1
|
Hopton RE, Jahahn NJ, Zemper AE. Lrig1 drives cryptogenesis and restrains proliferation during colon development. Am J Physiol Gastrointest Liver Physiol 2023; 325:G570-G581. [PMID: 37873577 PMCID: PMC11192189 DOI: 10.1152/ajpgi.00094.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 10/04/2023] [Accepted: 10/13/2023] [Indexed: 10/25/2023]
Abstract
Growth and specification of the mouse intestine occurs in utero and concludes after birth. Although numerous studies have examined this developmental process in the small intestine, far less is known about the cellular and molecular cues required for colon development. In this study, we examine the morphological events leading to crypt formation, epithelial cell differentiation, proliferation, and the emergence and expression of a stem and progenitor cell marker Lrig1. Through multicolor lineage tracing, we show Lrig1-expressing cells are present at birth and behave as stem cells to establish clonal crypts within 3 wk of life. In addition, we use an inducible knockout mouse to eliminate Lrig1 and show Lrig1 restrains proliferation within a critical developmental time window, without impacting colonic epithelial cell differentiation. Our study illustrates morphological changes during crypt development and the importance of Lrig1 in the developing colon.NEW & NOTEWORTHY Our studies define the importance of studying Lrig1 in colon development. We address a critical gap in the intestinal development literature and provide new information about the molecular cues that guide colon development. Using a novel, inducible knockout of Lrig1, we show Lrig1 is required for appropriate colon epithelial growth and illustrate the importance of Lrig1-expressing cells in the establishment of colonic crypts.
Collapse
Affiliation(s)
- Rachel E Hopton
- Institute of Molecular Biology, University of Oregon, Eugene, Oregon, United States
- Department of Biology, University of Oregon, Eugene, Oregon, United States
| | - Nicholas J Jahahn
- Institute of Molecular Biology, University of Oregon, Eugene, Oregon, United States
- Department of Biology, University of Oregon, Eugene, Oregon, United States
| | - Anne E Zemper
- Department of Biology, University of Oregon, Eugene, Oregon, United States
| |
Collapse
|
2
|
Shimizu S, Kondo J, Onuma K, Coppo R, Ota K, Kamada M, Harada Y, Tanaka Y, Nakazawa MA, Tamada Y, Okuno Y, Kawada K, Obama K, Coffey RJ, Fujiwara Y, Inoue M. Inhibition of the bone morphogenetic protein pathway suppresses tumor growth through downregulation of epidermal growth factor receptor in MEK/ERK-dependent colorectal cancer. Cancer Sci 2023; 114:3636-3648. [PMID: 37357017 PMCID: PMC10475764 DOI: 10.1111/cas.15882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 05/22/2023] [Accepted: 05/27/2023] [Indexed: 06/27/2023] Open
Abstract
The bone morphogenetic protein (BMP) pathway promotes differentiation and induces apoptosis in normal colorectal epithelial cells. However, its role in colorectal cancer (CRC) is controversial, where it can act as context-dependent tumor promoter or tumor suppressor. Here we have found that CRC cells reside in a BMP-rich environment based on curation of two publicly available RNA-sequencing databases. Suppression of BMP using a specific BMP inhibitor, LDN193189, suppresses the growth of select CRC organoids. Colorectal cancer organoids treated with LDN193189 showed a decrease in epidermal growth factor receptor, which was mediated by protein degradation induced by leucine-rich repeats and immunoglobulin-like domains protein 1 (LRIG1) expression. Among 18 molecularly characterized CRC organoids, suppression of growth by BMP inhibition correlated with induction of LRIG1 gene expression. Notably, knockdown of LRIG1 in organoids diminished the growth-suppressive effect of LDN193189. Furthermore, in CRC organoids, which are susceptible to growth suppression by LDN193189, simultaneous treatment with LDN193189 and trametinib, an FDA-approved MEK inhibitor, resulted in cooperative growth inhibition both in vitro and in vivo. Taken together, the simultaneous inhibition of BMP and MEK could be a novel treatment option in CRC cases, and evaluating in vitro growth suppression and LRIG1 induction by BMP inhibition using patient-derived organoids could offer functional biomarkers for predicting potential responders to this regimen.
Collapse
Affiliation(s)
- Shota Shimizu
- Department of Clinical Bioresource Research and DevelopmentKyoto University Graduate School of MedicineKyotoJapan
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of MedicineTottori University Faculty of MedicineTottoriJapan
| | - Jumpei Kondo
- Department of Clinical Bioresource Research and DevelopmentKyoto University Graduate School of MedicineKyotoJapan
- Department of BiochemistryOsaka International Cancer InstituteOsakaJapan
- Department of Molecular Biochemistry and Clinical Investigation, Division of Health ScienceOsaka University Graduate School of MedicineOsakaJapan
- Epithelial Biology CenterVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Kunishige Onuma
- Department of Clinical Bioresource Research and DevelopmentKyoto University Graduate School of MedicineKyotoJapan
| | - Roberto Coppo
- Department of Clinical Bioresource Research and DevelopmentKyoto University Graduate School of MedicineKyotoJapan
| | - Kasumi Ota
- Graduate School of Medicine and Faculty of MedicineKyoto UniversityKyotoJapan
| | - Mayumi Kamada
- Graduate School of Medicine and Faculty of MedicineKyoto UniversityKyotoJapan
| | - Yohei Harada
- Graduate School of Medicine and Faculty of MedicineKyoto UniversityKyotoJapan
| | - Yoshihisa Tanaka
- Graduate School of Pharmaceutical SciencesKyoto UniversityKyotoJapan
- RIKEN Center for Computational Science(R‐CCS)HPC/HPC‐ and AI‐driven Drug Development Platform DivisionKobeJapan
| | - Mai Adachi Nakazawa
- Graduate School of Medicine and Faculty of MedicineKyoto UniversityKyotoJapan
- Department of Medical Data Intelligence and Data Analysis Division, Innovation Center for Health PromotionHirosaki UniversityHirosakiJapan
| | - Yoshinori Tamada
- Department of Medical Data Intelligence and Data Analysis Division, Innovation Center for Health PromotionHirosaki UniversityHirosakiJapan
| | - Yasushi Okuno
- Graduate School of Medicine and Faculty of MedicineKyoto UniversityKyotoJapan
- RIKEN Center for Computational Science(R‐CCS)HPC/HPC‐ and AI‐driven Drug Development Platform DivisionKobeJapan
