1
|
Adkins-Threats M, Arimura S, Huang YZ, Divenko M, To S, Mao H, Zeng Y, Hwang JY, Burclaff JR, Jain S, Mills JC. Metabolic regulator ERRγ governs gastric stem cell differentiation into acid-secreting parietal cells. Cell Stem Cell 2024; 31:886-903.e8. [PMID: 38733994 PMCID: PMC11162331 DOI: 10.1016/j.stem.2024.04.016] [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: 06/06/2023] [Revised: 02/26/2024] [Accepted: 04/19/2024] [Indexed: 05/13/2024]
Abstract
Parietal cells (PCs) produce gastric acid to kill pathogens and aid digestion. Dysregulated PC census is common in disease, yet how PCs differentiate is unclear. Here, we identify the PC progenitors arising from isthmal stem cells, using mouse models and human gastric cells, and show that they preferentially express cell-metabolism regulator and orphan nuclear receptor Estrogen-related receptor gamma (Esrrg, encoding ERRγ). Esrrg expression facilitated the tracking of stepwise molecular, cellular, and ultrastructural stages of PC differentiation. EsrrgP2ACreERT2 lineage tracing revealed that Esrrg expression commits progenitors to differentiate into mature PCs. scRNA-seq indicated the earliest Esrrg+ PC progenitors preferentially express SMAD4 and SP1 transcriptional targets and the GTPases regulating acid-secretion signal transduction. As progenitors matured, ERRγ-dependent metabolic transcripts predominated. Organoid and mouse studies validated the requirement of ERRγ for PC differentiation. Our work chronicles stem cell differentiation along a single lineage in vivo and suggests ERRγ as a therapeutic target for PC-related disorders.
Collapse
Affiliation(s)
- Mahliyah Adkins-Threats
- Section of Gastroenterology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Division of Biomedical and Biological Sciences, Washington University, St. Louis, MO 63130, USA; Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Sumimasa Arimura
- Section of Gastroenterology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yang-Zhe Huang
- Section of Gastroenterology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Margarita Divenko
- Section of Gastroenterology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sarah To
- Section of Gastroenterology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Heather Mao
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yongji Zeng
- Section of Gastroenterology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jenie Y Hwang
- Section of Gastroenterology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pathology and Laboratory Medicine, University of Texas Health San Antonio, San Antonio, TX 78249, USA
| | - Joseph R Burclaff
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, NC 27599, USA
| | - Shilpa Jain
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jason C Mills
- Section of Gastroenterology, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
2
|
Adkins-Threats M, Huang YZ, Mills JC. Highlights of how single-cell analyses are illuminating differentiation and disease in the gastric corpus. Am J Physiol Gastrointest Liver Physiol 2024; 326:G205-G215. [PMID: 38193187 PMCID: PMC11211037 DOI: 10.1152/ajpgi.00164.2023] [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: 08/02/2023] [Revised: 12/18/2023] [Accepted: 12/23/2023] [Indexed: 01/10/2024]
Abstract
Single-cell RNA-sequencing (scRNA-seq) has emerged as a powerful technique to identify novel cell markers, developmental trajectories, and transcriptional changes during cell differentiation and disease onset and progression. In this review, we highlight recent scRNA-seq studies of the gastric corpus in both human and murine systems that have provided insight into gastric organogenesis, identified novel markers for the various gastric lineages during development and in adults, and revealed transcriptional changes during regeneration and tumorigenesis. Overall, by elucidating transcriptional states and fluctuations at the cellular level in healthy and disease contexts, scRNA-seq may lead to better, more personalized clinical treatments for disease progression.
Collapse
Affiliation(s)
- Mahliyah Adkins-Threats
- Section of Gastroenterology, Department of Medicine, Baylor College of Medicine, Houston, Texas, United States
| | - Yang-Zhe Huang
- Section of Gastroenterology, Department of Medicine, Baylor College of Medicine, Houston, Texas, United States
- Graduate School of Biomedical Sciences, Cancer and Cell Biology Program, Baylor College of Medicine, Houston, Texas, United States
| | - Jason C Mills
- Section of Gastroenterology, Department of Medicine, Baylor College of Medicine, Houston, Texas, United States
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, United States
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States
| |
Collapse
|
3
|
Sirajudeen S, Shah I, Karam SM, Al Menhali A. Seven-Month Vitamin D Deficiency Inhibits Gastric Epithelial Cell Proliferation, Stimulates Acid Secretion, and Differentially Alters Cell Lineages in the Gastric Glands. Nutrients 2023; 15:4648. [PMID: 37960302 PMCID: PMC10649607 DOI: 10.3390/nu15214648] [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: 09/21/2023] [Revised: 10/26/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
Vitamin D (VD) deficiency can result from insufficiency of either light exposure or VD intake. We investigated the biological effects of VD deficiency for 7 months on the mouse gastric glands. Varying degrees of VD deficiency were induced in C57BL/6 mice by keeping them on standard diet with constant-dark conditions (SDD) or VD deficient diet with constant-dark conditions (VDD). Samples of serum, glandular stomach, and gastric contents were collected for LCMS/MS, RT-PCR, immunohistochemistry, and acid content measurements. Both SDD and VDD mice had a significant decline in 25OHVD metabolite, gastric epithelial cell proliferation, and mucin 6 gene expression. These effects were enhanced with the severity of VD deficiency from SDD to VDD. Besides and compared to the control group, SDD mice only displayed a significant increase in the number of zymogenic cells (p ≤ 0.0001) and high expression of the adiponectin (p ≤ 0.05), gastrin (p ≤ 0.0001), mucin 5AC (*** p ≤ 0.001) and the Cyclin-dependent kinase inhibitor 1A (**** p ≤ 0.0001). These phenotypes were unique to SDD gastric samples and not seen in the VDD or control groups. This study suggests that the body reacts differently to diverse VD deficiency sources, light or diet.
Collapse
Affiliation(s)
- Shaima Sirajudeen
- Department of Biology, College of Science, United Arab Emirates University (UAEU), Al Ain 15551, United Arab Emirates;
| | - Iltaf Shah
- Zayed bin Sultan Al Nahyan Center for Health Sciences, United Arab Emirates University (UAEU), Al Ain 15551, United Arab Emirates; (I.S.); (S.M.K.)
- Department of Chemistry, College of Science, United Arab Emirates University (UAEU), Al Ain 15551, United Arab Emirates
| | - Sherif M. Karam
- Zayed bin Sultan Al Nahyan Center for Health Sciences, United Arab Emirates University (UAEU), Al Ain 15551, United Arab Emirates; (I.S.); (S.M.K.)
