, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| | - Yueqing Mo
- Department of Pathology, The Second Affiliated Hospital of Guangxi Medical University, 166 Daxuedong Road, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| | - Jiean Huang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, 166 Daxuedong Road, Nanning 530000, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
21
|
Owji AP, Kittredge A, Zhang Y, Yang T. Structure and Function of the Bestrophin family of calcium-activated chloride channels. Channels (Austin) 2021; 15:604-623. [PMID: 34612806 PMCID: PMC8496536 DOI: 10.1080/19336950.2021.1981625] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Bestrophins are a family of calcium-activated chloride channels (CaCCs) with relevance to human physiology and a myriad of eye diseases termed "bestrophinopathies". Since the identification of bestrophins as CaCCs nearly two decades ago, extensive studies from electrophysiological and structural biology perspectives have sought to define their key channel features including calcium sensing, gating, inactivation, and anion selectivity. The initial X-ray crystallography studies on the prokaryotic homolog of Best1, Klebsiella pneumoniae (KpBest), and the Best1 homolog from Gallus gallus (chicken Best1, cBest1), laid the foundational groundwork for establishing the architecture of Best1. Recent progress utilizing single-particle cryogenic electron microscopy has further elucidated the molecular mechanism of gating in cBest1 and, separately, the structure of Best2 from Bos taurus (bovine Best2, bBest2). Meanwhile, whole-cell patch clamp, planar lipid bilayer, and other electrophysiologic analyses using these models as well as the human Best1 (hBest1) have provided ample evidence describing the functional properties of the bestrophin channels. This review seeks to consolidate these structural and functional results to paint a broad picture of the underlying mechanisms comprising the bestrophin family's structure-function relationship.
Collapse
Affiliation(s)
- Aaron P Owji
- Department of Pharmacology, Columbia University, NY, USA
| | - Alec Kittredge
- Department of Pharmacology, Columbia University, NY, USA
| | - Yu Zhang
- Department of Ophthalmology, Columbia University, NY, USA
| | - Tingting Yang
- Department of Ophthalmology, Columbia University, NY, USA
| |
Collapse
|
22
|
Rehman T, Karp PH, Tan P, Goodell BJ, Pezzulo AA, Thurman AL, Thornell IM, Durfey SL, Duffey ME, Stoltz DA, McKone EF, Singh PK, Welsh MJ. Inflammatory cytokines TNF-α and IL-17 enhance the efficacy of cystic fibrosis transmembrane conductance regulator modulators. J Clin Invest 2021; 131:e150398. [PMID: 34166230 DOI: 10.1172/jci150398] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 06/22/2021] [Indexed: 12/12/2022] Open
Abstract
Without cystic fibrosis transmembrane conductance regulator-mediated (CFTR-mediated) HCO3- secretion, airway epithelia of newborns with cystic fibrosis (CF) produce an abnormally acidic airway surface liquid (ASL), and the decreased pH impairs respiratory host defenses. However, within a few months of birth, ASL pH increases to match that in non-CF airways. Although the physiological basis for the increase is unknown, this time course matches the development of inflammation in CF airways. To learn whether inflammation alters CF ASL pH, we treated CF epithelia with TNF-α and IL-17 (TNF-α+IL-17), 2 inflammatory cytokines that are elevated in CF airways. TNF-α+IL-17 markedly increased ASL pH by upregulating pendrin, an apical Cl-/HCO3- exchanger. Moreover, when CF epithelia were exposed to TNF-α+IL-17, clinically approved CFTR modulators further alkalinized ASL pH. As predicted by these results, in vivo data revealed a positive correlation between airway inflammation and CFTR modulator-induced improvement in lung function. These findings suggest that inflammation is a key regulator of HCO3- secretion in CF airways. Thus, they explain earlier observations that ASL pH increases after birth and indicate that, for similar levels of inflammation, the pH of CF ASL is abnormally acidic. These results also suggest that a non-cell-autonomous mechanism, airway inflammation, is an important determinant of the response to CFTR modulators.
Collapse
Affiliation(s)
- Tayyab Rehman
- Department of Internal Medicine and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine and
| | - Philip H Karp
- Department of Internal Medicine and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine and.,Howard Hughes Medical Institute, University of Iowa, Iowa City, Iowa, USA
| | - Ping Tan
- Department of Internal Medicine and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine and
| | - Brian J Goodell
- Department of Internal Medicine and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine and
| | - Alejandro A Pezzulo
- Department of Internal Medicine and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine and
| | - Andrew L Thurman
- Department of Internal Medicine and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine and
| | - Ian M Thornell
- Department of Internal Medicine and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine and
| | - Samantha L Durfey
- Departments of Medicine and Microbiology, University of Washington, Seattle, Washington, USA
| | - Michael E Duffey
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, USA
| | - David A Stoltz
- Department of Internal Medicine and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine and.,Department of Molecular Physiology and Biophysics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Edward F McKone
- National Referral Centre for Adult Cystic Fibrosis, St. Vincent's University Hospital and University College Dublin School of Medicine, Dublin, Ireland
| | - Pradeep K Singh
- Departments of Medicine and Microbiology, University of Washington, Seattle, Washington, USA
| | - Michael J Welsh
- Department of Internal Medicine and Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine and.,Howard Hughes Medical Institute, University of Iowa, Iowa City, Iowa, USA.,Department of Molecular Physiology and Biophysics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
23
|
Madka V, Kumar G, Pathuri G, Panneerselvam J, Zhang Y, Ganta V, Lightfoot S, Lubet RA, Suen CS, Steele VE, Janakiram NB, Mohammed A, Rao CV. Proton pump inhibitor omeprazole suppresses carcinogen induced colonic adenoma progression to adenocarcinoma in F344 rat. Cancer Prev Res (Phila) 2021; 14:1009-1020. [PMID: 34341012 DOI: 10.1158/1940-6207.capr-21-0057] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 05/25/2021] [Accepted: 07/09/2021] [Indexed: 11/16/2022]
Abstract
Colorectal cancer (CRC) causes over 53,000 deaths annually in the United States. Its rising incidences worldwide and particularly in young adults is a major concern. Here, we evaluated the efficacy of omeprazole (OME) that is clinically approved for treating acid-reflux, to enable its repurposing for CRC prevention. In the azoxymethane (AOM)-induced rat CRC model, dietary OME (250 and 500 ppm) was administered at early adenoma stage (8 weeks after AOM) to assess the progression of early lesions to adenocarcinoma. Administration of OME at 250 ppm or 500 ppm doses led to suppression of total colon adenocarcinoma incidence by 15.7% and 32% (p<0.01), respectively. Importantly, invasive carcinoma incidence was reduced by 59% (p<0.0005) and 90% (p<0.0001) in OME administered rats in a dose-dependent manner. There was also a strong and dose-dependent inhibition in the adenocarcinoma multiplicity in rats exposed to OME. Administration of 250 and 500 ppm OME inhibited total colon adenocarcinoma multiplicity by ~49% and ~65% (p<0.0001), respectively. While non-invasive adenocarcinomas multiplicity was suppressed by ~34% to ~48% (p<0.02), the invasive carcinomas multiplicity was reduced by ~74% to ~94% (p<0.0001) in OME exposed rats in comparison to the untreated rats. Biomarker analysis results showed a decrease in cell proliferation and anti-apoptotic/pro-survival proteins with an increase in apoptosis. Transcriptome analysis of treated tumors revealed a significant increase in adenocarcinoma inhibitory genes (Olmf4; Spink4) expression and down regulation of progression promoting genes (SerpinA1, MMP21, IL6). In summary, OME showed significant protection against the progression of adenoma to adenocarcinoma.