| | - Kenji Kawada
- Department of Surgery, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Kazutaka Obama
- Department of Surgery, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Robert J. Coffey
- Epithelial Biology CenterVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Yoshiyuki Fujiwara
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of MedicineTottori University Faculty of MedicineTottoriJapan
| | - Masahiro Inoue
- Department of Clinical Bioresource Research and DevelopmentKyoto University Graduate School of MedicineKyotoJapan
- Department of BiochemistryOsaka International Cancer InstituteOsakaJapan
| |
Collapse
|
3
|
Hopton RE, Jahahn NJ, Zemper AE. The Role of Lrig1 in the Development of the Colonic Epithelium. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.02.539114. [PMID: 37205411 PMCID: PMC10187246 DOI: 10.1101/2023.05.02.539114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Growth and specification of the mouse intestine occurs in utero and concludes after birth. While numerous studies have examined this developmental process in the small intestine, far less is known about the cellular and molecular cues required for colon development. In this study, we examine the morphological events leading to crypt formation, epithelial cell differentiation, areas of proliferation, and the emergence and expression of a stem and progenitor cell marker Lrig1. Through multicolor lineage tracing, we show Lrig1 expressing cells are present at birth and behave as stem cells to establish clonal crypts within three weeks after birth. In addition, we use an inducible knockout mouse to eliminate Lrig1 during colon development and show loss of Lrig1 restrains proliferation within a critical developmental time window, without impacting colonic epithelial cell differentiation. Our study illustrates the morphological changes that occur during crypt development and the importance of Lrig1 in the developing colon.
Collapse
|
4
|
Huizinga JD, Hussain A, Chen JH. Interstitial cells of Cajal and human colon motility in health and disease. Am J Physiol Gastrointest Liver Physiol 2021; 321:G552-G575. [PMID: 34612070 DOI: 10.1152/ajpgi.00264.2021] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Our understanding of human colonic motility, and autonomic reflexes that generate motor patterns, has increased markedly through high-resolution manometry. Details of the motor patterns are emerging related to frequency and propagation characteristics that allow linkage to interstitial cells of Cajal (ICC) networks. In studies on colonic motor dysfunction requiring surgery, ICC are almost always abnormal or significantly reduced. However, there are still gaps in our knowledge about the role of ICC in the control of colonic motility and there is little understanding of a mechanistic link between ICC abnormalities and colonic motor dysfunction. This review will outline the various ICC networks in the human colon and their proven and likely associations with the enteric and extrinsic autonomic nervous systems. Based on our extensive knowledge of the role of ICC in the control of gastrointestinal motility of animal models and the human stomach and small intestine, we propose how ICC networks are underlying the motor patterns of the human colon. The role of ICC will be reviewed in the autonomic neural reflexes that evoke essential motor patterns for transit and defecation. Mechanisms underlying ICC injury, maintenance, and repair will be discussed. Hypotheses are formulated as to how ICC dysfunction can lead to motor abnormalities in slow transit constipation, chronic idiopathic pseudo-obstruction, Hirschsprung's disease, fecal incontinence, diverticular disease, and inflammatory conditions. Recent studies on ICC repair after injury hold promise for future therapies.
Collapse
Affiliation(s)
- Jan D Huizinga
- Division of Gastroenterology, Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Amer Hussain
- Division of Gastroenterology, Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Ji-Hong Chen
- Division of Gastroenterology, Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
5
|
Li T, Hu M, Jiang C, Zhang D, Gao M, Xia J, Miao M, Shi G, Li H, Zhang J, Yin Z. Laxative effect and mechanism of Tiantian Capsule on loperamide-induced constipation in rats. JOURNAL OF ETHNOPHARMACOLOGY 2021; 266:113411. [PMID: 32980482 DOI: 10.1016/j.jep.2020.113411] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 09/08/2020] [Accepted: 09/08/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tiantian capsule (TTC), as a functional food, which consists of four herb medicines, including Aloe vera Burm.f. (25%), leaf juices, dried; Cucurbita moschata Duch. (25%), fructus, dried; Poria cocos (Schw.) Wolf. (12.5%), sclerotium, dried; Tremella fuciformis Berk. (12.5%), fruiting bodies, dried, and one extract xylooligosaccharides (25%) from Maize Cob by enzymolysis, has been commonly used in China to ameliorate constipation. AIM OF THE STUDY The aim of the work is to elucidate the potential laxative mechanisms of TTC in loperamide-induced constipated rats. MATERIALS AND METHODS LC-MS/MS was employed for analyzing the TTC extract. The gastrointestinal transit was evaluated by X-ray. The H&E and Alcian-Blue stain were applied to determine the changes of goblet cells and mucus layer, respectively. Meanwhile, levels of neurotransmitters were evaluated by enzyme-linked immunosorbent assay. The protein expressions were also measured by immunohistochemistry and Western blot. RESULTS Our results showed that TTC administration attenuated constipation responses in aspects of fecal pellets number, water content of feces, stomach emptying and gastrointestinal transit. Further investigations revealed that TTC treatment not only induced the recovery of neurotransmitters, such as motilin, substance P, somatostatin, endothelin and vasoactive intestinal peptide, but also up-regulated the expressions of c-kit and stem cell factor (SCF). Additionally, the number of goblet cells and thickness of the mucus layer were elevated, and the guanylate cyclase C-cGMP signal pathway was also up-regulated after TTC treatment. CONCLUSION Our findings demonstrated that the laxative effect of TTC in constipation rats is probably due to the regulation of bowel movement and intestinal fluid secretion.