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University (UAEU), Al Ain 15551, United Arab Emirates
| | - Asma Al Menhali
- Department of Biology, College of Science, United Arab Emirates University (UAEU), Al Ain 15551, United Arab Emirates;
- Zayed bin Sultan Al Nahyan Center for Health Sciences, United Arab Emirates University (UAEU), Al Ain 15551, United Arab Emirates; (I.S.); (S.M.K.)
| |
Collapse
|
4
|
Innate immune activation and modulatory factors of Helicobacter pylori towards phagocytic and nonphagocytic cells. Curr Opin Immunol 2023; 82:102301. [PMID: 36933362 DOI: 10.1016/j.coi.2023.102301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 02/17/2023] [Indexed: 03/18/2023]
Abstract
Helicobacter pylori is an intriguing obligate host-associated human pathogen with a specific host interaction biology, which has been shaped by thousands of years of host-pathogen coevolution. Molecular mechanisms of interaction of H. pylori with the local immune cells in the human system are less well defined than epithelial cell interactions, although various myeloid cells, including neutrophils and other phagocytes, are locally present or attracted to the sites of infection and interact with H. pylori. We have recently addressed the question of novel bacterial innate immune stimuli, including bacterial cell envelope metabolites, that can activate and modulate cell responses via the H. pylori Cag type IV secretion system. This review article gives an overview of what is currently known about the interaction modes and mechanisms of H. pylori with diverse human cell types, with a focus on bacterial metabolites and cells of the myeloid lineage including phagocytic and antigen-presenting cells.
Collapse
|
5
|
Zhang ZS, Deng WY, Huang SL, Yang BF, Zhu FH, Jiang B, Wang SN, Wang YK. Clinicopathological characteristics of signet-ring cell carcinoma derived from gastric fovelar epithelium. J Dig Dis 2022; 23:396-403. [PMID: 36111615 PMCID: PMC9826366 DOI: 10.1111/1751-2980.13120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/28/2022] [Accepted: 08/10/2022] [Indexed: 01/11/2023]
Abstract
OBJECTIVE We aimed to investigate the immunophenotype, differential diagnosis, and clinicopathological characteristics of signet-ring cell carcinoma (SRCC) derived from gastric foveolar epithelium. METHODS Clinical characteristics, endoscopic findings, histopathological features, and follow-up data of seven cases of SRCC derived from gastric foveolar epithelium with small intramucosal lesions were analyzed. RESULTS Seven patients with a mean age of 38.3 years were diagnosed with SRCC derived from gastric foveolar epithelium and small intramucosal lesions, all of them were negative for CDH-1 germline mutation. The glands proliferated and expanded, and then morphologically transformed into signet-ring cells and formed clonal hyperplastic SRCC, which expanded laterally along the gastric foveolar cells to a length of 3-6 mm. Periodic acid Schiff staining was positive, while CK7 and MUC6 were negative, in all cases. Ki-67-positive cells ranged 37%-60%. During a follow-up period of 6-30 months, no patients experienced tumor recurrence or metastasis. CONCLUSIONS SRCC derived from gastric foveolar epithelium is originated from the proliferative region of the bottom of the gastric pit and gland neck. It is easily missed diagnosed or misdiagnosed as it grows laterally along the gastric foveolar cells. Biological behavior, genetics, and etiology of such SRCC, as well as the clinicopathological characteristics, need to be further studied.
Collapse
Affiliation(s)
- Zhi Shang Zhang
- Department of PathologyShenzhen Hospital of Southern Medical UniversityShenzhenGuangdong ProvinceChina
| | - Wei Yi Deng
- Department of PathologyShenzhen Hospital of Southern Medical UniversityShenzhenGuangdong ProvinceChina
| | - Si Lin Huang
- Department of PathologyShenzhen Hospital of Southern Medical UniversityShenzhenGuangdong ProvinceChina
| | - Bin Feng Yang
- Department of PathologyXinxiang Central HospitalXinxiangHenan ProvinceChina
| | - Fang Heng Zhu
- Department of PathologyXinxiang Central HospitalXinxiangHenan ProvinceChina
| | - Bo Jiang
- Department of PathologyThe 990th Hospital of the PLA Joint Logistics Support ForceZhumadianHenan ProvinceChina
| | - Su Nan Wang
- Shenzhen PolytechnicShenzhenGuangdong ProvinceChina
| | - Yang Kun Wang
- Department of PathologyForesea Life Insurance Guangzhou General HospitalGuangzhouGuangdong ProvinceChina
| |
Collapse
|
6
|
Hoffmann W. Self-Renewal and Cancers of the Gastric Epithelium: An Update and the Role of the Lectin TFF1 as an Antral Tumor Suppressor. Int J Mol Sci 2022; 23:ijms23105377. [PMID: 35628183 PMCID: PMC9141172 DOI: 10.3390/ijms23105377] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/09/2022] [Accepted: 05/10/2022] [Indexed: 11/16/2022] Open
Abstract
In 2020, gastric cancer was the fourth leading cause of cancer deaths globally. About 90% of gastric cancers are sporadic and the vast majority are correlated with Helicobacter pylori infection; whereas familial clustering is observed in about 10% of cases. Gastric cancer is now considered to be a disease originating from dysregulated self-renewal of the gastric glands in the setting of an inflammatory environment. The human stomach contains two types of gastric units, which show bi-directional self-renewal from a complex variety of stem cells. This review focuses on recent progress concerning the characterization of the different stem cell populations and the mainly mesenchymal signals triggering their stepwise differentiation as well as the genesis of pre-cancerous lesions and carcinogenesis. Furthermore, a model is presented (Lectin-triggered Receptor Blocking Hypothesis) explaining the role of the lectin TFF1 as an antral tumor suppressor possibly regulating Lgr5+ antral stem cells in a paracrine or maybe autocrine fashion, with neighboring antral gland cells having a role as niche cells.
Collapse
Affiliation(s)
- Werner Hoffmann
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany
| |
Collapse
|
7
|
Ibrahim NA, Elmorshedy KE, Radwan DA, Buabeid MA. The impact of oral ciprofloxacin on the structure and functions of rat gastric mucosa. Saudi J Biol Sci 2022; 29:2187-2198. [PMID: 35531231 PMCID: PMC9073028 DOI: 10.1016/j.sjbs.2021.11.042] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/07/2021] [Accepted: 11/17/2021] [Indexed: 11/27/2022] Open
Abstract
Ciprofloxacin (CPX), is a fluoroquinolone antibiotic used to treat a number of gram-negative and gram-positive bacterial infections. Ciprofloxacin can cause severe side effects, ranging from tendon problems, nerve damage, to serious mood or behavior changes. The purpose of this study was to investigate how ciprofloxacin affects gastric cell lines in rats with a distinctive emphasis on physiological, histopathological, and bacteriological changes. Male albino rats (n = 21) were distributed into three groups; control, CPX, and CPX-withdrawal groups. The treated rats were given CPX tablets (12.5 mg/kg) dissolved in carboxymethyl cellulose (CMC) 0.5% orally once daily via gavage for sixty consecutive days. Control rats received only the vehicle. The withdrawal group was treated for 60 days and the drug was withdrawn for another sixty days. After completion of the experiment, all rats were sacrificed and gastric tissues were treated for light, immunohistochemical, and scanning electron microscopic examination. Image J software was used to measure immune-labeled gastric epithelial cells. Blood samples were also collected for H. Pylori immunoglobulins IgM, IgA, and IgG. Results showed that treated rats acquired significantly strongly positive tumor necrosis factor (TNFα) and significant reduction of serum level of H. pylori IgM, IgA, and IgG in all the study groups. It could be concluded that prolonged oral CPX administration to albino rats changes the gastric mucosal architecture and bacteriology.