Collapse
Affiliation(s)
- Venkateshwar Madka
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology/Oncology Section, University of Oklahoma Health Sciences Center
| | - Gaurav Kumar
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology/Oncology Section, University of Oklahoma Health Sciences Center
| | - Gopal Pathuri
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology/Oncology Section, University of Oklahoma Health Sciences Center
| | - Janani Panneerselvam
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology/Oncology Section, University of Oklahoma Health Sciences Center
| | - Yuting Zhang
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology/Oncology Section, University of Oklahoma Health Sciences Center
| | - Vishal Ganta
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology/Oncology Section, University of Oklahoma Health Sciences Center
| | - Stanley Lightfoot
- Pathology-Retired, Center for Cancer Prevention and Drug Development
| | - Ronald A Lubet
- Division of Cancer Prevention, National Cancer Institute
| | - Chen S Suen
- Cancer Prevention, National Cancer Institute
| | | | | | - Altaf Mohammed
- Division of Cancer Prevention, National Cancer Institute
| | - Chinthalapally V Rao
- Center for Cancer Prevention and Drug Development, Department of Medicine, Hematology/Oncology Section, University of Oklahoma Health Sciences Center
| |
Collapse
|
24
|
Goblet cell LRRC26 regulates BK channel activation and protects against colitis in mice. Proc Natl Acad Sci U S A 2021; 118:2019149118. [PMID: 33431687 DOI: 10.1073/pnas.2019149118] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Goblet cells (GCs) are specialized cells of the intestinal epithelium contributing critically to mucosal homeostasis. One of the functions of GCs is to produce and secrete MUC2, the mucin that forms the scaffold of the intestinal mucus layer coating the epithelium and separates the luminal pathogens and commensal microbiota from the host tissues. Although a variety of ion channels and transporters are thought to impact on MUC2 secretion, the specific cellular mechanisms that regulate GC function remain incompletely understood. Previously, we demonstrated that leucine-rich repeat-containing protein 26 (LRRC26), a known regulatory subunit of the Ca2+-and voltage-activated K+ channel (BK channel), localizes specifically to secretory cells within the intestinal tract. Here, utilizing a mouse model in which MUC2 is fluorescently tagged, thereby allowing visualization of single GCs in intact colonic crypts, we show that murine colonic GCs have functional LRRC26-associated BK channels. In the absence of LRRC26, BK channels are present in GCs, but are not activated at physiological conditions. In contrast, all tested MUC2- cells completely lacked BK channels. Moreover, LRRC26-associated BK channels underlie the BK channel contribution to the resting transepithelial current across mouse distal colonic mucosa. Genetic ablation of either LRRC26 or BK pore-forming α-subunit in mice results in a dramatically enhanced susceptibility to colitis induced by dextran sodium sulfate. These results demonstrate that normal potassium flux through LRRC26-associated BK channels in GCs has protective effects against colitis in mice.
Collapse
|
25
|
|
26
|
Rehman S, Narayanan K, Nickerson AJ, Coon SD, Hoque KM, Sandle GI, Rajendran VM. Parallel intermediate conductance K + and Cl - channel activity mediates electroneutral K + exit across basolateral membranes in rat distal colon. Am J Physiol Gastrointest Liver Physiol 2020; 319:G142-G150. [PMID: 32567323 PMCID: PMC7500264 DOI: 10.1152/ajpgi.00011.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Transepithelial K+ absorption requires apical K+ uptake and basolateral K+ exit. In the colon, apical H+-K+-ATPase mediates cellular K+ uptake, and it has been suggested that electroneutral basolateral K+ exit reflects K+-Cl- cotransporter-1 (KCC1) operating in parallel with K+ and Cl- channels. The present study was designed to identify basolateral transporter(s) responsible for K+ exit in rat distal colon. Active K+ absorption was determined by measuring 86Rb+ (K+ surrogate) fluxes across colonic epithelia under voltage-clamp conditions. With zero Cl- in the mucosal solution, net K+ absorption was reduced by 38%, indicating that K+ absorption was partially Cl--dependent. Serosal addition of DIOA (KCC1 inhibitor) or Ba2+ (nonspecific K+ channel blocker) inhibited net K+ absorption by 21% or 61%, respectively, suggesting that both KCC1 and K+ channels contribute to basolateral K+ exit. Clotrimazole and TRAM34 (IK channel blockers) added serosally inhibited net K+ absorption, pointing to the involvement of IK channels in basolateral K+ exit. GaTx2 (CLC2 blocker) added serosally also inhibited net K+ absorption, suggesting that CLC2-mediated Cl- exit accompanies IK channel-mediated K+ exit across the basolateral membrane. Net K+ absorption was not inhibited by serosal addition of either IbTX (BK channel blocker), apamin (SK channel blocker), chromanol 293B (KV7 channel blocker), or CFTRinh172 (CFTR blocker). Immunofluorescence studies confirmed basolateral membrane colocalization of CLC2-like proteins and Na+-K+-ATPase α-subunits. We conclude that active K+ absorption in rat distal colon involves electroneutral basolateral K+ exit, which may reflect IK and CLC2 channels operating in parallel.NEW & NOTEWORTHY This study demonstrates that during active electroneutral K+ absorption in rat distal colon, K+ exit across the basolateral membrane mainly reflects intermediate conductance K+ channels operating in conjunction with chloride channel 2, with a smaller, but significant, contribution from K+-Cl- cotransporter-1 (KCC1) activity.
Collapse
Affiliation(s)
- Shabina Rehman
- 1Departments of Biochemistry West Virginia University School of Medicine, Morgantown, West Virginia
| | - Karthikeyan Narayanan
- 1Departments of Biochemistry West Virginia University School of Medicine, Morgantown, West Virginia
| | - Andrew J. Nickerson
- 1Departments of Biochemistry West Virginia University School of Medicine, Morgantown, West Virginia,2Departments of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Steven D. Coon
- 3Department of Biological Sciences, Port Peck Community College, Poplar, Montana
| | - Kazi Mirajul Hoque
- 4Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Geoffrey I. Sandle
- 5Leeds Institute for Medical Research at St. James’s, St. James’s University Hospital. Leeds, United Kingdom
| | - Vazhaikkurichi M. Rajendran
- 1Departments of Biochemistry West Virginia University School of Medicine, Morgantown, West Virginia,6Departments of Medicine, West Virginia University School of Medicine, Morgantown, West Virginia
| |
Collapse
|
27
|
Shin DH, Kim M, Kim Y, Jun I, Jung J, Nam JH, Cheng MH, Lee MG. Bicarbonate permeation through anion channels: its role in health and disease. Pflugers Arch 2020; 472:1003-1018. [PMID: 32621085 DOI: 10.1007/s00424-020-02425-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/19/2020] [Accepted: 06/26/2020] [Indexed: 12/31/2022]
Abstract
Many anion channels, frequently referred as Cl- channels, are permeable to different anions in addition to Cl-. As the second-most abundant anion in the human body, HCO3- permeation via anion channels has many important physiological roles. In addition to its classical role as an intracellular pH regulator, HCO3- also controls the activity and stability of dissolved proteins in bodily fluids such as saliva, pancreatic juice, intestinal fluid, and airway surface liquid. Moreover, HCO3- permeation through these channels affects membrane potentials that are the driving forces for transmembrane transport of solutes and water in epithelia and affect neuronal excitability in nervous tissue. Consequently, aberrant HCO3- transport via anion channels causes a number of human diseases in respiratory, gastrointestinal, genitourinary, and neuronal systems. Notably, recent studies have shown that the HCO3- permeabilities of several anion channels are not fixed and can be altered by cellular stimuli, findings which may have both physiological and pathophysiological significance. In this review, we summarize recent progress in understanding the molecular mechanisms and the physiological roles of HCO3- permeation through anion channels. We hope that the present discussions can stimulate further research into this very important topic, which will provide the basis for human disorders associated with aberrant HCO3- transport.
Collapse
Affiliation(s)
- Dong Hoon Shin
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Minjae Kim
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Yonjung Kim
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Ikhyun Jun
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, South Korea
- The Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Jinsei Jung
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, South Korea
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Joo Hyun Nam
- Department of Physiology, Dongguk University College of Medicine, 123 Dongdae-ro, Kyungju, 780-714, Republic of Korea
| | - Mary Hongying Cheng
- Department of Computational & Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Min Goo Lee
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| |
Collapse
|
28
|
Qin L, Zeng J, Shi N, Chen L, Wang L. Application of weighted gene co‑expression network analysis to explore the potential diagnostic biomarkers for colorectal cancer. Mol Med Rep 2020; 21:2533-2543. [PMID: 32323816 PMCID: PMC7185270 DOI: 10.3892/mmr.2020.11047] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Accepted: 03/02/2020] [Indexed: 12/13/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignant diseases in the world. Although mechanistic studies have been conducted on the pathogenesis of CRC, the molecular mechanism of CRC tumorigenesis remains unclear. In the present study, the weighted gene co-expression network analysis was performed for the Gene Expression Omnibus (GEO) dataset GSE87211, in order to analyze the key modules involved in the pathogenesis of CRC. Next, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses were performed on the key module genes to analyze the functional pathways involved. The hub genes were screened using the Cytoscape platform and verified by a second GEO dataset, GSE21510. Finally, 10 hub genes were identified in 2 key modules (the green and brown modules) as the genes most significantly associated with the tumorigenesis of CRC. The 5 hub genes from the green module included collagen type I α1 chain, collagen type XII α1 chain, collagen triple helix repeat containing 1, inhibin subunit βa (INHBA) and chromobox 2 (CBX2), while the 5 hub genes from the brown module included bestrophin 2 (BEST2), carbonic anhydrase 2, glucagon, solute carrier family 4 member 4 and gliomedin. The 2 key modules with the 10 hub genes identified may regulate the occurrence and development of CRC through the extracellular matrix pathway, PI3K-Akt and chemokine signaling pathways, thus providing a reference for understanding the complex mechanism of tumorigenesis in CRC. Of note, few studies have reported the pathogenesis of CRC with the 3 identified hub genes, INHBA, CBX2 and BEST2. Further investigation of the molecular mechanism of these genes in CRC is recommended.