Collapse
Affiliation(s)
- Tian Li
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, PR China; Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, PR China; Department of TCMs Pharmaceuticals & State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Mengmeng Hu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, PR China; Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, PR China; Department of TCMs Pharmaceuticals & State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Cuihua Jiang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, PR China; Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, PR China
| | - Dongjian Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, PR China; Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, PR China
| | - Meng Gao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, PR China; Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, PR China
| | - Jianwei Xia
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, PR China; Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, PR China; Department of TCMs Pharmaceuticals & State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Mengqi Miao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, PR China
| | - Gaofeng Shi
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, PR China
| | - Hui Li
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, PR China
| | - Jian Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, PR China; Laboratories of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, PR China.
| | - Zhiqi Yin
- Department of TCMs Pharmaceuticals & State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China.
| |
Collapse
|
6
|
Ji Y, Kumar R, Gokhale A, Chao HP, Rycaj K, Chen X, Li Q, Tang DG. LRIG1, a regulator of stem cell quiescence and a pleiotropic feedback tumor suppressor. Semin Cancer Biol 2021; 82:120-133. [PMID: 33476721 PMCID: PMC8286266 DOI: 10.1016/j.semcancer.2020.12.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 12/04/2020] [Accepted: 12/16/2020] [Indexed: 12/14/2022]
Abstract
LRIG1, leucine-rich repeats and immunoglobulin-like domains protein 1, was discovered more than 20 years ago and has been shown to be downregulated or lost, and to function as a tumor suppressor in several cancers. Another well-reported biological function of LRIG1 is to regulate and help enforce the quiescence of adult stem cells (SCs). In both contexts, LRIG1 regulates SC quiescence and represses tumor growth via, primarily, antagonizing the expression and activities of ERBB and other receptor tyrosine kinases (RTKs). We have recently reported that in treatment-naïve human prostate cancer (PCa), LRIG1 is primarily regulated by androgen receptor (AR) and is prominently overexpressed. In castration-resistant PCa (CRPC), both LRIG1 and AR expression becomes heterogeneous and, frequently, discordant. Importantly, in both androgen-dependent PCa and CRPC models, LRIG1 exhibits tumor-suppressive functions. Moreover, LRIG1 induction inhibits the growth of pre-established AR+ and AR− PCa. Here, upon a brief introduction of the LRIG1 and the LRIG family, we provide an updated overview on LRIG1 functions in regulating SC quiescence and repressing tumor development. We further highlight the expression, regulation and functions of LRIG1 in treatment-naïve PCa and CRPC. We conclude by offering the perspectives of identifying novel cancer-specific LRIG1-interacting signaling partners and developing LRIG1-based anti-cancer therapeutics and diagnostic/prognostic biomarkers.
Collapse
Affiliation(s)
- Yibing Ji
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| | - Rahul Kumar
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Abhiram Gokhale
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Hseu-Ping Chao
- Department of Epigenetics & Mol. Carcinogenesis, the University of Texas M.D Anderson Cancer Center, Smithville, TX 78957, USA
| | - Kiera Rycaj
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; Department of Epigenetics & Mol. Carcinogenesis, the University of Texas M.D Anderson Cancer Center, Smithville, TX 78957, USA
| | - Xin Chen
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Qiuhui Li
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| | - Dean G Tang
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; Department of Epigenetics & Mol. Carcinogenesis, the University of Texas M.D Anderson Cancer Center, Smithville, TX 78957, USA.
| |
Collapse
|
7
|
Kondo J, Huh WJ, Franklin JL, Heinrich MC, Rubin BP, Coffey RJ. A smooth muscle-derived, Braf-driven mouse model of gastrointestinal stromal tumor (GIST): evidence for an alternative GIST cell-of-origin. J Pathol 2020; 252:441-450. [PMID: 32944951 PMCID: PMC7802691 DOI: 10.1002/path.5552] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 08/27/2020] [Accepted: 09/14/2020] [Indexed: 12/21/2022]
Abstract
Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumor of the gut. GISTs are thought to arise solely from interstitial cells of Cajal (ICC), a KIT-positive population that controls gut motility. Activating gain-of-function mutations in KIT and PDGFRA are the most frequent driver events, and most of these tumors are responsive to the tyrosine kinase inhibitor imatinib. Less common drivers include mutant BRAFV600E and these tumors are resistant to imatinib. A mouse model of GIST was recently reported using Etv1, the master transcriptional regulator of ICC-intramuscular (IM) and ICC-myenteric (MY), to induce mutant Braf expression. ICC hyperplasia was observed in Etv1CreERT2 ;BrafLSL-V600E/+ mice but loss of Trp53 was required for development of GIST. We identified previously expression of the pan-ErbB negative regulator, LRIG1, in two distinct subclasses of ICC [ICC-deep muscular plexus (DMP) in small intestine and ICC-submucosal plexus (SMP) in colon] and that LRIG1 regulated their development from smooth muscle cell progenitors. Using Lrig1CreERT2 to induce BrafV600E , we observed ICC hyperplasia beyond the confines of ICC-DMP and ICC-SMP expression, suggesting smooth muscle cells as the cell-of-origin. To examine this possibility, we selectively activated BrafV600E in smooth muscle cells. Myh11CreERT2 ;BrafLSL-V600E/+ mice developed not only ICC hyperplasia but also GIST and in the absence of Trp53 disruption. In addition to providing a simpler model for mutant Braf GIST, these results provide conclusive evidence for smooth muscle cells as an alternative cell-of-origin for GIST. © 2020 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jumpei Kondo
- Department of Medicine, Vanderbilt University Medical Center, TN, USA
- Epithelial Biology Center, Vanderbilt University Medical Center, TN, USA
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine Kyoto University, Kyoto, Japan
| | - Won Jae Huh
- Epithelial Biology Center, Vanderbilt University Medical Center, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, TN, USA
| | - Jeffrey L Franklin
- Department of Medicine, Vanderbilt University Medical Center, TN, USA
- Epithelial Biology Center, Vanderbilt University Medical Center, TN, USA
| | - Michael C Heinrich
- Hematology/Medical Oncology, Portland VA Health Care System and OHSU Knight Cancer Institute, OR, USA
| | - Brian P Rubin
- Robert J Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic Foundation, OH, USA
| | - Robert J Coffey
- Department of Medicine, Vanderbilt University Medical Center, TN, USA
- Epithelial Biology Center, Vanderbilt University Medical Center, TN, USA
| |
Collapse
|
8
|
Iino S, Horiguchi K, Horiguchi S. Investigation of Novel c-Kit-expressing Smooth Muscle Cells in Murine Cecum. Acta Histochem Cytochem 2020; 53:11-19. [PMID: 32410749 PMCID: PMC7212203 DOI: 10.1267/ahc.20003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 03/03/2020] [Indexed: 12/28/2022] Open
Abstract
In the gastrointestinal tract musculatures, c-Kit receptor tyrosine kinase is specifically expressed in interstitial cells of Cajal (ICC). ICC are distributed among the smooth muscle cells and are either bipolar or multipolar in shape. Our previous and current study shows that c-Kit-immunopositive smooth muscle cells are present in the murine cecum. Here, we found that c-Kit-expressing smooth muscle cells (named Kit-SM cells) are situated at the submucosal surface of the circular muscle layer. These cells showed smooth muscle actin and myosin immunoreactivities and ultrastructural features such as thick and thin filaments and caveolae. Kit-SM cells also expressed TMEM16A and LRIG1, which are known to be expressed in ICC. Although the functional significance of Kit-SM cells has yet to be revealed, these cells can be considered to have proliferation or differentiation potential in the cecal musculature.