Collapse
Affiliation(s)
- Nihal A Ibrahim
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, United Arab Emirates.,Centre of Medical and Bio-allied Health Sciences Research (CMBAHSR), Ajman University, Ajman, United Arab Emirates
| | - Kadreya E Elmorshedy
- Anatomy Department, Tanta College of Medicine, Egypt.,Almaakal University, Basra, Iraq
| | - Doaa A Radwan
- Anatomy Department, Tanta College of Medicine, Egypt
| | - Manal A Buabeid
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, United Arab Emirates.,Centre of Medical and Bio-allied Health Sciences Research (CMBAHSR), Ajman University, Ajman, United Arab Emirates
| |
Collapse
|
8
|
Sato Y, Ban S, Katayama Y, Mitsui T. Unique membranous gastrin receptor expression of parietal cells, and its distribution pattern in the gastric oxyntic mucosa and fundic gland polyps. Hum Pathol 2022; 125:23-34. [DOI: 10.1016/j.humpath.2022.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 04/03/2022] [Accepted: 04/04/2022] [Indexed: 11/04/2022]
|
9
|
Yang H, Yang WJ, Hu B. Gastric epithelial histology and precancerous conditions. World J Gastrointest Oncol 2022; 14:396-412. [PMID: 35317321 PMCID: PMC8919001 DOI: 10.4251/wjgo.v14.i2.396] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/08/2021] [Accepted: 01/05/2022] [Indexed: 02/06/2023] Open
Abstract
The most common histological type of gastric cancer (GC) is gastric adenocarcinoma arising from the gastric epithelium. Less common variants include mesenchymal, lymphoproliferative and neuroendocrine neoplasms. The Lauren scheme classifies GC into intestinal type, diffuse type and mixed type. The WHO classification includes papillary, tubular, mucinous, poorly cohesive and mixed GC. Chronic atrophic gastritis (CAG) and intestinal metaplasia are recommended as common precancerous conditions. No definite precancerous condition of diffuse/poorly/undifferentiated type is recommended. Chronic superficial inflammation and hyperplasia of foveolar cells may be the focus. Presently, the management of early GC and precancerous conditions mainly relies on endoscopy including diagnosis, treatment and surveillance. Management of precancerous conditions promotes the early detection and treatment of early GC, and even prevent the occurrence of GC. In the review, precancerous conditions including CAG, metaplasia, foveolar hyperplasia and gastric hyperplastic polyps derived from the gastric epithelium have been concluded, based on the overview of gastric epithelial histological organization and its renewal.
Collapse
Affiliation(s)
- Hang Yang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Wen-Juan Yang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Bing Hu
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
10
|
Al-Yassir F, Khoder G, Sugathan S, Saseedharan P, Al Menhali A, Karam SM. Modulation of Stem Cell Progeny by Probiotics during Regeneration of Gastric Mucosal Erosions. BIOLOGY 2021; 10:biology10070596. [PMID: 34203400 PMCID: PMC8301058 DOI: 10.3390/biology10070596] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/23/2021] [Accepted: 06/25/2021] [Indexed: 02/07/2023]
Abstract
Patients with gastric mucosal erosions are predisposed to chronic gastritis, ulcer or even cancer. The repair of mucosal erosions involves several events including proliferation of gastric epithelial stem cells. The aim of this study was to investigate the effects of the probiotic mixture of De Simone Formulation on gastric epithelial stem cell lineages in mouse models of gastric mucosal erosions. Gastric erosions were induced by a single oral gavage of 80% ethanol containing 15 mg/mL acetylsalicylic acid (5 mL/kg) following a daily dose of probiotic mixture (5 mg/day/mouse) for 10 days. In another protocol, erosions were induced by a daily gavage of acetylsalicylic acid (400 mg/kg/day/mouse) for 5 days before or after daily administration of probiotic mixture for 5 days. Control mice received water gavage for 10 days. All mice were injected with bromodeoxyuridine two hours before sacrifice to label S-phase cells. The stomachs of all mice were processed for histological examination, lectin binding, and immunohistochemical analysis. The results reveal that mice that received probiotics before or after the induction of erosion showed a decrease in erosion index with an increase in gastric epithelial stem/progenitor cell proliferation and enhanced production of mucus, trefoil factors, and ghrelin by mucous and enteroendocrine cell lineages. These mice also showed restoration of the amount of H+,K+-ATPase and pepsinogen involved in the production of the harsh acidic environment by parietal and chief cell lineages. In conclusion, this study demonstrates the beneficial effects of probiotics against gastric mucosal erosion and highlights the involvement and modulation of proliferative stem cells and their multiple glandular epithelial cell lineages.
Collapse
Affiliation(s)
- Farah Al-Yassir
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, United Arab Emirates; (F.A.-Y.); (S.S.); (P.S.)
- Department of Biological Sciences, Faculty of Science, Debbieh Campus, Beirut Arab University, P.O. Box 11-50-20 Riad El Solh 11072809, Beirut, Lebanon
| | - Ghalia Khoder
- Department of Pharmaceutics and Pharmaceutical Technology, Sharjah Institute for Medical Research, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
- Correspondence: (G.K.); (A.A.M.); (S.M.K.); Tel.: +971-3-713-7493 (S.M.K.)
| | - Subi Sugathan
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, United Arab Emirates; (F.A.-Y.); (S.S.); (P.S.)
| | - Prashanth Saseedharan
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, United Arab Emirates; (F.A.-Y.); (S.S.); (P.S.)
| | - Asma Al Menhali
- Department of Biology, College of Science, United Arab Emirates University, Al Ain 15551, United Arab Emirates
- Zayed Research Center for Health Sciences, United Arab Emirates University, Al Ain 17666, United Arab Emirates
- Correspondence: (G.K.); (A.A.M.); (S.M.K.); Tel.: +971-3-713-7493 (S.M.K.)
| | - Sherif M. Karam
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, United Arab Emirates; (F.A.-Y.); (S.S.); (P.S.)
- Zayed Research Center for Health Sciences, United Arab Emirates University, Al Ain 17666, United Arab Emirates
- Correspondence: (G.K.); (A.A.M.); (S.M.K.); Tel.: +971-3-713-7493 (S.M.K.)
| |
Collapse
|
11
|
Ribeiro-Parenti L, Jarry AC, Cavin JB, Willemetz A, Le Beyec J, Sannier A, Benadda S, Pelletier AL, Hourseau M, Léger T, Morlet B, Couvelard A, Anini Y, Msika S, Marmuse JP, Ledoux S, Le Gall M, Bado A. Bariatric surgery induces a new gastric mucosa phenotype with increased functional glucagon-like peptide-1 expressing cells. Nat Commun 2021; 12:110. [PMID: 33397977 PMCID: PMC7782689 DOI: 10.1038/s41467-020-20301-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 11/19/2020] [Indexed: 12/19/2022] Open
Abstract
Glucagon-Like Peptide-1 (GLP-1) undergoes rapid inactivation by dipeptidyl peptidase-4 (DPP4) suggesting that target receptors may be activated by locally produced GLP-1. Here we describe GLP-1 positive cells in the rat and human stomach and found these cells co-expressing ghrelin or somatostatin and able to secrete active GLP-1 in the rats. In lean rats, a gastric load of glucose induces a rapid and parallel rise in GLP-1 levels in both the gastric and the portal veins. This rise in portal GLP-1 levels was abrogated in HFD obese rats but restored after vertical sleeve gastrectomy (VSG) surgery. Finally, obese rats and individuals operated on Roux-en-Y gastric bypass and SG display a new gastric mucosa phenotype with hyperplasia of the mucus neck cells concomitant with increased density of GLP-1 positive cells. This report brings to light the contribution of gastric GLP-1 expressing cells that undergo plasticity changes after bariatric surgeries, to circulating GLP-1 levels.