Collapse
Affiliation(s)
- Liping Qin
- Molecular Laboratory, Institute of Aging Research, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| | - Jianping Zeng
- Department of Neurosurgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 311121, P.R. China
| | - Nannan Shi
- Molecular Laboratory, Institute of Aging Research, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| | - Liu Chen
- Molecular Laboratory, Institute of Aging Research, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| | - Li Wang
- Molecular Laboratory, Institute of Aging Research, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| |
Collapse
|
29
|
Owji AP, Zhao Q, Ji C, Kittredge A, Hopiavuori A, Fu Z, Ward N, Clarke OB, Shen Y, Zhang Y, Hendrickson WA, Yang T. Structural and functional characterization of the bestrophin-2 anion channel. Nat Struct Mol Biol 2020; 27:382-391. [PMID: 32251414 PMCID: PMC7150642 DOI: 10.1038/s41594-020-0402-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 02/28/2020] [Indexed: 01/21/2023]
Abstract
The bestrophin family of calcium (Ca2+)-activated chloride (Cl-) channels, which mediate the influx and efflux of monovalent anions in response to the levels of intracellular Ca2+, comprises four members in mammals (bestrophin 1-4). Here we report cryo-EM structures of bovine bestrophin-2 (bBest2) bound and unbound by Ca2+ at 2.4- and 2.2-Å resolution, respectively. The bBest2 structure highlights four previously underappreciated pore-lining residues specifically conserved in Best2 but not in Best1, illustrating the differences between these paralogs. Structure-inspired electrophysiological analysis reveals that, although the channel is sensitive to Ca2+, it has substantial Ca2+-independent activity for Cl-, reflecting the opening at the cytoplasmic restriction of the ion conducting pathway even when Ca2+ is absent. Moreover, the ion selectivity of bBest2 is controlled by multiple residues, including those involved in gating.
Collapse
Affiliation(s)
- Aaron P Owji
- Department of Pharmacology, Columbia University, New York, NY, USA
| | - Qingqing Zhao
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, NY, USA
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Changyi Ji
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, NY, USA
| | - Alec Kittredge
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, NY, USA
| | - Austin Hopiavuori
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, NY, USA
| | - Ziao Fu
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Nancy Ward
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, NY, USA
| | - Oliver B Clarke
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Yin Shen
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Yu Zhang
- Department of Ophthalmology, Columbia University, New York, NY, USA.
| | - Wayne A Hendrickson
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA.
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA.
- New York Structural Biology Center, New York, NY, USA.
| | - Tingting Yang
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, NY, USA.
- Department of Ophthalmology, Columbia University, New York, NY, USA.
| |
Collapse
|
30
|
Dayama G, Priya S, Niccum DE, Khoruts A, Blekhman R. Interactions between the gut microbiome and host gene regulation in cystic fibrosis. Genome Med 2020; 12:12. [PMID: 31992345 PMCID: PMC6988342 DOI: 10.1186/s13073-020-0710-2] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 01/05/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Cystic fibrosis is the most common autosomal recessive genetic disease in Caucasians. It is caused by mutations in the CFTR gene, leading to poor hydration of mucus and impairment of the respiratory, digestive, and reproductive organ functions. Advancements in medical care have led to markedly increased longevity of patients with cystic fibrosis, but new complications have emerged, such as early onset of colorectal cancer. Although the pathogenesis of colorectal cancer in cystic fibrosis remains unclear, altered host-microbe interactions might play a critical role. To investigate this, we characterized changes in the microbiome and host gene expression in the colonic mucosa of cystic fibrosis patients relative to healthy controls, and identified host gene-microbiome interactions in the colon of cystic fibrosis patients. METHODS We performed RNA-seq on colonic mucosa samples from cystic fibrosis patients and healthy controls to determine differentially expressed host genes. We also performed 16S rRNA sequencing to characterize the colonic mucosal microbiome and identify gut microbes that are differentially abundant between patients and healthy controls. Lastly, we modeled associations between relative abundances of specific bacterial taxa in the gut mucosa and host gene expression. RESULTS We find that 1543 genes, including CFTR, show differential expression in the colon of cystic fibrosis patients compared to healthy controls. These genes are enriched with functions related to gastrointestinal and colorectal cancer, such as metastasis of colorectal cancer, tumor suppression, p53, and mTOR signaling pathways. In addition, patients with cystic fibrosis show decreased gut microbial diversity, decreased abundance of butyrate producing bacteria, such as Ruminococcaceae and Butyricimonas, and increased abundance of other taxa, such as Actinobacteria and Clostridium. An integrative analysis identified colorectal cancer-related genes, including LCN2 and DUOX2, for which gene expression is correlated with the abundance of colorectal cancer-associated bacteria, such as Ruminococcaceae and Veillonella. CONCLUSIONS In addition to characterizing host gene expression and mucosal microbiome in cystic fibrosis patients, our study explored the potential role of host-microbe interactions in the etiology of colorectal cancer in cystic fibrosis. Our results provide biomarkers that may potentially serve as targets for stratifying risk of colorectal cancer in patients with cystic fibrosis.
Collapse
Affiliation(s)
- Gargi Dayama
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, USA
| | - Sambhawa Priya
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, USA
| | - David E Niccum
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Alexander Khoruts
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA.
- Center for Immunology, BioTechnology Institute, University of Minnesota, Minneapolis, MN, USA.
| | - Ran Blekhman
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, USA.
- Department of Ecology, Evolution, and Behavior, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
31
|
Yoo JH, Donowitz M. Intestinal enteroids/organoids: A novel platform for drug discovery in inflammatory bowel diseases. World J Gastroenterol 2019; 25:4125-4147. [PMID: 31435168 PMCID: PMC6700704 DOI: 10.3748/wjg.v25.i30.4125] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 06/14/2019] [Accepted: 07/19/2019] [Indexed: 02/06/2023] Open
Abstract
The introduction of biologics such as anti-tumor necrosis factor (TNF) monoclonal antibodies followed by anti-integrins has dramatically changed the therapeutic paradigm of inflammatory bowel diseases (IBD). Furthermore, a newly developed anti-p40 subunit of interleukin (IL)-12 and IL-23 (ustekinumab) has been recently approved in the United States for patients with moderate to severe Crohn’s disease who have failed treatment with anti-TNFs. However, these immunosuppressive therapeutics which focus on anti-inflammatory mechanisms or immune cells still fail to achieve long-term remission in a significant percentage of patients. This strongly underlines the need to identify novel treatment targets beyond immune suppression to treat IBD. Recent studies have revealed the critical role of intestinal epithelial cells (IECs) in the pathogenesis of IBD. Physical, biochemical and immunologic driven barrier dysfunctions of epithelial cells contribute to the development of IBD. In addition, the recent establishment of adult stem cell-derived intestinal enteroid/organoid culture technology has allowed an exciting opportunity to study human IECs comprising all normal epithelial cells. This long-term epithelial culture model can be generated from endoscopic biopsies or surgical resections and recapitulates the tissue of origin, representing a promising platform for novel drug discovery in IBD. This review describes the advantages of intestinal enteroids/organoids as a research tool for intestinal diseases, introduces studies with these models in IBD, and gives a description of the current status of therapeutic approaches in IBD. Finally, we provide an overview of the current endeavors to identify a novel drug target for IBD therapy based on studies with human enteroids/organoids and describe the challenges in using enteroids/organoids as an IBD model.