Collapse
Affiliation(s)
- Satoshi Iino
- Department of Anatomy, University of Fukui Faculty of Medical Sciences
| | | | - Satomi Horiguchi
- Department of Anatomy, University of Fukui Faculty of Medical Sciences
| |
Collapse
|
9
|
Li Q, Liu B, Chao HP, Ji Y, Lu Y, Mehmood R, Jeter C, Chen T, Moore JR, Li W, Liu C, Rycaj K, Tracz A, Kirk J, Calhoun-Davis T, Xiong J, Deng Q, Huang J, Foster BA, Gokhale A, Chen X, Tang DG. LRIG1 is a pleiotropic androgen receptor-regulated feedback tumor suppressor in prostate cancer. Nat Commun 2019; 10:5494. [PMID: 31792211 PMCID: PMC6889295 DOI: 10.1038/s41467-019-13532-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 11/06/2019] [Indexed: 12/13/2022] Open
Abstract
LRIG1 has been reported to be a tumor suppressor in gastrointestinal tract and epidermis. However, little is known about the expression, regulation and biological functions of LRIG1 in prostate cancer (PCa). We find that LRIG1 is overexpressed in PCa, but its expression correlates with better patient survival. Functional studies reveal strong tumor-suppressive functions of LRIG1 in both AR+ and AR- xenograft models, and transgenic expression of LRIG1 inhibits tumor development in Hi-Myc and TRAMP models. LRIG1 also inhibits castration-resistant PCa and exhibits therapeutic efficacy in pre-established tumors. We further show that 1) AR directly transactivates LRIG1 through binding to several AR-binding sites in LRIG1 locus, and 2) LRIG1 dampens ERBB expression in a cell type-dependent manner and inhibits ERBB2-driven tumor growth. Collectively, our study indicates that LRIG1 represents a pleiotropic AR-regulated feedback tumor suppressor that functions to restrict oncogenic signaling from AR, Myc, ERBBs, and, likely, other oncogenic drivers.
Collapse
Affiliation(s)
- Qiuhui Li
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, 430079, Wuhan, China
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Bigang Liu
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Hsueh-Ping Chao
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Yibing Ji
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Yue Lu
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Rashid Mehmood
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Collene Jeter
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Taiping Chen
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - John R Moore
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Wenqian Li
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Can Liu
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Kiera Rycaj
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Amanda Tracz
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Jason Kirk
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Tammy Calhoun-Davis
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Jie Xiong
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Qu Deng
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Jiaoti Huang
- Department of Pathology, Duke University of School of Medicine, Durham, NC, 27710, USA
| | - Barbara A Foster
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Abhiram Gokhale
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Xin Chen
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA.
- Department of Oncology, Tongji Hospital, Tongji Medical School, Huazhong University of Science and Technology (HUST), 430030, Wuhan, China.
| | - Dean G Tang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA.
- Cancer Stem Cell Institute, Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, 200120, Shanghai, China.
| |
Collapse
|
10
|
Walsh KT, Zemper AE. The Enteric Nervous System for Epithelial Researchers: Basic Anatomy, Techniques, and Interactions With the Epithelium. Cell Mol Gastroenterol Hepatol 2019; 8:369-378. [PMID: 31108231 PMCID: PMC6718943 DOI: 10.1016/j.jcmgh.2019.05.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 05/02/2019] [Accepted: 05/03/2019] [Indexed: 02/08/2023]
Abstract
The intestinal epithelium does not function in isolation, but interacts with many components including the Enteric Nervous System (ENS). Understanding ENS and intestinal epithelium interactions requires multidisciplinary approaches to uncover cells involved, mechanisms used, and the ultimate influence on intestinal physiology. This review is intended to serve as a reference for epithelial biologists interested in studying these interactions. With this in mind, this review aims to summarize the basic anatomy of the epithelium and ENS, mechanisms by which they interact, and techniques used to study these interactions. We highlight in vitro, ex vivo and in vivo techniques. Additionally, ENS influence on epithelial proliferation and gene expression within stem and differentiated cells as well as gastrointestinal cancer are discussed.