Collapse
Affiliation(s)
- Lara Ribeiro-Parenti
- Université de Paris, Inserm U1149, Centre de Recherche sur l'inflammation, Paris, France
- Service de Chirurgie Générale Œsogastrique et Bariatrique, Hôpital Bichat - Claude-Bernard, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Anne-Charlotte Jarry
- Université de Paris, Inserm U1149, Centre de Recherche sur l'inflammation, Paris, France
| | - Jean-Baptiste Cavin
- Université de Paris, Inserm U1149, Centre de Recherche sur l'inflammation, Paris, France
| | - Alexandra Willemetz
- Université de Paris, Inserm U1149, Centre de Recherche sur l'inflammation, Paris, France
| | - Johanne Le Beyec
- Université de Paris, Inserm U1149, Centre de Recherche sur l'inflammation, Paris, France
- Sorbonne Université, AP-HP, Hôpital Pitié-Salpêtrière-Charles Foix, Biochimie Endocrinienne et Oncologique, Paris, France
| | - Aurélie Sannier
- Department of Pathology Bichat Hospital, AP-HP, 75018, Paris, France
| | - Samira Benadda
- Université de Paris, Inserm U1149, Centre de Recherche sur l'inflammation, Paris, France
- Cell and Tissue Imaging Platform, Inserm, U1149, CNRS, ERL8252, 75018, Paris, France
| | - Anne-Laure Pelletier
- Université de Paris, Inserm U1149, Centre de Recherche sur l'inflammation, Paris, France
| | - Muriel Hourseau
- Department of Pathology Bichat Hospital, AP-HP, 75018, Paris, France
| | - Thibaut Léger
- Université de Paris, Mass Spectrometry Laboratory, Institut Jacques Monod, UMR 7592, CNRS, 75205, Paris, France
- Université Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, 35000, Rennes, France
| | - Bastien Morlet
- Université de Paris, Mass Spectrometry Laboratory, Institut Jacques Monod, UMR 7592, CNRS, 75205, Paris, France
| | - Anne Couvelard
- Université de Paris, Inserm U1149, Centre de Recherche sur l'inflammation, Paris, France
- Department of Pathology Bichat Hospital, AP-HP, 75018, Paris, France
| | - Younes Anini
- Department of Obstetrics and Gynecology, Dalhousie University, IWK Health Centre, Halifax, New Brunswick, Canada
| | - Simon Msika
- Université de Paris, Inserm U1149, Centre de Recherche sur l'inflammation, Paris, France
- Service de Chirurgie Générale Œsogastrique et Bariatrique, Hôpital Bichat - Claude-Bernard, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Jean-Pierre Marmuse
- Université de Paris, Inserm U1149, Centre de Recherche sur l'inflammation, Paris, France
- Service de Chirurgie Générale Œsogastrique et Bariatrique, Hôpital Bichat - Claude-Bernard, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Sévérine Ledoux
- Université de Paris, Inserm U1149, Centre de Recherche sur l'inflammation, Paris, France
- Service des Explorations Fonctionnelles Hôpital Louis Mourier, AP-HP, Centre Intégré Nord Francilien de prise en charge de l'Obésité (CINFO), 92701, Colombes, France
| | - Maude Le Gall
- Université de Paris, Inserm U1149, Centre de Recherche sur l'inflammation, Paris, France.
| | - André Bado
- Université de Paris, Inserm U1149, Centre de Recherche sur l'inflammation, Paris, France.
| |
Collapse
|
12
|
Hu MN, Hu SH, Zhang XW, Xiong SM, Deng H. Overview on new progress of hereditary diffuse gastric cancer with CDH1 variants. TUMORI JOURNAL 2020; 106:346-355. [PMID: 32811340 DOI: 10.1177/0300891620949668] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Hereditary diffuse gastric cancer (HDGC), comprising 1%-3% of gastric malignances, has been associated with CDH1 variants. Accumulating evidence has demonstrated more than 100 germline CDH1 variant types. E-cadherin encoded by the CDH1 gene serves as a tumor suppressor protein. CDH1 promoter hypermethylation and other molecular mechanisms resulting in E-cadherin dysfunction are involved in the tumorigenesis of HDGC. Histopathology exhibits characteristic signet ring cells, and immunohistochemical staining may show negativity for E-cadherin and other signaling proteins. Early HDGC is difficult to detect by endoscopy due to the development of lesions beneath the mucosa. Prophylactic gastrectomy is the most recommended treatment for pathogenic CDH1 variant carriers. Recent studies have promoted the progression of promising molecular-targeted therapies and management strategies. This review summarizes recent advances in CDH1 variant types, tumorigenesis mechanisms, diagnosis, and therapy, as well as clinical implications for future gene therapies.
Collapse
Affiliation(s)
- Mu-Ni Hu
- Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Shu-Hui Hu
- Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Xing-Wei Zhang
- Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Shu-Min Xiong
- Department of Ophthalmology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Huan Deng
- Molecular Medicine and Genetics Center, the Fourth Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China.,Renmin Institute of Forensic Medicine in Jiangxi, Nanchang, Jiangxi Province, China
| |
Collapse
|
13
|
Cao Y, Zeng Y, Qin X, Tan Y, Zeng M, Wang L, Cao X, Zou L, Wang C. A rare case report of pituicytoma with biphasic pattern and admixed with scattered Herring bodies. World J Surg Oncol 2020; 18:108. [PMID: 32460843 PMCID: PMC7254636 DOI: 10.1186/s12957-020-01889-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 05/19/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Pituicytoma is a rare pituitary non-neuroendocrine tumour. The awareness of pituitary non-neuroendocrine tumours has gradually increased over the past several decades, but the knowledge of some histological variants of the tumours is limited, particularly in clinicopathological significance. Here, we report a rare case of pituicytoma variant. CASE PRESENTATION A 71-year-old man presented with sudden symptoms of stroke including urinary incontinence, weakness in right lower limb, and trouble speaking. Physical examinations showed a right facial paralysis. The radiological examinations eventually found a 1.7 × 1.4 × 1.3 cm sellar occupied lesion. After symptomatic treatment improved the symptoms, the patient underwent transsphenoidal resection of the pituitary mass. Histologically, the tumour contained hypocellular area and hypercellular area. The hypocellular area showed elongated spindle cells arranged in a fascicular pattern around small vessels and scattered Herring bodies; the hypercellular area showed a large number of pseudorosettes. Immunohistochemistrically, the tumour cells were positive for thyroid transcription factor-1, S100, and neuron-specific enolase. Neurofilament only showed a little positive in the hypocellular area, and silver impregnation was only noted in a perivascular distribution. The patient had no recurrence 4 months after the surgery. CONCLUSIONS The rare variant of pituicytoma has a favourable prognosis. Moreover, it needs to be distinguished pituicytomas with pseudorosettes from ependymomas because of different prognosis. Lastly, Herring bodies may occasionally be seen in the pituicytoma, which could be a potential diagnostic pitfall.