Collapse
Affiliation(s)
- Jun-Hwan Yoo
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
- Digestive Disease Center, CHA Bundang Medical Center, CHA University, Seongnam 13496, South Korea
| | - Mark Donowitz
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| |
Collapse
|
32
|
Lee B, Hong GS, Lee SH, Kim H, Kim A, Hwang EM, Kim J, Lee MG, Yang JY, Kweon MN, Tse CM, Mark D, Oh U. Anoctamin 1/TMEM16A controls intestinal Cl - secretion induced by carbachol and cholera toxin. Exp Mol Med 2019; 51:1-14. [PMID: 31383845 PMCID: PMC6802608 DOI: 10.1038/s12276-019-0287-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 03/07/2019] [Accepted: 03/11/2019] [Indexed: 12/17/2022] Open
Abstract
Calcium-activated chloride channels (CaCCs) mediate numerous physiological functions and are best known for the transport of electrolytes and water in epithelia. In the intestine, CaCC currents are considered necessary for the secretion of fluid to protect the intestinal epithelium. Although genetic ablation of ANO1/TMEM16A, a gene encoding a CaCC, reduces the carbachol-induced secretion of intestinal fluid, its mechanism of action is still unknown. Here, we confirm that ANO1 is essential for the secretion of intestinal fluid. Carbachol-induced transepithelial currents were reduced in the proximal colon of Ano1-deficient mice. Surprisingly, cholera toxin-induced and cAMP-induced fluid secretion, believed to be mediated by CFTR, were also significantly reduced in the intestine of Ano1-deficient mice. ANO1 is largely expressed in the apical membranes of intestines, as predicted for CaCCs. The Ano1-deficient colons became edematous under basal conditions and had a greater susceptibility to dextran sodium sulfate-induced colitis. However, Ano1 depletion failed to affect tumor development in a model of colorectal cancer. We thus conclude that ANO1 is necessary for cAMP- and carbachol-induced Cl− secretion in the intestine, which is essential for the protection of the intestinal epithelium from colitis. An ion channel, a membrane protein allowing ion transport, that controls the flow of chloride is needed for proper secretion of protective fluids in the intestine. Uhtaek Oh from the Korea Institute of Science & Technology in Seoul, South Korea, and colleagues showed that cells lining the intestinal surface express a calcium-activated chloride channel called anoctamin-1 (ANO1) that regulates fluid secretion in the gut. Compared to control animals, ANO1-deficient mice released less fluid into their intestines following exposure to a diarrhea-inducing toxin or to a chloride transport–stimulating signaling molecule. This fluid secretion was previously thought to be mediated via a different ion channel. The ANO1-deficient mice accumulated fluid within colonic tissues, which increased their susceptibility to colitis. The findings point to ANO1 activation as a potential therapeutic strategy for treating colitis.
Collapse
Affiliation(s)
- Byeongjun Lee
- College of Pharmacy, Seoul National University, Seoul, 08826, Korea
| | - Gyu-Sang Hong
- Brain Science Institute, Korea Institute of Science & Technology (KIST), Seoul, 02792, Korea
| | - Sung Hoon Lee
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Korea
| | - Hyungsup Kim
- Brain Science Institute, Korea Institute of Science & Technology (KIST), Seoul, 02792, Korea
| | - Ajung Kim
- Brain Science Institute, Korea Institute of Science & Technology (KIST), Seoul, 02792, Korea
| | - Eun Mi Hwang
- Brain Science Institute, Korea Institute of Science & Technology (KIST), Seoul, 02792, Korea
| | - Jiyoon Kim
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Min Goo Lee
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, 03722, Korea
| | - Jin-Young Yang
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine/Asan Medical Center, Seoul, 05505, Korea
| | - Mi-Na Kweon
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine/Asan Medical Center, Seoul, 05505, Korea
| | - Chung-Ming Tse
- Departments of Physiology and Medicine, Division of Gastroenterois maintained by the opening of plasmalogy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Donowitz Mark
- Departments of Physiology and Medicine, Division of Gastroenterois maintained by the opening of plasmalogy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Uhtaek Oh
- College of Pharmacy, Seoul National University, Seoul, 08826, Korea. .,Brain Science Institute, Korea Institute of Science & Technology (KIST), Seoul, 02792, Korea.
| |
Collapse
|
33
|
Vega G, Guequén A, Johansson MEV, Arike L, Martínez-Abad B, Nyström EEL, Scudieri P, Pedemonte N, Millar-Büchner P, Philp AR, Galietta LJ, Hansson GC, Flores CA. Normal Calcium-Activated Anion Secretion in a Mouse Selectively Lacking TMEM16A in Intestinal Epithelium. Front Physiol 2019; 10:694. [PMID: 31263421 PMCID: PMC6585864 DOI: 10.3389/fphys.2019.00694] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/16/2019] [Indexed: 01/24/2023] Open
Abstract
Calcium-activated anion secretion is expected to ameliorate cystic fibrosis, a genetic disease that carries an anion secretory defect in exocrine tissues. Human patients and animal models of the disease that present a mild intestinal phenotype have been postulated to bear a compensatory calcium-activated anion secretion in the intestine. TMEM16A is calcium-activated anion channel whose presence in the intestinal epithelium is contradictory. We aim to test the functional expression of TMEM16A using animal models with Cftr and/or Tmem16a intestinal silencing. Expression of TMEM16A was studied in a wild type and intestinal Tmem16a knockout mice by mRNA-seq, mass-spectrometry, q-PCR, Western blotting and immunolocalization. Calcium-activated anion secretion was recorded in the ileum and proximal colon of these animals including intestinal Cftr knockout and double mutants with dual Tmem16a and Cftr intestinal ablation. Mucus homeostasis was studied by immune-analysis of Mucin-2 (Muc2) and survival curves were recorded. Tmem16a transcript was found in intestine. Nevertheless, protein was barely detected in colon samples. Electrophysiological measurements demonstrated that the intestinal deletion of Tmem16a did not change calcium-activated anion secretion induced by carbachol or ATP in ileum and proximal colon. Muc2 architecture was not altered by Tmem16a silencing as was observed when Cftr was deleted from mouse intestine. Tmem16a silencing neither affected animal survival nor modified the lethality observed in the intestinal Cftr-null mouse. Our results demonstrate that TMEM16A function in the murine intestine is not related to electrogenic calcium-activated anion transport and does not affect mucus homeostasis and survival of animals.
Collapse
Affiliation(s)
- Génesis Vega
- Centro de Estudios Científicos (CECs), Valdivia, Chile
- Universidad Austral de Chile, Valdivia, Chile
| | - Anita Guequén
- Centro de Estudios Científicos (CECs), Valdivia, Chile
| | | | - Liisa Arike
- Department of Medical Biochemistry, University of Gothenburg, Gothenburg, Sweden
| | | | | | - Paolo Scudieri
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | | | | | - Amber R. Philp
- Centro de Estudios Científicos (CECs), Valdivia, Chile
- Universidad Austral de Chile, Valdivia, Chile
| | - Luis J. Galietta
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
- Department of Translational Medical Sciences (DISMET), University of Naples Federico II, Naples, Italy
| | - Gunnar C. Hansson
- Department of Medical Biochemistry, University of Gothenburg, Gothenburg, Sweden
| | | |
Collapse
|
34
|
Rao MC. Physiology of Electrolyte Transport in the Gut: Implications for Disease. Compr Physiol 2019; 9:947-1023. [PMID: 31187895 DOI: 10.1002/cphy.c180011] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We now have an increased understanding of the genetics, cell biology, and physiology of electrolyte transport processes in the mammalian intestine, due to the availability of sophisticated methodologies ranging from genome wide association studies to CRISPR-CAS technology, stem cell-derived organoids, 3D microscopy, electron cryomicroscopy, single cell RNA sequencing, transgenic methodologies, and tools to manipulate cellular processes at a molecular level. This knowledge has simultaneously underscored the complexity of biological systems and the interdependence of multiple regulatory systems. In addition to the plethora of mammalian neurohumoral factors and their cross talk, advances in pyrosequencing and metagenomic analyses have highlighted the relevance of the microbiome to intestinal regulation. This article provides an overview of our current understanding of electrolyte transport processes in the small and large intestine, their regulation in health and how dysregulation at multiple levels can result in disease. Intestinal electrolyte transport is a balance of ion secretory and ion absorptive processes, all exquisitely dependent on the basolateral Na+ /K+ ATPase; when this balance goes awry, it can result in diarrhea or in constipation. The key transporters involved in secretion are the apical membrane Cl- channels and the basolateral Na+ -K+ -2Cl- cotransporter, NKCC1 and K+ channels. Absorption chiefly involves apical membrane Na+ /H+ exchangers and Cl- /HCO3 - exchangers in the small intestine and proximal colon and Na+ channels in the distal colon. Key examples of our current understanding of infectious, inflammatory, and genetic diarrheal diseases and of constipation are provided. © 2019 American Physiological Society. Compr Physiol 9:947-1023, 2019.