Collapse
Key Words
- 5-ht, 5-hydroxytryptamine
- 5-ht3r, 5-hydroxytryptamine 3 receptor
- ach, acetylcholine
- aitc, allyl isothicyanate
- cpi, crypt proliferation index
- eec, enteroendocrine cell
- ens, enteric nervous system
- gi, gastrointestinal
- hio, human intestinal organoid
- isc, intestinal stem cell
- lgr5, leucine-rich repeat–containing g protein–coupled receptor
- ne, norepinephrine
- ngf, nerve growth factor
- si, small intestine
- ta, transit-amplifying
Collapse
Affiliation(s)
- Kathleen T. Walsh
- Institute of Molecular Biology, University of Oregon, Eugene, Oregon,Institute of Neuroscience, University of Oregon, Eugene, Oregon,Department of Biology, University of Oregon, Eugene, Oregon
| | - Anne E. Zemper
- Institute of Molecular Biology, University of Oregon, Eugene, Oregon,Department of Biology, University of Oregon, Eugene, Oregon,Correspondence Address correspondence to: Anne E. Zemper, PhD, University of Oregon, 218 Streisinger Hall, 1370 Franklin Boulevard, Eugene, Oregon 97401. fax: (541) 346–6056.
| |
Collapse
|
11
|
Hwang SJ, Pardo DM, Zheng H, Bayguinov Y, Blair PJ, Fortune‐Grant R, Cook RS, Hennig GW, Shonnard MC, Grainger N, Peri LE, Verma SD, Rock J, Sanders KM, Ward SM. Differential sensitivity of gastric and small intestinal muscles to inducible knockdown of anoctamin 1 and the effects on gastrointestinal motility. J Physiol 2019; 597:2337-2360. [PMID: 30843201 PMCID: PMC6487927 DOI: 10.1113/jp277335] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 02/22/2019] [Indexed: 12/13/2022] Open
Abstract
KEY POINTS Electrical pacemaking in gastrointestinal muscles is generated by specialized interstitial cells of Cajal that produce the patterns of contractions required for peristalsis and segmentation in the gut. The calcium-activated chloride conductance anoctamin-1 (Ano1) has been shown to be responsible for the generation of pacemaker activity in GI muscles, but this conclusion is established from studies of juvenile animals in which effects of reduced Ano1 on gastric emptying and motor patterns could not be evaluated. Knocking down Ano1 expression using Cre/LoxP technology caused dramatic changes in in gastric motor activity, with disrupted slow waves, abnormal phasic contractions and delayed gastric emptying; modest changes were noted in the small intestine. Comparison of the effects of Ano1 antagonists on muscles from juvenile and adult small intestinal muscles suggests that conductances in addition to Ano1 may develop with age and contribute to pacemaker activity. ABSTRACT Interstitial cells of Cajal (ICC) generate slow waves and transduce neurotransmitter signals in the gastrointestinal (GI) tract, facilitating normal motility patterns. ICC express a Ca2+ -activated Cl- conductance (CaCC), and constitutive knockout of the channel protein anoctamin-1 leads to loss of slow waves in gastric and intestinal muscles. These knockout experiments were performed on juvenile mice. However, additional experiments demonstrated significant differences in the sensitivity of gastric and intestinal muscles to antagonists of anoctamin-1 channels. Furthermore, the significance of anoctamin-1 and the electrical and mechanical behaviours facilitated by this conductance have not been evaluated on the motor behaviours of adult animals. Cre/loxP technology was used to generate cell-specific knockdowns of anoctamin-1 in ICC (KitCreERT2/+ ;Ano1tm2jrr/+ ) in GI muscles. The recombination efficiency of KitCreERT was evaluated with an eGFP reporter, molecular techniques and immunohistochemistry. Electrical and contractile experiments were used to examine the consequences of anoctamin-1 knockdown on pacemaker activity, mechanical responses, gastric motility patterns, gastric emptying and GI transit. Reduced anoctamin-1 caused loss of gastric, but not intestinal slow waves. Irregular spike complexes developed in gastric muscles, leading to uncoordinated antral contractions, delayed gastric emptying and increased total GI transit time. Slow waves in intestinal muscles of juvenile mice were more sensitive to anoctamin-1 antagonists than slow waves in adult muscles. The low susceptibility to anoctamin-1 knockdown and weak efficacy of anoctamin-1 antagonists in inhibiting slow waves in adult small intestinal muscles suggest that a conductance in addition to anoctamin-1 may develop in small intestinal ICC with ageing and contribute to pacemaker activity.