Collapse
Affiliation(s)
- Youde Cao
- Department of Pathology, Chongqing Hospital of Traditional Chinese Medicine, 6 Seventh Panxi Branch Road, Jiangbei District, Chongqing, 400021, China.,Department of Pathology, College of Basic Medicine, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong, Chongqing, 400016, China
| | - Yan Zeng
- Department of Pathology, College of Basic Medicine, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong, Chongqing, 400016, China
| | - Xue Qin
- Department of Pathology, College of Basic Medicine, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong, Chongqing, 400016, China
| | - Yiwen Tan
- Department of Pathology, College of Basic Medicine, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong, Chongqing, 400016, China
| | - Min Zeng
- Department of Pathology, Chongqing Hospital of Traditional Chinese Medicine, 6 Seventh Panxi Branch Road, Jiangbei District, Chongqing, 400021, China
| | - Lijuan Wang
- Department of Pathology, Chongqing Hospital of Traditional Chinese Medicine, 6 Seventh Panxi Branch Road, Jiangbei District, Chongqing, 400021, China
| | - Xiaojing Cao
- Department of Pathology, Chongqing Hospital of Traditional Chinese Medicine, 6 Seventh Panxi Branch Road, Jiangbei District, Chongqing, 400021, China
| | - Lingfeng Zou
- Department of Pathology, Chongqing Hospital of Traditional Chinese Medicine, 6 Seventh Panxi Branch Road, Jiangbei District, Chongqing, 400021, China
| | - Chenglong Wang
- Department of Pathology, Chongqing Hospital of Traditional Chinese Medicine, 6 Seventh Panxi Branch Road, Jiangbei District, Chongqing, 400021, China. .,Department of Pathology, College of Basic Medicine, Chongqing Medical University, 1 Yixueyuan Road, Yuzhong, Chongqing, 400016, China.
| |
Collapse
|
14
|
Tano de la Hoz MF, Longo MV, Vidal ME, Flamini MA, Díaz AO. The stomach of the wild herbivorous hystricomorph rodent
Lagostomus maximus
: A comparative histomorphological and histochemical study of the cardiac and pyloric regions. ACTA ZOOL-STOCKHOLM 2020. [DOI: 10.1111/azo.12335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- María Florencia Tano de la Hoz
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET Buenos Aires Argentina
- Departamento de Biología FCEyN IIMyC – Instituto de Investigaciones Marinas y Costeras CONICET‐Universidad Nacional de Mar del Plata Buenos Aires Argentina
| | - María Victoria Longo
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET Buenos Aires Argentina
- Departamento de Biología FCEyN IIMyC – Instituto de Investigaciones Marinas y Costeras CONICET‐Universidad Nacional de Mar del Plata Buenos Aires Argentina
| | - Mariana Elena Vidal
- Facultad de Ciencias Exactas y Naturales Universidad Nacional de Mar del Plata Buenos Aires Argentina
| | - Mirta Alicia Flamini
- Laboratorio de Histología y Embriología Descriptiva Experimental y Comparada Facultad de Ciencias Veterinarias Universidad Nacional de La Plata Buenos Aires Argentina
| | - Alcira Ofelia Díaz
- Departamento de Biología FCEyN IIMyC – Instituto de Investigaciones Marinas y Costeras CONICET‐Universidad Nacional de Mar del Plata Buenos Aires Argentina
| |
Collapse
|
15
|
Sheng W, Malagola E, Nienhüser H, Zhang Z, Kim W, Zamechek L, Sepulveda A, Hata M, Hayakawa Y, Zhao CM, Chen D, Wang TC. Hypergastrinemia Expands Gastric ECL Cells Through CCK2R + Progenitor Cells via ERK Activation. Cell Mol Gastroenterol Hepatol 2020; 10:434-449.e1. [PMID: 32330731 PMCID: PMC7371950 DOI: 10.1016/j.jcmgh.2020.04.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 04/11/2020] [Accepted: 04/13/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Enterochromaffin-like (ECL) cells in the stomach express gastrin/cholecystokinin 2 receptor CCK2R and are known to expand under hypergastrinemia, but whether this results from expansion of existing ECL cells or increased production from progenitors has not been clarified. METHODS We used mice with green fluorescent protein fluorescent reporter expression in ECL cells (histidine decarboxylase [Hdc]-green fluorescent protein), as well as Cck2r- and Hdc-driven Tamoxifen inducible recombinase Cre (Cck2r-CreERT2, Hdc-CreERT2) mice combined with Rosa26Sor-tdTomato (R26-tdTomato) mice, and studied their expression and cell fate in the gastric corpus by using models of hypergastrinemia (gastrin infusion, omeprazole treatment). RESULTS Hdc-GFP marked the majority of ECL cells, located in the lower third of the gastric glands. Hypergastrinemia led to expansion of ECL cells that was not restricted to the gland base, and promoted cellular proliferation (Ki67) in the gastric isthmus but not in basal ECL cells. Cck2r-CreERT2 mice marked most ECL cells, as well as scattered cell types located higher up in the glands, whose number was increased during hypergastrinemia. Cck2r-CreERT2+ isthmus progenitors, but not Hdc+ mature ECL cells, were the source of ECL cell hyperplasia during hypergastrinemia and could grow as 3-dimensional spheroids in vitro. Moreover, gastrin treatment in vitro promoted sphere formation from sorted Cck2r+Hdc- cells, and increased chromogranin A and phosphorylated- extracellular signal-regulated kinase expression in CCK2R-derived organoids. Gastrin activates extracellular signal-regulated kinase pathways in vivo and in vitro, and treatment with the Mitogen-activated protein kinase kinase 1 inhibitor U0126 blocked hypergastrinemia-mediated changes, including CCK2R-derived ECL cell hyperplasia in vivo as well as sphere formation and chromogranin A expression in vitro. CONCLUSIONS We show here that hypergastrinemia induces ECL cell hyperplasia that is derived primarily from CCK2R+ progenitors in the corpus. Gastrin-dependent function of CCK2R+ progenitors is regulated by the extracellular signal-regulated kinase pathway.
Collapse
Affiliation(s)
- Weiwei Sheng
- Division of Digestive and Liver Diseases, Department of Medicine,Department of Gastrointestinal Surgery, the First Hospital, China Medical University, Shenyang, China
| | - Ermanno Malagola
- Division of Digestive and Liver Diseases, Department of Medicine
| | - Henrik Nienhüser
- Division of Digestive and Liver Diseases, Department of Medicine
| | - Zhengyu Zhang
- Division of Digestive and Liver Diseases, Department of Medicine
| | - Woosook Kim
- Division of Digestive and Liver Diseases, Department of Medicine
| | - Leah Zamechek
- Division of Digestive and Liver Diseases, Department of Medicine
| | - Antonia Sepulveda
- Department of Pathology, Columbia University, College of Physicians and Surgeons, New York, New York
| | - Masahiro Hata
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoku Hayakawa
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Chun-Mei Zhao
- Department of Clinical and Molecular Medicine (Institutt for klinisk og molekylær medisin), Norwegian University of Science and Technology (Norges teknisk-naturvitenskaplige universitet), Trondheim, Norway
| | - Duan Chen
- Department of Clinical and Molecular Medicine (Institutt for klinisk og molekylær medisin), Norwegian University of Science and Technology (Norges teknisk-naturvitenskaplige universitet), Trondheim, Norway
| | - Timothy C. Wang
- Division of Digestive and Liver Diseases, Department of Medicine,Correspondence Address correspondence to: Timothy C. Wang, MD, Division of Digestive and Liver Disease, Department of Medicine, Columbia University Medical Center, New York, New York; fax: (212) 851-4590.