Collapse
Affiliation(s)
- Mrinalini C Rao
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
35
|
Enns CB, Harding JCS, Loewen ME. Decreased electrogenic anionic secretory response in the porcine colon following in vivo challenge with Brachyspira spp. supports an altered mucin environment. Am J Physiol Gastrointest Liver Physiol 2019; 316:G495-G508. [PMID: 30629469 DOI: 10.1152/ajpgi.00348.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Brachyspira spp. cause diarrheal disease in multiple animal species by colonization of the colon, resulting in colitis, mucus induction, and disrupted ion transport. Unique to spirochete pathogenesis is the immense production of mucus, resulting in a niche mucin environment likely favoring spirochete colonization. Mucin rheological properties are heavily influenced by anionic secretion, and loss of secretory function has been implicated in diseases such as cystic fibrosis. Here, the effects on the agonist-induced electrogenic anionic secretory response by infectious colonic spirochete bacteria Brachyspira hyodysenteriae and Brachyspira hampsonii were assessed in the proximal, apex, and distal sections of colon in Ussing chambers. Activation of secretion via isoproterenol, carbachol, and forskolin/3-isobutyl-1-methylxanthine demonstrated a significantly decreased change in short-circuit current ( Isc) in Brachyspira-infected pigs in all sections. Tissue resistances did not account for this difference, rather, it was attributed to a decrease in anionic secretion as indicated by a decrease in bumetanide inhibitable Isc. Quantitative RT-PCR and Western blot analyses determined that the major anionic channels of the epithelium were downregulated in diarrheic pigs paired with altered mucin gene expression. The investigated cytokines were not responsible for the downregulation of anion channel gene transcripts. Although IL-1α was upregulated in all segments, it did not alter cystic fibrosis transmembrane conductance regulator (CFTR) mRNA expression in Caco-2 monolayers. However, a whole cell Brachyspira hampsonii lysate significantly reduced CFTR mRNA expression in Caco-2 monolayers. Together, these findings indicate that these two Brachyspira spp. may directly cause a decreased anionic secretory response in the porcine colon, supporting an altered mucin environment likely favoring spirochete colonization. NEW & NOTEWORTHY This research demonstrates for the first time that the niche mucin environment produced by two infectious spirochete spp. is supported by a decrease in the electrogenic anionic secretory response throughout the porcine colon. Our findings suggest that the host's cytokine response is not likely responsible for the decrease in anionic secretory function. Rather, it appears that Brachyspira spp. directly impede ion channel transcription and translation, potentially altering colonic mucin rheological properties, which may favor spirochete colonization.
Collapse
Affiliation(s)
- Cole B Enns
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan , Saskatoon , Canada
| | - John C S Harding
- Department of Large Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan , Saskatoon , Canada
| | - Matthew E Loewen
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan , Saskatoon , Canada
| |
Collapse
|
36
|
Miller AN, Vaisey G, Long SB. Molecular mechanisms of gating in the calcium-activated chloride channel bestrophin. eLife 2019; 8:43231. [PMID: 30628889 PMCID: PMC6342527 DOI: 10.7554/elife.43231] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 01/02/2019] [Indexed: 11/13/2022] Open
Abstract
Bestrophin (BEST1-4) ligand-gated chloride (Cl-) channels are activated by calcium (Ca2+). Mutation of BEST1 causes retinal disease. Partly because bestrophin channels have no sequence or structural similarity to other ion channels, the molecular mechanisms underlying gating are unknown. Here, we present a series of cryo-electron microscopy structures of chicken BEST1, determined at 3.1 Å resolution or better, that represent the channel’s principal gating states. Unlike other channels, opening of the pore is due to the repositioning of tethered pore-lining helices within a surrounding protein shell that dramatically widens a neck of the pore through a concertina of amino acid rearrangements. The neck serves as both the activation and the inactivation gate. Ca2+ binding instigates opening of the neck through allosteric means whereas inactivation peptide binding induces closing. An aperture within the otherwise wide pore controls anion permeability. The studies define a new molecular paradigm for gating among ligand-gated ion channels.
Collapse
Affiliation(s)
- Alexandria N Miller
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, United States
| | - George Vaisey
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, United States.,Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, United States
| | - Stephen B Long
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, United States
| |
Collapse
|
37
|
Rottgen TS, Nickerson AJ, Minor EA, Stewart AB, Harold AD, Rajendran VM. Dextran sulfate sodium-induced chronic colitis attenuates Ca 2+-activated Cl - secretion in murine colon by downregulating TMEM16A. Am J Physiol Cell Physiol 2018; 315:C10-C20. [PMID: 29561662 PMCID: PMC6087728 DOI: 10.1152/ajpcell.00328.2017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 03/13/2018] [Accepted: 03/13/2018] [Indexed: 12/29/2022]
Abstract
Attenuated Ca2+-activated Cl- secretion has previously been observed in the model of dextran sulfate sodium (DSS)-induced colitis. Prior studies have implicated dysfunctional muscarinic signaling from basolateral membranes as the potential perpetrator leading to decreased Ca2+-activated Cl- secretion. However, in our chronic model of DSS-colitis, cholinergic receptor muscarinic 3 ( Chrm3) transcript (1.028 ± 0.12 vs. 1.029 ± 0.27, P > 0.05) and CHRM3 protein expression (1.021 ± 0.24 vs. 0.928 ± 0.09, P > 0.05) were unchanged. Therefore, we hypothesized that decreased carbachol (CCH)-stimulated Cl- secretion in DSS-induced colitis could be attributed to a loss of Ca2+-activated Cl- channels (CaCC) in apical membranes of colonic epithelium. To establish this chemically-induced colitis, Balb/C mice were exposed to 4% DSS for five alternating weeks to stimulate a more moderate, chronic colitis. Upon completion of the protocol, whole thickness sections of colon were mounted in an Ussing chamber under voltage-clamp conditions. DSS-induced colitis demonstrated a complete inhibition of basolateral administration of CCH-stimulated Cl- secretion that actually displayed a reversal in polarity (15.40 ± 2.22 μA/cm2 vs. -2.47 ± 0.25 μA/cm2). Western blotting of potential CaCCs, quantified by densitometric analysis, demonstrated no change in bestrophin-2 and cystic fibrosis transmembrane regulator, whereas anoctamin-1 [ANO1, transmembrane protein 16A (TMEM16A)] was significantly downregulated (1.001 ± 0.13 vs. 0.510 ± 0.12, P < 0.05). Our findings indicate that decreased expression of TMEM16A in DSS-induced colitis contributes to the decreased Ca2+-activated Cl- secretion in murine colon.
Collapse
Affiliation(s)
- Trey S Rottgen
- Department of Physiology, Pharmacology, and Neuroscience, West Virginia University School of Medicine , Morgantown, West Virginia
- Department of Biochemistry and Molecular Pharmacology, West Virginia University School of Medicine , Morgantown, West Virginia
| | - Andrew J Nickerson
- Department of Physiology, Pharmacology, and Neuroscience, West Virginia University School of Medicine , Morgantown, West Virginia
- Department of Biochemistry and Molecular Pharmacology, West Virginia University School of Medicine , Morgantown, West Virginia
| | - Emily A Minor
- Department of Physiology, Pharmacology, and Neuroscience, West Virginia University School of Medicine , Morgantown, West Virginia
- Department of Biochemistry and Molecular Pharmacology, West Virginia University School of Medicine , Morgantown, West Virginia
| | | | - Abby D Harold
- Department of Physiology, Pharmacology, and Neuroscience, West Virginia University School of Medicine , Morgantown, West Virginia
- Department of Biochemistry and Molecular Pharmacology, West Virginia University School of Medicine , Morgantown, West Virginia
| | - Vazhaikkurichi M Rajendran
- Department of Physiology, Pharmacology, and Neuroscience, West Virginia University School of Medicine , Morgantown, West Virginia
- Department of Biochemistry and Molecular Pharmacology, West Virginia University School of Medicine , Morgantown, West Virginia
| |
Collapse
|
38
|
Massip-Copiz MM, Santa-Coloma TA. Extracellular pH and lung infections in cystic fibrosis. Eur J Cell Biol 2018; 97:402-410. [PMID: 29933921 DOI: 10.1016/j.ejcb.2018.06.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 06/01/2018] [Accepted: 06/12/2018] [Indexed: 12/11/2022] Open
Abstract
Cystic fibrosis (CF) is an autosomal recessive disease caused by CFTR mutations. It is characterized by high NaCl concentration in sweat and the production of a thick and sticky mucus, occluding secretory ducts, intestine and airways, accompanied by chronic inflammation and infections of the lungs. This causes a progressive and lethal decline in lung function. Therefore, finding the mechanisms driving the high susceptibility to lung infections has been a key issue. For decades the prevalent hypothesis was that a reduced airway surface liquid (ASL) volume and composition, and the consequent increased mucus concentration (dehydration), create an environment favoring infections. However, a few years ago, in a pig model of CF, the Na+/K+ concentrations and the ASL volume were found intact. Immediately a different hypothesis arose, postulating a reduced ASL pH as the cause for the increased susceptibility to infections, due to a diminished bicarbonate secretion through CFTR. Noteworthy, a recent report found normal ASL pH values in CF children and in cultured primary airway cells, challenging the ASL pH hypothesis. On the other hand, recent evidences revitalized the hypothesis of a reduced ASL secretion. Thus, the role of the ASL pH in the CF is still a controversial matter. In this review we discuss the basis that sustain the role of CFTR in modulating the extracellular pH, and the recent results sustaining the different points of view. Finding the mechanisms of CFTR signaling that determine the susceptibility to infections is crucial to understand the pathophysiology of CF and related lung diseases.