Collapse
Affiliation(s)
- Sung Jin Hwang
- Department of Physiology & Cell BiologyUniversity of NevadaReno School of MedicineRenoNV89557USA
| | - David M. Pardo
- Department of AnatomyUniversity of CaliforniaSan FranciscoSan FranciscoCA94143USA
| | - Haifeng Zheng
- Department of Physiology & Cell BiologyUniversity of NevadaReno School of MedicineRenoNV89557USA
| | - Yulia Bayguinov
- Department of Physiology & Cell BiologyUniversity of NevadaReno School of MedicineRenoNV89557USA
| | - Peter J. Blair
- Department of Physiology & Cell BiologyUniversity of NevadaReno School of MedicineRenoNV89557USA
| | - Rachael Fortune‐Grant
- Faculty of BiologyMedicine and HealthSchool of Biological SciencesUniversity of ManchesterUK
| | - Robert S. Cook
- School of Molecular and Cellular BiologyUniversity of LeedsLeedsLS2 9JTUK
| | - Grant W. Hennig
- Department of PharmacologyThe University of VermontUVM College of MedicineBurlingtonVT05405USA
| | - Matthew C. Shonnard
- Department of Physiology & Cell BiologyUniversity of NevadaReno School of MedicineRenoNV89557USA
| | - Nathan Grainger
- Department of Physiology & Cell BiologyUniversity of NevadaReno School of MedicineRenoNV89557USA
| | - Lauren E. Peri
- Department of Physiology & Cell BiologyUniversity of NevadaReno School of MedicineRenoNV89557USA
| | - Sonali Deep Verma
- Department of AnatomyUniversity of CaliforniaSan FranciscoSan FranciscoCA94143USA
| | - Jason Rock
- Centre for Regenerative MedicineBoston University School of MedicineBostonMA02118USA
| | - Kenton M. Sanders
- Department of Physiology & Cell BiologyUniversity of NevadaReno School of MedicineRenoNV89557USA
| | - Sean M. Ward
- Department of Physiology & Cell BiologyUniversity of NevadaReno School of MedicineRenoNV89557USA
| |
Collapse
|
12
|
Lin Q, Qin M, Zhao SG, Liu ZX, Dou WJ, Zhang R, Li YL, Xi XH, Xu JQ, Ma LT, Wang JJ. The roles of PDGFRα signaling in the postnatal development and functional maintenance of the SMC-ICC-PDGFRα+ cell (SIP) syncytium in the colon. Neurogastroenterol Motil 2019; 31:e13568. [PMID: 30848008 DOI: 10.1111/nmo.13568] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 01/15/2019] [Accepted: 01/15/2019] [Indexed: 01/02/2023]
Abstract
BACKGROUND The SIP syncytium in the gut consists of smooth muscle cells, interstitial cells of Cajal, and PDGFRα+ cells. We studied the fate of SIP cells after blocking PDGFRα receptor to explore the roles of PDGFRα signaling in the postnatal development and functional maintenance of the SIP syncytium. METHODS Crenolanib was administered to mice from P0, P10, or P50. The morphological changes in SIP cells were examined by immunofluorescence. Protein expression in SIP cells was detected by Western blotting. Moreover, colonic transit was analyzed by testing the colonic bead expulsion time. KEY RESULTS A dose of 5 mg(kg•day)-1 crenolanib administered for 10 days beginning on P0 apparently hindered the development of PDGFRα+ cells in the colonic longitudinal muscularis and myenteric plexus without influencing their proliferative activity and apoptosis, but this result was not seen in the colonic circular muscularis. SMCs were also inhibited by crenolanib. A dose of 7.5 mg(kg•day)-1 crenolanib administered for 15 days beginning on P0 caused reductions in both PDGFRα+ cells and ICC in the longitudinal muscularis, myenteric plexus, and circular muscularis. However, when crenolanib was administered at a dose of 5 mg(kg•day)-1 beginning on P10 or P50, it only noticeably decreased the number of PDGFRα+ cells in the colonic longitudinal muscularis. Crenolanib also caused PDGFRα+ cells to transdifferentiate into SMC in adult mice. Colonic transit was delayed after administration of crenolanib. CONCLUSIONS & INFERENCES Therefore, PDGFRα signaling is essential for the development and functional maintenance of the SIP cells, especially PDGFRα+ cells.
Collapse
Affiliation(s)
- Qiang Lin
- Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Ming Qin
- Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Shu-Guang Zhao
- Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhen-Xiong Liu
- Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Wei-Jia Dou
- Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Rong Zhang
- Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Yu-Long Li
- Department of Gastroenterology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Xiao-Hou Xi
- Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Jia-Qiao Xu
- Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Li-Tian Ma
- Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Jing-Jie Wang
- Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
13
|
Lin Y, Liang Z, He L, Yang M, Liu D, Gu HF, Liu H, Zhu Z, Zheng H, Li L, Yang G. Gut ghrelin regulates hepatic glucose production and insulin signaling via a gut-brain-liver pathway. Cell Commun Signal 2019; 17:8. [PMID: 30683114 PMCID: PMC6347823 DOI: 10.1186/s12964-019-0321-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 01/21/2019] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Ghrelin modulates many physiological processes. However, the effects of intestinal ghrelin on hepatic glucose production (HGP) are still unclear. The current study was to explore the roles of intestinal ghrelin on glucose homeostasis and insulin signaling in the liver. METHODS The system of intraduodenal infusion and intracerebral microinfusion into the nucleus of the solitary tract (NTS) in the normal chow-diet rats and pancreatic-euglycemic clamp procedure (PEC) combined with [3-3H] glucose as a tracer were used to analyze the effect of intestinal ghrelin. Intraduodenal co-infusion of ghrelin, tetracaine and Activated Protein Kinase (AMPK) activator (AICAR), or pharmacologic and molecular inhibitor of N-methyl-D-aspartate receptors within the dorsal vagal complex, or hepatic vagotomy in rats were used to explore the possible mechanism of the effect of intestinal ghrelin on HGP. RESULTS Our results demonstrated that gut infusion of ghrelin inhibited duodenal AMP-dependent protein kinase (AMPK) signal pathways, increased HGP and expression of gluconeogenic enzymes, and decreased insulin signaling in the liver of the rat. Intraduodenal co-infusion of ghrelin receptor antagonist [D-Lys3]-GHRP-6 and AMPK agonist with ghrelin diminished gut ghrelin-induced increase in HGP and decrease in glucose infusion rate (GIR) and hepatic insulin signaling. The effects of gut ghrelin were also negated by co-infusion with tetracaine, or MK801, an N-methyl-D-aspartate (NMDA) receptor inhibitor, and adenovirus expressing the shRNA of NR1 subunit of NMDA receptors (Ad-shNR1) within the dorsal vagal complex, and hepatic vagotomy in rats. When ghrelin and lipids were co-infused into the duodenum, the roles of gut lipids in increasing the rate of glucose infusion (GIR) and lowering HGP were reversed. CONCLUSIONS The current study provided evidence that intestinal ghrelin has an effect on HGP and identified a neural glucoregulatory function of gut ghrelin signaling.