| |
Collapse
|
16
|
Khoder G, Al-Yassir F, Al Menhali A, Saseedharan P, Sugathan S, Tomasetto C, Karam SM. Probiotics Upregulate Trefoil Factors and Downregulate Pepsinogen in the Mouse Stomach. Int J Mol Sci 2019; 20:ijms20163901. [PMID: 31405107 PMCID: PMC6719917 DOI: 10.3390/ijms20163901] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 07/31/2019] [Accepted: 08/05/2019] [Indexed: 02/06/2023] Open
Abstract
Probiotics are used in the management of some gastrointestinal diseases. However, little is known about their effects on normal gastric epithelial biology. The aim of this study was to explore how the probiotic mixture VSL#3 affects gastric cell lineages in mice with a special focus on protective and aggressive factors. Weight-matching littermate male mice (n = 14) were divided into treated and control pairs. The treated mice received VSL#3 (5 mg/day/mouse) by gastric gavage for 10 days. Control mice received only the vehicle. Food consumption and bodyweight were monitored. All mice were injected intraperitoneally with bromodeoxyuridine (120 mg/Kg bodyweight) two hours before sacrificed to label S-phase cells. Stomach tissues were processed for lectin- and immunohistochemical examination. ImageJ software was used to quantify immunolabeled gastric epithelial cells. Real-time quantitative polymerase chain reaction was used to provide relative changes in expression of gastric cell lineages specific genes. Results revealed that treated mice acquired (i) increased production of mucus, trefoil factor (TFF) 1 and TFF2, (ii) decreased production of pepsinogen, and (iii) increased ghrelin-secreting cells. No significant changes were observed in bodyweight, food consumption, cell proliferation, or parietal cells. Therefore, VSL#3 administration amplifies specific cell types specialized in the protection of the gastric epithelium.
Collapse
Affiliation(s)
- Ghalia Khoder
- Department of Pharmaceutics and Pharmaceuticals Technology, College of Pharmacy, Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, UAE
| | - Farah Al-Yassir
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, AlAin 17666, UAE
- Department of Biological Sciences, Faculty of Science, Beirut Arab University, Debbieh Campus PO Box 11-50-20 Riad El Solh, Beirut 11072809, Lebanon
| | - Asma Al Menhali
- Department of Biology, College of Science, United Arab Emirates University, Al-Ain 15551, UAE
| | - Prashanth Saseedharan
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, AlAin 17666, UAE
| | - Subi Sugathan
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, AlAin 17666, UAE
| | - Catherine Tomasetto
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Centre National de la Recherche Scientifique (CNRS), UMR7104, Université de Strasbourg, F-67404 Illkirch, France
| | - Sherif M Karam
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, AlAin 17666, UAE.
| |
Collapse
|
17
|
Ogawa T, Wada Y, Takemura K, Board PG, Uchida K, Kitagaki K, Tamura T, Suzuki T, Tokairin Y, Nakajima Y, Eishi Y. CHAC1 overexpression in human gastric parietal cells with Helicobacter pylori infection in the secretory canaliculi. Helicobacter 2019; 24:e12598. [PMID: 31111570 PMCID: PMC6618068 DOI: 10.1111/hel.12598] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/29/2019] [Accepted: 04/29/2019] [Indexed: 12/30/2022]
Abstract
BACKGROUND Cation transport regulator 1 (CHAC1), a newly discovered enzyme that degrades glutathione, is induced in Helicobacter pylori (H. pylori)-infected gastric epithelial cells in culture. The CHAC1-induced decrease in glutathione leads to an accumulation of reactive oxygen species and somatic mutations in TP53. We evaluated the possible correlation between H. pylori infection and CHAC1 expression in human gastric mucosa. MATERIALS AND METHODS Both fresh-frozen and formalin-fixed paraffin-embedded tissue samples of gastric mucosa with or without H. pylori infection were obtained from 41 esophageal cancer patients that underwent esophago-gastrectomy. Fresh samples were used for real-time polymerase chain reaction for H. pylori DNA and CHAC1 mRNA, and formalin-fixed samples were used for immunohistochemistry with anti-CHAC1 and anti-H. pylori monoclonal antibodies. Double-enzyme or fluorescence immunohistochemistry and immuno-electron microscopy were used for further analysis. RESULTS Significant CHAC1 overexpression was detected in H. pylori-infected parietal cells that expressed the human proton pump/H,K-ATPase α subunit, whereas a constitutively low level of CHAC1 mRNA expression was observed in the other samples regardless of the H. pylori infection status, reflecting the weak CHAC1 expression detected by immunohistochemistry in the fundic-gland areas. Immuno-electron microscopy revealed intact H. pylori cells in the secretory canaliculi of infected parietal cells. Some parietal cells exhibited positive nuclear signals for Ki67 in the neck zone of the gastric fundic-gland mucosa with H. pylori infection. CONCLUSION Cation transport regulator 1 overexpression in H. pylori-infected parietal cells may cause the H. pylori-induced somatic mutations that contribute to the development of gastric cancer.