Collapse
Affiliation(s)
- María Macarena Massip-Copiz
- Laboratory of Cellular and Molecular Biology, Institute for Biomedical Research (BIOMED UCA-CONICET), The National Scientific and Technical Research Council (CONICET), and School of Medical Sciences, The Pontifical Catholic University of Argentina (UCA), Buenos Aires, Argentina
| | - Tomás Antonio Santa-Coloma
- Laboratory of Cellular and Molecular Biology, Institute for Biomedical Research (BIOMED UCA-CONICET), The National Scientific and Technical Research Council (CONICET), and School of Medical Sciences, The Pontifical Catholic University of Argentina (UCA), Buenos Aires, Argentina.
| |
Collapse
|
39
|
Marmorstein AD, Johnson AA, Bachman LA, Andrews-Pfannkoch C, Knudsen T, Gilles BJ, Hill M, Gandhi JK, Marmorstein LY, Pulido JS. Mutant Best1 Expression and Impaired Phagocytosis in an iPSC Model of Autosomal Recessive Bestrophinopathy. Sci Rep 2018. [PMID: 29540715 PMCID: PMC5852082 DOI: 10.1038/s41598-018-21651-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Autosomal recessive bestrophinopathy (ARB) is caused by mutations in the gene BEST1 which encodes bestrophin 1 (Best1), an anion channel expressed in retinal pigment epithelial (RPE) cells. It has been hypothesized that ARB represents the human null phenotype for BEST1 and that this occurs due to nonsense mediated decay (NMD). To test this hypothesis, we generated induced pluripotent stem cells (iPSCs) from a patient with ARB and her parents. After differentiation to retinal pigment epithelial (iPSC-RPE) cells, both BEST1 mRNA and Best1 protein expression were compared to controls. BEST1 mRNA expression levels, determined by quantitative PCR, were similar in ARB iPSC-RPE, parental cells, and genetically unrelated controls. Western blotting revealed that CRALBP and RPE65 were expressed within the range delineated by unrelated controls in iPSC-RPE from the ARB donor and her parents. Best1 protein was detected in different clones of ARB iPSC-RPE, but at reduced levels compared to all controls. When tested for the ability to phagocytose photoreceptor outer segments, ARB iPSC-RPE exhibited impaired internalization. These data suggest that impaired phagocytosis is a trait common to the bestrophinopathies. Furthermore, ARB is not universally the result of NMD and ARB, in this patient, is not due to the absence of Best1.
Collapse
Affiliation(s)
- Alan D Marmorstein
- Department of Ophthalmology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Adiv A Johnson
- Department of Ophthalmology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Lori A Bachman
- Department of Ophthalmology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | | | - Travis Knudsen
- Department of Ophthalmology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Benjamin J Gilles
- Department of Ophthalmology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Matthew Hill
- Department of Ophthalmology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Jarel K Gandhi
- Department of Ophthalmology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Lihua Y Marmorstein
- Department of Ophthalmology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Jose S Pulido
- Department of Ophthalmology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| |
Collapse
|
40
|
Kumar V, Mansfield J, Fan R, MacLean A, Li J, Mohan M. miR-130a and miR-212 Disrupt the Intestinal Epithelial Barrier through Modulation of PPARγ and Occludin Expression in Chronic Simian Immunodeficiency Virus-Infected Rhesus Macaques. THE JOURNAL OF IMMUNOLOGY 2018. [PMID: 29514950 DOI: 10.4049/jimmunol.1701148] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Intestinal epithelial barrier dysfunction is a well-known sequela of HIV/SIV infection that persists despite antiretroviral therapy. Although inflammation is a triggering factor, the underlying molecular mechanisms remain unknown. Emerging evidence suggests that epithelial barrier function is epigenetically regulated by inflammation-induced microRNAs (miRNAs). Accordingly, we profiled and characterized miRNA/mRNA expression exclusively in colonic epithelium and identified 46 differentially expressed miRNAs (20 upregulated and 26 downregulated) in chronically SIV-infected rhesus macaques (Macaca mulatta). We bioinformatically crossed the predicted miRNA targets to transcriptomic data and characterized miR-130a and miR-212 as both were predicted to interact with critical epithelial barrier-associated genes. Next, we characterized peroxisome proliferator-activated receptor γ (PPARγ) and occludin (OCLN), predicted targets of miR-130a and miR-212, respectively, as their downregulation has been strongly linked to epithelial barrier disruption and dysbiosis. Immunofluorescence, luciferase reporter, and overexpression studies confirmed the ability of miR-130a and miR-212 to decrease protein expression of PPARγ and OCLN, respectively, and reduce transepithelial electrical resistance. Because Δ-9-tetrahydrocannabinol exerted protective effects in the intestine in our previous studies, we successfully used it to reverse miR-130a- and miR-212-mediated reduction in transepithelial electrical resistance. Finally, ex vivo Δ-9-tetrahydrocannabinol treatment of colon tissue from chronically SIV-infected rhesus macaques significantly increased PPARγ expression. Our findings suggest that dysregulated miR-130a and miR-212 expression in colonic epithelium during chronic HIV/SIV infection can facilitate epithelial barrier disruption by downregulating OCLN and PPARγ expression. Most importantly, our results highlight the beneficial effects of cannabinoids on epithelial barrier function in not just HIV/SIV but potentially other chronic intestinal inflammatory diseases.
Collapse
Affiliation(s)
- Vinay Kumar
- Eurofins Bioanalytics USA, Saint Charles, MO 63304
| | - Joshua Mansfield
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA 70433; and
| | - Rong Fan
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA 70433; and
| | - Andrew MacLean
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA 70433; and
| | - Jian Li
- Department of Global Biostatistics and Data Science, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112
| | - Mahesh Mohan
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA 70433; and
| |
Collapse
|
41
|
Gao DY, Zhang BL, Leung MCT, Au SCL, Wong PYD, Shum WWC. Coupling of TRPV6 and TMEM16A in epithelial principal cells of the rat epididymis. J Gen Physiol 2017; 148:161-82. [PMID: 27481714 PMCID: PMC4969799 DOI: 10.1085/jgp.201611626] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Accepted: 07/13/2016] [Indexed: 01/31/2023] Open
Abstract
Principal cells regulate the ionic environment of the epididymal lumen via unknown mechanisms. Gao et al. use electrophysiological and pharmacological tools to characterize rat principal cells and reveal a TRPV6-mediated calcium conductance and TMEM16A-mediated calcium-activated chloride conductance. The epididymis establishes a congenial environment for sperm maturation and protection. Its fluid is acidic, and the calcium concentration is low and declines along the length of the epididymal tubule. However, our knowledge of ionic currents and mechanisms of calcium homeostasis in rat epididymal epithelial cells remains enigmatic. In this study, to better understand calcium regulation in the epididymis, we use the patch-clamp method to record from single rat cauda epididymal principal cells. We detect a constitutively active Ca2+ current with characteristics that match the epithelial calcium channel TRPV6. Electrophysiological and pharmacological data also reveal a constitutively active calcium-activated chloride conductance (CaCC). Removal of extracellular calcium attenuates not only the TRPV6-like conductance, but also the CaCC. Lanthanide block is time dependent such that the TRPV6-like component is inhibited first, followed by the CaCC. The putative CaCC blocker niflumic acid partially inhibits whole-cell currents, whereas La3+ almost abolishes whole-cell currents in principal cells. Membrane potential measurements reveal an interplay between La3+-sensitive ion channels and those that are sensitive to the specific TMEM16A inhibitor tannic acid. In vivo perfusion of the cauda epididymal tubule shows a substantial rate of Ca2+ reabsorption from the luminal side, which is dose-dependently suppressed by ruthenium red, a putative blocker of epithelial Ca2+ channels and CaCC. Finally, we discover messenger RNA for both TRPV6 and TMEM16A in the rat epididymis and show that their proteins colocalize in the apical membrane of principal cells. Collectively, these data provide evidence for a coupling mechanism between TRPV6 and TMEM16A in principal cells that may play an important role in the regulation of calcium homeostasis in the epididymis.