Collapse
Affiliation(s)
- Yao Lin
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China.,The Key Laboratory of Laboratory Medical Diagnostics in the Ministry of Education and Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400010, China
| | - Zerong Liang
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China
| | - Liping He
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China
| | - Mengliu Yang
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China
| | - Dongfang Liu
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China
| | - Harvest F Gu
- Center for Pathophysiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Hua Liu
- Department of Pediatrics, University of Mississippi Medical Center, 2500 North State Street, Jackson, Mississippi, MS 39216-4505, USA
| | - Zhiming Zhu
- Department of Hypertension and Endocrinology, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, 400010, China
| | - Hongting Zheng
- Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Ling Li
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China.
| | - Gangyi Yang
- Department of Endocrinology, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China.
| |
Collapse
|
14
|
Choi E, Lantz TL, Vlacich G, Keeley TM, Samuelson LC, Coffey RJ, Goldenring JR, Powell AE. Lrig1+ gastric isthmal progenitor cells restore normal gastric lineage cells during damage recovery in adult mouse stomach. Gut 2018; 67:1595-1605. [PMID: 28814482 PMCID: PMC5815959 DOI: 10.1136/gutjnl-2017-313874] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 06/26/2017] [Accepted: 06/29/2017] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Lrig1 is a marker of proliferative and quiescent stem cells in the skin and intestine. We examined whether Lrig1-expressing cells are long-lived gastric progenitors in gastric glands in the mouse stomach. We also investigated how the Lrig1-expressing progenitor cells contribute to the regeneration of normal gastric mucosa by lineage commitment to parietal cells after acute gastric injury in mice. DESIGN We performed lineage labelling using Lrig1-CreERT2/+;R26R-YFP/+ (Lrig1/YFP) or R26R-LacZ/+ (Lrig1/LacZ) mice to examine whether the Lrig1-YFP-marked cells are gastric progenitor cells. We studied whether Lrig1-YFP-marked cells give rise to normal gastric lineage cells in damaged mucosa using Lrig1/YFP mice after treatment with DMP-777 to induce acute injury. We also studied Lrig1-CreERT2/CreERT2 (Lrig1 knockout) mice to examine whether the Lrig1 protein is required for regeneration of gastric corpus mucosa after acute injury. RESULTS Lrig1-YFP-marked cells give rise to gastric lineage epithelial cells both in the gastric corpus and antrum, in contrast to published results that Lgr5 only marks progenitor cells within the gastric antrum. Lrig1-YFP-marked cells contribute to replacement of damaged gastric oxyntic glands during the recovery phase after acute oxyntic atrophy in the gastric corpus. Lrig1 null mice recovered normally from acute gastric mucosal injury indicating that Lrig1 protein is not required for lineage differentiation. Lrig1+ isthmal progenitor cells did not contribute to transdifferentiating chief cell lineages after acute oxyntic atrophy. CONCLUSIONS Lrig1 marks gastric corpus epithelial progenitor cells capable of repopulating the damaged oxyntic mucosa by differentiating into normal gastric lineage cells in mouse stomach.
Collapse
Affiliation(s)
- Eunyoung Choi
- Nashville VA Medical Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Section of Surgical Sciences, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Epithelial Biology Center, Nashville, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Tyler L Lantz
- Department of Biology, Institute of Molecular Biology, University of Oregon, Oregon, USA
| | - Gregory Vlacich
- Department of Radiation Oncology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Theresa M Keeley
- Department of Molecular & Integrative Physiology, The University of Michigan, Michigan, USA
| | - Linda C Samuelson
- Department of Molecular & Integrative Physiology, The University of Michigan, Michigan, USA
| | - Robert J Coffey
- Nashville VA Medical Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Epithelial Biology Center, Nashville, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - James R Goldenring
- Nashville VA Medical Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Section of Surgical Sciences, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Epithelial Biology Center, Nashville, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Anne E Powell
- Department of Biology, Institute of Molecular Biology, University of Oregon, Oregon, USA
| |
Collapse
|
15
|
Ward SM. Hyperplasia of Interstitial Cells of Cajal Leads to Rapid Gastric Emptying in Diabetes. Gastroenterology 2017; 153:350-352. [PMID: 28655507 DOI: 10.1053/j.gastro.2017.06.039] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Affiliation(s)
- Sean M Ward
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, Nevada.
| |
Collapse
|
16
|
McKinley ET, Sui Y, Al-Kofahi Y, Millis BA, Tyska MJ, Roland JT, Santamaria-Pang A, Ohland CL, Jobin C, Franklin JL, Lau KS, Gerdes MJ, Coffey RJ. Optimized multiplex immunofluorescence single-cell analysis reveals tuft cell heterogeneity. JCI Insight 2017; 2:93487. [PMID: 28570279 DOI: 10.1172/jci.insight.93487] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 04/27/2017] [Indexed: 12/17/2022] Open
Abstract
Intestinal tuft cells are a rare, poorly understood cell type recently shown to be a critical mediator of type 2 immune response to helminth infection. Here, we present advances in segmentation algorithms and analytical tools for multiplex immunofluorescence (MxIF), a platform that enables iterative staining of over 60 antibodies on a single tissue section. These refinements have enabled a comprehensive analysis of tuft cell number, distribution, and protein expression profiles as a function of anatomical location and physiological perturbations. Based solely on DCLK1 immunoreactivity, tuft cell numbers were similar throughout the mouse small intestine and colon. However, multiple subsets of tuft cells were uncovered when protein coexpression signatures were examined, including two new intestinal tuft cell markers, Hopx and EGFR phosphotyrosine 1068. Furthermore, we identified dynamic changes in tuft cell number, composition, and protein expression associated with fasting and refeeding and after introduction of microbiota to germ-free mice. These studies provide a foundational framework for future studies of intestinal tuft cell regulation and demonstrate the utility of our improved MxIF computational methods and workflow for understanding cellular heterogeneity in complex tissues in normal and disease states.