Collapse
Affiliation(s)
- Tomohisa Ogawa
- Department of Human Pathology, Graduate School and Faculty of MedicineTokyo Medical and Dental UniversityTokyoJapan
| | - Yuriko Wada
- Department of Human Pathology, Graduate School and Faculty of MedicineTokyo Medical and Dental UniversityTokyoJapan
| | - Kosuke Takemura
- Department of UrologyTokyo Metropolitan Cancer and Infectious Diseases Center Komagome HospitalTokyoJapan
| | - Philip G. Board
- The ACRF Department of Cancer Biology and Therapeutics, Group of Molecular GeneticsThe John Curtin School of Medical Research, Australian National UniversityCanberraAustralian Capital TerritoryAustralia
| | - Keisuke Uchida
- Division of Surgical PathologyTokyo Medical and Dental University HospitalTokyoJapan
| | - Keisuke Kitagaki
- Division of Surgical PathologyTokyo Medical and Dental University HospitalTokyoJapan
| | - Tomoki Tamura
- Division of Surgical PathologyTokyo Medical and Dental University HospitalTokyoJapan
| | - Takashige Suzuki
- Department of Human Pathology, Graduate School and Faculty of MedicineTokyo Medical and Dental UniversityTokyoJapan
| | - Yutaka Tokairin
- Department of Gastrointestinal SurgeryTokyo Medical and Dental UniversityTokyoJapan
| | - Yasuaki Nakajima
- Department of Gastrointestinal SurgeryTokyo Medical and Dental UniversityTokyoJapan
| | - Yoshinobu Eishi
- Department of Human Pathology, Graduate School and Faculty of MedicineTokyo Medical and Dental UniversityTokyoJapan
| |
Collapse
|
18
|
do Nascimento Santos CA, Borojevic R, Nasciutti LE, Maedatakiya CM. Characterization of Gastrospheres Using 3D Coculture System. Methods Mol Biol 2019; 1842:105-121. [PMID: 30196405 DOI: 10.1007/978-1-4939-8697-2_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/01/2023]
Abstract
To understand the molecular mechanisms involved in gastric disorders and regeneration, we need an in vitro tridimensional (3D) culture model, which can mimic the in vivo gastric microenvironment. A 3D coculture system named gastrosphere is proposed herein, composed of primary human gastric epithelial and stromal cells. The primary cultures were obtained from endoscopic gastric biopsies, and after mechanical and enzymatic dispersion, epithelial (HGE3) and stromal (HGS12) cells were expanded. After extensive immunocytochemical characterization, cells were seeded onto 96-well round bottom plates previously covered with 1% agarose. Cells were cultured in KM-F12 culture medium with 10% fetal bovine serum (FBS), antibiotics, and antimycotics, in humidified air at 37 °C and atmosphere containing 5% CO2 for 72 h or until spheres formation. Then gastrospheres were carefully transferred to a rotary cell culture system (RCCS-4), and maintained for 07, 14, 21, and 28 days. Gastrospheres were morphologically characterized by immunocytochemistry [cytokeratins (CK), vimentin, α-smooth muscle actin (α-SMA), laminin (LN), fibronectin (FN), and type IV collagen (CIV), proliferating cell nuclear antigen (PCNA)], and electron microscopy. In gastrospheres, the cytokeratin-positive epithelial cells were found in the outer layer, while vimentin-positive stromal cells were localized in the center of the gastrospheres. PCNA+ cells were mainly seen at the peripheral and in the intermediary region while nestin+ cells were also depicted in the latter zone. Scanning electron microscopy revealed groups of cohesive gastric cells at the periphery, while transmission electron microscopy demonstrated some differentiated mucous-like or zymogenic-like cells in the periphery and stromal structures located at the center of the 3D structures. Extracellular matrix was deposed between cells. Our data suggest that in vitro gastrospheres recapitulate the in vivo gastric microenvironment.
Collapse
Affiliation(s)
| | - Radovan Borojevic
- Centro de Medicina Regenerativa, Faculdade de Medicina de Petrópolis - FASE, Rio de Janeiro, Brazil
| | - Luiz Eurico Nasciutti
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Cidade Universitária, Rio de Janeiro, Brazil
| | - Christina M Maedatakiya
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Cidade Universitária, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
19
|
Isolation of Human Gastric Epithelial Cells from Gastric Surgical Tissue and Gastric Biopsies for Primary Culture. Methods Mol Biol 2019. [PMID: 29959708 DOI: 10.1007/978-1-4939-8600-2_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
Abstract
The epithelium of human gastric mucosa is involved in secretory and digestive functions, and distinct epithelial populations located in specific compartments of the gastric pit-gland units mediate these functions. We successfully developed a method for the isolation and growth of normal human gastric epithelial cells using biopsies or surgically resected tissues as the source of the cells. Gastric epithelial cell aggregates were released from the underlying tissues by gentle enzymatic digestion of collagenase type IV. Primary cultures were generated by seeding viable multicellular aggregates on plastic without a biological matrix. Characterized by immunostaining of cell-specific antigens, the cells were confirmed to be heterogenous gastric epithelial primary cultures containing mucus cells, neck cells, parietal cells, chief cells, and gastrin-secreting endocrine cells. This simple and convenient method will prove useful in the isolation of normal human gastric mucous epithelial cells for in vitro studies of gastric epithelial biology.
Collapse
|
20
|
Ni X, Tan Z, Ding C, Zhang C, Song L, Yang S, Liu M, Jia R, Zhao C, Song L, Liu W, Zhou Q, Gong T, Li X, Tai Y, Zhu W, Shi T, Wang Y, Xu J, Zhen B, Qin J. A region-resolved mucosa proteome of the human stomach. Nat Commun 2019; 10:39. [PMID: 30604760 PMCID: PMC6318339 DOI: 10.1038/s41467-018-07960-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 12/06/2018] [Indexed: 12/13/2022] Open
Abstract
The human gastric mucosa is the most active layer of the stomach wall, involved in food digestion, metabolic processes and gastric carcinogenesis. Anatomically, the human stomach is divided into seven regions, but the protein basis for cellular specialization is not well understood. Here we present a global analysis of protein profiles of 82 apparently normal mucosa samples obtained from living individuals by endoscopic stomach biopsy. We identify 6,258 high-confidence proteins and estimate the ranges of protein expression in the seven stomach regions, presenting a region-resolved proteome reference map of the near normal, human stomach. Furthermore, we measure mucosa protein profiles of tumor and tumor nearby tissues (TNT) from 58 gastric cancer patients, enabling comparisons between tumor, TNT, and normal tissue. These datasets provide a rich resource for the gastrointestinal tract research community to investigate the molecular basis for region-specific functions in mucosa physiology and pathology including gastric cancer. The human stomach is divided into seven anatomically distinct regions but their protein composition is largely unknown. Here, the authors present a region-resolved map of the healthy human stomach mucosa as well as mucosa proteomes of tumor and tumor nearby tissue from gastric cancer patients.
Collapse
Affiliation(s)
- Xiaotian Ni
- Department of Gastrointestinal Oncology, The Fifth Medical Center, General Hospital of PLA, Beijing, 100071, China.,State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (The PHOENIX Center, Beijing), Institute of lifeomics, Beijing, 102206, China.,Center for Bioinformatics, East China Normal University, Shanghai, 200241, China
| | - Zhaoli Tan
- Department of Gastrointestinal Oncology, The Fifth Medical Center, General Hospital of PLA, Beijing, 100071, China
| | - Chen Ding
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (The PHOENIX Center, Beijing), Institute of lifeomics, Beijing, 102206, China.,State Key Laboratory of Genetic Engineering, Human Phenome Institute, Institutes of Biomedical Sciences, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Chunchao Zhang
- Alkek Center for Molecular Discovery, Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Lan Song
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (The PHOENIX Center, Beijing), Institute of lifeomics, Beijing, 102206, China.,Department of Bioinformatics, College of Life Science, Hebei University, Baoding, 071002, China
| | - Shuai Yang
- Department of Gastrointestinal Oncology, The Fifth Medical Center, General Hospital of PLA, Beijing, 100071, China
| | - Mingwei Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (The PHOENIX Center, Beijing), Institute of lifeomics, Beijing, 102206, China
| | - Ru Jia
- Department of Gastrointestinal Oncology, The Fifth Medical Center, General Hospital of PLA, Beijing, 100071, China
| | - Chuanhua Zhao
- Department of Gastrointestinal Oncology, The Fifth Medical Center, General Hospital of PLA, Beijing, 100071, China
| | - Lei Song
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (The PHOENIX Center, Beijing), Institute of lifeomics, Beijing, 102206, China
| | - Wanlin Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (The PHOENIX Center, Beijing), Institute of lifeomics, Beijing, 102206, China
| | - Quan Zhou
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (The PHOENIX Center, Beijing), Institute of lifeomics, Beijing, 102206, China
| | - Tongqing Gong
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (The PHOENIX Center, Beijing), Institute of lifeomics, Beijing, 102206, China
| | - Xianju Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (The PHOENIX Center, Beijing), Institute of lifeomics, Beijing, 102206, China
| | - Yanhong Tai
- Department of Gastrointestinal Oncology, The Fifth Medical Center, General Hospital of PLA, Beijing, 100071, China
| | - Weimin Zhu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (The PHOENIX Center, Beijing), Institute of lifeomics, Beijing, 102206, China
| | - Tieliu Shi
- Center for Bioinformatics, East China Normal University, Shanghai, 200241, China
| | - Yi Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (The PHOENIX Center, Beijing), Institute of lifeomics, Beijing, 102206, China.,Alkek Center for Molecular Discovery, Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jianming Xu
- Department of Gastrointestinal Oncology, The Fifth Medical Center, General Hospital of PLA, Beijing, 100071, China.