Collapse
Affiliation(s)
- Da Yuan Gao
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Bao Li Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Matthew C T Leung
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Simon C L Au
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Patrick Y D Wong
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Winnie W C Shum
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
42
|
Abstract
Glaucoma is characterized by a slow and progressive degeneration of the optic nerve, including retinal ganglion cell (RGC) axons in the optic nerve head (ONH), leading to visual impairment. Despite its high prevalence, the biological basis of glaucoma pathogenesis still is not yet fully understood, and the factors contributing to its progression are currently not well characterized. Intraocular pressure (IOP) is the only modifiable risk factor, and reduction of IOP is the standard treatment for glaucoma. However, lowering IOP itself is not always effective for preserving visual function in patients with primary open-angle glaucoma. The second messenger cyclic adenosine 3′,5′-monophosphate (cAMP) regulates numerous biological processes in the central nervous system including the retina and the optic nerve. Although recent studies revealed that cAMP generated by adenylyl cyclases (ACs) is important in regulating aqueous humor dynamics in ocular tissues, such as the ciliary body and trabecular meshwork, as well as cell death and growth in the retina and optic nerve, the functional role and significance of cAMP in glaucoma remain to be elucidated. In this review, we will discuss the functional role of cAMP in aqueous humor dynamics and IOP regulation, and review the current medications, which are related to the cAMP signaling pathway, for glaucoma treatment. Also, we will further focus on cAMP signaling in RGC growth and regeneration by soluble AC as well as ONH astrocytes by transmembrane ACs to understand its potential role in the pathogenesis of glaucoma neurodegeneration
Collapse
Affiliation(s)
- Myoung Sup Shim
- Hamilton Glaucoma Center and Department of Ophthalmology, Shiley Eye Institute, University of California San Diego, La Jolla, CA 92037, USA
| | - Keun-Young Kim
- Center for Research on Biological Systems, National Center for Microscopy and Imaging Research and Department of Neuroscience, University of California San Diego, La Jolla 92093, CA 92093, USA
| | - Won-Kyu Ju
- Hamilton Glaucoma Center and Department of Ophthalmology, Shiley Eye Institute, University of California San Diego, La Jolla, CA 92037, USA
| |
Collapse
|
43
|
Okamoto R, Watanabe M. Functional relevance of intestinal epithelial cells in inflammatory bowel disease. ACTA ACUST UNITED AC 2017; 39:522-527. [PMID: 28049961 DOI: 10.2177/jsci.39.522] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The intestinal epithelium constitutes a physical barrier between inner and outer side of our body. It also functions as a "hub" which connects factors that determine the development of inflammatory bowel disease, such as microbiota, susceptibility genes, and host immune response. Accordingly, recent studies have implicated and further featured the role of intestinal epithelial cell dysfunction in the pathophysiology of inflammatory bowel disease. For example, mucin producing goblet cells are usually "depleted" in ulcerative colitis patients. Studies have shown that those goblet cells exhibit various immune-regulatory functions in addition to mucin production, such as antigen presentation or cytokine production. Paneth cells are another key cell lineage that has been deeply implicated in the pathophysiology of Crohn's disease. Several susceptibility genes for Crohn's disease may lead to impairment of anti-bacterial peptide production and secretion by Paneth cells. Also, other susceptibility genes may determine the survival of Paneth cells, which leads to reduced Paneth cell function in the patient small intestinal mucosa. Further studies may reveal other unexpected roles of the intestinal epithelium in the pathophysiology of inflammatory bowel disease, and may help to develop alternative therapies targeted to intestinal epithelial cell functions.
Collapse
Affiliation(s)
- Ryuichi Okamoto
- Tokyo Medical and Dental University, Center for stem cells and regenerative medicine
| | | |
Collapse
|
44
|
Johnson AA, Guziewicz KE, Lee CJ, Kalathur RC, Pulido JS, Marmorstein LY, Marmorstein AD. Bestrophin 1 and retinal disease. Prog Retin Eye Res 2017; 58:45-69. [PMID: 28153808 DOI: 10.1016/j.preteyeres.2017.01.006] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 01/23/2017] [Accepted: 01/25/2017] [Indexed: 12/18/2022]
Abstract
Mutations in the gene BEST1 are causally associated with as many as five clinically distinct retinal degenerative diseases, which are collectively referred to as the "bestrophinopathies". These five associated diseases are: Best vitelliform macular dystrophy, autosomal recessive bestrophinopathy, adult-onset vitelliform macular dystrophy, autosomal dominant vitreoretinochoroidopathy, and retinitis pigmentosa. The most common of these is Best vitelliform macular dystrophy. Bestrophin 1 (Best1), the protein encoded by the gene BEST1, has been the subject of a great deal of research since it was first identified nearly two decades ago. Today we know that Best1 functions as both a pentameric anion channel and a regulator of intracellular Ca2+ signaling. Best1 is an integral membrane protein which, within the eye, is uniquely expressed in the retinal pigment epithelium where it predominantly localizes to the basolateral plasma membrane. Within the brain, Best1 expression has been documented in both glial cells and astrocytes where it functions in both tonic GABA release and glutamate transport. The crystal structure of Best1 has revealed critical information about how Best1 functions as an ion channel and how Ca2+ regulates that function. Studies using animal models have led to critical insights into the physiological roles of Best1 and advances in stem cell technology have allowed for the development of patient-derived, "disease in a dish" models. In this article we review our knowledge of Best1 and discuss prospects for near-term clinical trials to test therapies for the bestrophinopathies, a currently incurable and untreatable set of diseases.
Collapse
Affiliation(s)
- Adiv A Johnson
- Department of Ophthalmology, Mayo Clinic, Rochester, MN, USA; Nikon Instruments, Melville, NY, USA
| | - Karina E Guziewicz
- Department of Clinical Studies-Philadelphia, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - C Justin Lee
- Center for Neuroscience and Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology, Seoul, South Korea
| | - Ravi C Kalathur
- New York Structural Biology Center, New York Consortium on Membrane Protein Structure, New York, NY, USA
| | - Jose S Pulido
- Department of Ophthalmology, Mayo Clinic, Rochester, MN, USA
| | | | | |
Collapse
|
45
|
Concepcion AR, Vaeth M, Wagner LE, Eckstein M, Hecht L, Yang J, Crottes D, Seidl M, Shin HP, Weidinger C, Cameron S, Turvey SE, Issekutz T, Meyts I, Lacruz RS, Cuk M, Yule DI, Feske S. Store-operated Ca2+ entry regulates Ca2+-activated chloride channels and eccrine sweat gland function. J Clin Invest 2016; 126:4303-4318. [PMID: 27721237 DOI: 10.1172/jci89056] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 08/31/2016] [Indexed: 01/06/2023] Open
Abstract
Eccrine sweat glands are essential for sweating and thermoregulation in humans. Loss-of-function mutations in the Ca2+ release-activated Ca2+ (CRAC) channel genes ORAI1 and STIM1 abolish store-operated Ca2+ entry (SOCE), and patients with these CRAC channel mutations suffer from anhidrosis and hyperthermia at high ambient temperatures. Here we have shown that CRAC channel-deficient patients and mice with ectodermal tissue-specific deletion of Orai1 (Orai1K14Cre) or Stim1 and Stim2 (Stim1/2K14Cre) failed to sweat despite normal sweat gland development. SOCE was absent in agonist-stimulated sweat glands from Orai1K14Cre and Stim1/2K14Cre mice and human sweat gland cells lacking ORAI1 or STIM1 expression. In Orai1K14Cre mice, abolishment of SOCE was associated with impaired chloride secretion by primary murine sweat glands. In human sweat gland cells, SOCE mediated by ORAI1 was necessary for agonist-induced chloride secretion and activation of the Ca2+-activated chloride channel (CaCC) anoctamin 1 (ANO1, also known as TMEM16A). By contrast, expression of TMEM16A, the water channel aquaporin 5 (AQP5), and other regulators of sweat gland function was normal in the absence of SOCE. Our findings demonstrate that Ca2+ influx via store-operated CRAC channels is essential for CaCC activation, chloride secretion, and sweat production in humans and mice.
Collapse
|
46
|
Boedtkjer E, Matchkov VV, Boedtkjer DMB, Aalkjaer C. Negative News: Cl− and HCO3− in the Vascular Wall. Physiology (Bethesda) 2016; 31:370-83. [DOI: 10.1152/physiol.00001.2016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Cl− and HCO3− are the most prevalent membrane-permeable anions in the intra- and extracellular spaces of the vascular wall. Outwardly directed electrochemical gradients for Cl− and HCO3− permit anion channel opening to depolarize vascular smooth muscle and endothelial cells. Transporters and channels for Cl− and HCO3− also modify vascular contractility and structure independently of membrane potential. Transport of HCO3− regulates intracellular pH and thereby modifies the activity of enzymes, ion channels, and receptors. There is also evidence that Cl− and HCO3− transport proteins affect gene expression and protein trafficking. Considering the extensive implications of Cl− and HCO3− in the vascular wall, it is critical to understand how these ions are transported under physiological conditions and how disturbances in their transport can contribute to disease development. Recently, sensing mechanisms for Cl− and HCO3− have been identified in the vascular wall where they modify ion transport and vasomotor function, for instance, during metabolic disturbances. This review discusses current evidence that transport (e.g., via NKCC1, NBCn1, Ca2+-activated Cl− channels, volume-regulated anion channels, and CFTR) and sensing (e.g., via WNK and RPTPγ) of Cl− and HCO3− influence cardiovascular health and disease.