Collapse
Affiliation(s)
- Eliot T McKinley
- Epithelial Biology Center and.,Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Yunxia Sui
- General Electric Global Research Center, Niskayuna, New York, USA
| | - Yousef Al-Kofahi
- General Electric Global Research Center, Niskayuna, New York, USA
| | - Bryan A Millis
- Department of Cell and Developmental Biology.,Cell Imaging Shared Resource, and
| | - Matthew J Tyska
- Epithelial Biology Center and.,Department of Cell and Developmental Biology
| | - Joseph T Roland
- Epithelial Biology Center and.,Department of Surgery, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | | | | | - Christian Jobin
- Department of Medicine.,Department of Infectious Diseases and Pathology, and.,Department of Anatomy and Cell Physiology, University of Florida, Gainesville, Florida, USA
| | - Jeffrey L Franklin
- Epithelial Biology Center and.,Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Cell and Developmental Biology.,Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Ken S Lau
- Epithelial Biology Center and.,Department of Cell and Developmental Biology
| | - Michael J Gerdes
- General Electric Global Research Center, Niskayuna, New York, USA
| | - Robert J Coffey
- Epithelial Biology Center and.,Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Department of Cell and Developmental Biology.,Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| |
Collapse
|
17
|
Yang YP, Ma H, Starchenko A, Huh WJ, Li W, Hickman FE, Zhang Q, Franklin JL, Mortlock DP, Fuhrmann S, Carter BD, Ihrie RA, Coffey RJ. A Chimeric Egfr Protein Reporter Mouse Reveals Egfr Localization and Trafficking In Vivo. Cell Rep 2017; 19:1257-1267. [PMID: 28494873 PMCID: PMC5517093 DOI: 10.1016/j.celrep.2017.04.048] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 03/31/2017] [Accepted: 04/17/2017] [Indexed: 01/06/2023] Open
Abstract
EGF receptor (EGFR) is a critical signaling node throughout life. However, it has not been possible to directly visualize endogenous Egfr in mice. Using CRISPR/Cas9 genome editing, we appended a fluorescent reporter to the C terminus of the Egfr. Homozygous reporter mice appear normal and EGFR signaling is intact in vitro and in vivo. We detect distinct patterns of Egfr expression in progenitor and differentiated compartments in embryonic and adult mice. Systemic delivery of EGF or amphiregulin results in markedly different patterns of Egfr internalization and trafficking in hepatocytes. In the normal intestine, Egfr localizes to the crypt rather than villus compartment, expression is higher in adjacent epithelium than in intestinal tumors, and following colonic injury expression appears in distinct cell populations in the stroma. This reporter, under control of its endogenous regulatory elements, enables in vivo monitoring of the dynamics of Egfr localization and trafficking in normal and disease states.
Collapse
Affiliation(s)
- Yu-Ping Yang
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Haiting Ma
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Alina Starchenko
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Won Jae Huh
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Wei Li
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - F Edward Hickman
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - Qin Zhang
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jeffrey L Franklin
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA; Department of Veterans Affairs Medical Center, Nashville, TN 37232, USA
| | - Douglas P Mortlock
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Sabine Fuhrmann
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA; Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Bruce D Carter
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - Rebecca A Ihrie
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA; Department of Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA; Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Robert J Coffey
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA; Department of Veterans Affairs Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
18
|
Neirinckx V, Hedman H, Niclou SP. Harnessing LRIG1-mediated inhibition of receptor tyrosine kinases for cancer therapy. Biochim Biophys Acta Rev Cancer 2017; 1868:109-116. [PMID: 28259645 DOI: 10.1016/j.bbcan.2017.02.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 02/27/2017] [Accepted: 02/28/2017] [Indexed: 02/07/2023]
Abstract
Leucine-rich repeats and immunoglobulin-like domains containing protein 1 (LRIG1) is an endogenous feedback regulator of receptor tyrosine kinases (RTKs) and was recently shown to inhibit growth of different types of malignancies. Additionally, this multifaceted RTK inhibitor was reported to be a tumor suppressor, a stem cell regulator, and a modulator of different cellular phenotypes. This mini-review provides a concise and up-to-date summary about the known functions of LRIG1 and its related family members, with a special emphasis on underlying molecular mechanisms and the opportunities for harnessing its therapeutic potential against cancer.
Collapse
Affiliation(s)
- Virginie Neirinckx
- NorLux Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, 1526, Luxembourg
| | - Hakan Hedman
- Oncology Research Laboratory, Department of Radiation Sciences, Umeå University, 90187 Umeå, Sweden
| | - Simone P Niclou
- NorLux Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, 1526, Luxembourg; K.G. Jebsen Brain Tumour Research Centre, Department of Biomedicine, University of Bergen, 5020 Bergen, Norway.
| |
Collapse
|
19
|
Zebrafish as a model for understanding enteric nervous system interactions in the developing intestinal tract. Methods Cell Biol 2016; 134:139-64. [PMID: 27312493 DOI: 10.1016/bs.mcb.2016.02.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The enteric nervous system (ENS) forms intimate connections with many other intestinal cell types, including immune cells and bacterial consortia resident in the intestinal lumen. In this review, we highlight contributions of the zebrafish model to understanding interactions among these cells. Zebrafish is a powerful model for forward genetic screens, several of which have uncovered genes previously unknown to be important for ENS development. More recently, zebrafish has emerged as a model for testing functions of genes identified in human patients or large-scale human susceptibility screens. In several cases, zebrafish studies have revealed mechanisms connecting intestinal symptoms with other, seemingly unrelated disease phenotypes. Importantly, chemical library screens in zebrafish have provided startling new insights into potential effects of common drugs on ENS development. A key feature of the zebrafish model is the ability to rear large numbers of animals germ free or in association with only specific bacterial species. Studies utilizing these approaches have demonstrated the importance of bacterial signals for normal intestinal development. These types of studies also show how luminal bacteria and the immune system can contribute to inflammatory processes that can feedback to influence ENS development. The excellent optical properties of zebrafish embryos and larvae, coupled with the ease of generating genetically marked cells of both the host and its resident bacteria, allow visualization of multiple intestinal cell types in living larvae and should promote a more in-depth understanding of intestinal cell interactions, especially interactions between other intestinal cell types and the ENS.
Collapse
|
20
|
Sanders KM. New Molecular Tools to Investigate the Development and Functions of Interstitial Cells of Cajal in the GI Tract. Gastroenterology 2015; 149:283-6. [PMID: 26116799 DOI: 10.1053/j.gastro.2015.06.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Affiliation(s)
- Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada.
| |
Collapse
|