Collapse
Affiliation(s)
| | | | - Donna M. B. Boedtkjer
- Department of Biomedicine, Aarhus University, Denmark
- Department of Clinical Medicine, Aarhus University, Denmark; and
| | - Christian Aalkjaer
- Department of Biomedicine, Aarhus University, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
47
|
Wu L, Sun Y, Ma L, Zhu J, Zhang B, Pan Q, Li Y, Liu H, Diao A, Li Y. A C-terminally truncated mouse Best3 splice variant targets and alters the ion balance in lysosome-endosome hybrids and the endoplasmic reticulum. Sci Rep 2016; 6:27332. [PMID: 27265833 PMCID: PMC4893618 DOI: 10.1038/srep27332] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 05/16/2016] [Indexed: 02/07/2023] Open
Abstract
The Bestrophin family has been characterized as Cl(-) channels in mammals and Na(+) channels in bacteria, but their exact physiological roles remian unknown. In this study, a natural C-terminally truncated variant of mouse Bestrophin 3 (Best3V2) expression in myoblasts and muscles is demonstrated. Unlike full-length Best3, Best3V2 targets the two important intracellular Ca stores: the lysosome and the ER. Heterologous overexpression leads to lysosome swelling and renders it less acidic. Best3V2 overexpression also results in compromised Ca(2+) release from the ER. Knocking down endogenous Best3 expression in myoblasts makes these cells more excitable in response to Ca(2+) mobilizing reagents, such as caffeine. We propose that Best3V2 in myoblasts may work as a tuner to control Ca(2+) release from intracellular Ca(2+) stores.
Collapse
Affiliation(s)
- Lichang Wu
- Department of Animal Sciences and Technology, Qingdao Agricultural University, Qingdao, China.,College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Yu Sun
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Liqiao Ma
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Jun Zhu
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Baoxia Zhang
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Qingjie Pan
- Department of Animal Sciences and Technology, Qingdao Agricultural University, Qingdao, China
| | - Yuyin Li
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Huanqi Liu
- Department of Animal Sciences and Technology, Qingdao Agricultural University, Qingdao, China
| | - Aipo Diao
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Yinchuan Li
- Department of Animal Sciences and Technology, Qingdao Agricultural University, Qingdao, China
| |
Collapse
|
48
|
Wu MM, Lou J, Song BL, Gong YF, Li YC, Yu CJ, Wang QS, Ma TX, Ma K, Hartzell HC, Duan DD, Zhao D, Zhang ZR. Hypoxia augments the calcium-activated chloride current carried by anoctamin-1 in cardiac vascular endothelial cells of neonatal mice. Br J Pharmacol 2016; 171:3680-92. [PMID: 24758567 PMCID: PMC4128065 DOI: 10.1111/bph.12730] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 04/01/2014] [Accepted: 04/06/2014] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND AND PURPOSE The molecular identity of calcium-activated chloride channels (CaCCs) in vascular endothelial cells remains unknown. This study sought to identify whether anoctamin-1 (Ano1, also known as TMEM16A) functions as a CaCC and whether hypoxia alters the biophysical properties of Ano1 in mouse cardiac vascular endothelial cells (CVECs). EXPERIMENTAL APPROACH Western blot, quantitative real-time PCR, confocal imaging analysis and patch-clamp analysis combined with pharmacological approaches were used to determine whether Ano1 was expressed and functioned as CaCC in CVECs. KEY RESULTS Ano1 was expressed in CVECs. The biophysical properties of the current generated in the CVECs, including the Ca2+ and voltage dependence, outward rectification, anion selectivity and the pharmacological profile, are similar to those described for CaCCs. The density of ICl(Ca) detected in CVECs was significantly inhibited by T16Ainh-A01, an Ano1 inhibitor, and a pore-targeting, specific anti-Ano1 antibody, and was markedly decreased in Ano1 gene knockdown CVECs. The density of ICl(Ca) was significantly potentiated in CVECs exposed to hypoxia, and this hypoxia-induced increase in the density of ICl(Ca) was inhibited by T16Ainh-A01 or anti-Ano1 antibody. Hypoxia also increased the current density of ICl(Ca) in Ano1 gene knockdown CVECs. CONCLUSIONS AND IMPLICATIONS Ano1 formed CaCC in CVECs of neonatal mice. Hypoxia enhances Ano1-mediated ICl(Ca) density via increasing its expression, altering the ratio of its splicing variants, sensitivity to membrane voltage and to Ca2+. Ano1 may play a role in the pathophysiological processes during ischaemia in heart, and therefore, Ano1 might be a potential therapeutic target to prevent ischaemic damage.
Collapse
Affiliation(s)
- Ming-Ming Wu
- Departments of Clinical Pharmacy and Cardiology, Institute of Clinical Pharmacy, The 2nd Affiliated Hospital, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin Medical University, Harbin, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
A Novel Mechanism of pH Buffering in C. elegans Glia: Bicarbonate Transport via the Voltage-Gated ClC Cl- Channel CLH-1. J Neurosci 2016; 35:16377-97. [PMID: 26674864 DOI: 10.1523/jneurosci.3237-15.2015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED An important function of glia is the maintenance of the ionic composition and pH of the synaptic microenvironment. In terms of pH regulation, HCO3 (-) buffering has been shown to be important in both glia and neurons. Here, we used in vivo fluorescent pH imaging and RNA sequencing of the amphid sheath glia of Caenorhabditis elegans to reveal a novel mechanism of cellular HCO3 (-) uptake. While the classical mechanism of HCO3 (-) uptake involves Na(+)/HCO3 (-) cotransporters, here we demonstrate that the C. elegans ClC Cl(-) channel CLH-1 is highly permeable to HCO3 (-) and mediates HCO3 (-) uptake into amphid sheath glia. CLH-1 has homology and electrophysiological properties similar to the mammalian ClC-2 Cl(-) channel. Our data suggest that, in addition to maintaining synaptic Cl(-) concentration, these channels may also be involved in maintenance of synaptic pH via HCO3 (-) flux. These findings provide an exciting new facet of study regarding how pH is regulated in the brain. SIGNIFICANCE STATEMENT Maintenance of pH is essential for the physiological function of the nervous system. HCO3 (-) is crucial for pH regulation and is transported into the cell via ion transporters, including ion channels, the molecular identity of which remains unclear. In this manuscript, we describe our discovery that the C. elegans amphid sheath glia regulate intracellular pH via HCO3 (-) flux through the voltage-gated ClC channel CLH-1. This represents a novel function for ClC channels, which has implications for their possible role in mammalian glial pH regulation. This discovery may also provide a novel therapeutic target for pathologic conditions, such as ischemic stroke where acidosis leads to widespread death of glia and subsequently neurons.
Collapse
|
50
|
Okamoto R, Watanabe M. Role of epithelial cells in the pathogenesis and treatment of inflammatory bowel disease. J Gastroenterol 2016; 51:11-21. [PMID: 26138071 DOI: 10.1007/s00535-015-1098-4] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Accepted: 06/15/2015] [Indexed: 02/04/2023]
Abstract
In the past decades, continuous effort has been paid to deeply understanding the pathophysiology of inflammatory bowel diseases (IBD), such as ulcerative colitis or Crohn's disease. As the disease typically arises as chronic inflammation of the gastrointestinal mucosa, research has been focused on how such an uncontrolled, deleterious immune response may arise and persist in a certain cohort of patients. Based on those immunologic analyses, the establishment of anti-TNF-α therapy, and the following series of biologic agents achieved great success and dramatically changed the therapeutic strategy of IBD patients. However, to guarantee long-term remission of the disease, the therapeutic standard has been raised to achieve "mucosal healing", which requires complete repair of the gastrointestinal mucosa. Recent studies have revealed the unexpected importance of epithelial cells in the pathophysiology of IBD. The general barrier function as well as the cell lineage-specific functions have been deeply attributed to the development of chronic intestinal inflammation. Also, the groundbreaking establishment of the in vitro intestinal stem cell culture system has opened up a way of developing stem cell transplantation therapy to treat otherwise refractory ulcers that may persist in IBD patients. In this review, we would like to focus on the role of epithelial cells in the pathophysiology of IBD, and also give a perspective to the upcoming development of regenerative therapies that may become one of the therapeutic choices to achieve mucosal healing in refractory patients of IBD.
Collapse
Affiliation(s)
- Ryuichi Okamoto
- Center for Stem Cell and Regenerative Medicine, Graduate School, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-Ku, Tokyo, 113-8519, Japan.
| | - Mamoru Watanabe
- Department of Gastroenterology and Hepatology, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|