| | - Bei Zhen
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (The PHOENIX Center, Beijing), Institute of lifeomics, Beijing, 102206, China.
| | - Jun Qin
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (The PHOENIX Center, Beijing), Institute of lifeomics, Beijing, 102206, China. .,State Key Laboratory of Genetic Engineering, Human Phenome Institute, Institutes of Biomedical Sciences, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200032, China. .,Alkek Center for Molecular Discovery, Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
21
|
Luo W, Fedda F, Lynch P, Tan D. CDH1 Gene and Hereditary Diffuse Gastric Cancer Syndrome: Molecular and Histological Alterations and Implications for Diagnosis And Treatment. Front Pharmacol 2018; 9:1421. [PMID: 30568591 PMCID: PMC6290068 DOI: 10.3389/fphar.2018.01421] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 11/19/2018] [Indexed: 12/12/2022] Open
Abstract
Gastric cancer, a group of common malignancies, results in the most cancer mortality worldwide after only lung and colorectal cancer. Although familial gastric cancers have long been recognized, it was not until recently that they were discovered to be associated with mutations of specific genes. Mutations of CDH1, the gene encoding E-cadherin, are the most common germline mutations detected in gastric cancer and underlie hereditary diffuse gastric cancer (HDGC) syndrome. All reported HDGCs are the pure diffuse type by Lauren classification and are associated with dismal prognosis once the tumor invades the submucosa. Because CDH1 germline mutations are inherited in an autosomal-dominant fashion and have high penetrance, the International Gastric Cancer Linkage Consortium (IGCLC) developed criteria to facilitate the screening of CDH1 mutation carriers; these criteria have been proven to have excellent sensitivity and specificity. Recent histologic studies suggest that HDGC progresses through several stages. Even when the tumor becomes "invasive" in lamina propria, it may stay indolent for a long time. However, the molecular mechanisms that induce the transitions between stages and determine the length of the indolent phase remain to be determined. Although the standard management for CDH1 mutation carriers is prophylactic total gastrectomy, many questions must be answered before the surgery can be done. These include the optimal surveillance strategy, the best strategy to choose surgical candidates, and the ideal time to perform surgery. In addition to increasing the risk of gastric cancer, CDH1 germline mutations also increase the risk of invasive lobular carcinoma of the breast, and possibly colorectal adenocarcinoma, and are associated with blepharocheilodontic syndrome (a congenital development disorder). However, the optimal management of these conditions is less established owing to insufficient data regarding the risk of cancer development. This review focuses on molecular and histological findings in HDGC, as opposed to sporadic diffuse gastric cancer, and their implications for the management of CDH1 mutation carriers and the diagnosis and treatment of HDGC. Other conditions associated with CDH1 germline mutations and future research directions are also discussed.
Collapse
Affiliation(s)
- Wenyi Luo
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Faysal Fedda
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Patrick Lynch
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Dongfeng Tan
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
22
|
Abstract
Chronic injury and inflammation in the esophagus can cause a change in cellular differentiation known as metaplasia. Most commonly, the differentiation changes manifest as Barrett's esophagus (BE), characterized by the normal stratified squamous epithelium converting into a cuboidal-columnar, glandular morphology. BE cells can phenotypically resemble specific normal cell types of the stomach or intestine, or they can have overlapping phenotypes in disorganized admixtures. The stomach can also undergo metaplasia characterized by aberrant gastric or intestinal differentiation patterns. In both organs, it has been argued that metaplasia may represent a recapitulation of the embryonic or juvenile gastrointestinal tract, as cells access a developmental progenitor genetic program that can help repair damaged tissue. Here, we review the normal development of esophagus and stomach, and describe how BE represents an intermixing of cells resembling gastric pseudopyloric (SPEM) and intestinal metaplasia. We discuss a cellular process recently termed "paligenosis" that governs how mature, differentiated cells can revert to a proliferating progenitor state in metaplasia. We discuss the "Cyclical Hit" theory in which paligenosis might be involved in the increased risk of metaplasia for progression to cancer. However, somatic mutations might occur in proliferative phases and then be warehoused upon redifferentiation. Through years of chronic injury and many rounds of paligenosis and dedifferentiation, eventually a cell with a mutation that prevents dedifferentiation may arise and clonally expand fueling stable metaplasia and potentially thereafter acquiring additional mutations and progressing to dysplasia and cancer.
Collapse
Affiliation(s)
- Ramon U Jin
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Jason C Mills
- Division of Gastroenterology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
23
|
Gómez-Santos L, Alonso E, Díaz-Flores L, Madrid JF, Sáez FJ. Transdifferentiation of mucous neck cells into chief cells in fundic gastric glands shown by GNA lectin histochemistry. Tissue Cell 2017; 49:746-750. [PMID: 29089090 DOI: 10.1016/j.tice.2017.10.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 10/16/2017] [Accepted: 10/25/2017] [Indexed: 12/13/2022]
Abstract
The epithelium of the gastric mucosa and its glands in the corpus of rat stomach contains mucous surface cells (MSCs), parietal cells, mucous neck cells (MNCs), zymogenic or chief cells (ZCs), several types of enteroendocrine cells, and intermediate cells with characteristics between MNCs and ZCs also called transitional or prezymogenic cells (pre-ZCs). The aim of our work was to analyze the expression of Mannose (Man) in the rat gastric glands by means of Galanthus nivalis lectin (GNA) histochemistry to identify the differences between MNC, pre-ZCs and ZCs and to establish the relationships between these cells. Most of the cytoplasm of MNCs was negative for GNA histochemistry. Intensity of GNA labeling in the gastric gland showed a graduation from pre-ZCs (weak labeling) to ZCs (moderate labeling). Labeling of ZCs was stronger at the perinuclear and apical cytoplasm. In the last years, strong evidence has been reported supporting that ZCs differentiate from MNCs. Our work also supports the origin of ZCs from MNCs, because the GNA labeling graduation might be due to oligosaccharides which are not expressed in MNCs, start to express in pre-ZCs and are more abundant in ZCs, indicating that differentiation from MNCs to ZCs is a process in which glycans with Man moieties are synthesized.
Collapse
Affiliation(s)
- Laura Gómez-Santos
- Department of Cell Biology and Histology, Training and Research Unit: Reproduction, Development, Aging and Cancer (TRU/UFI 11/44), University of the Basque Country UPV/EHU, Leioa, Vizcaya, Spain
| | - Edurne Alonso
- Department of Cell Biology and Histology, Training and Research Unit: Reproduction, Development, Aging and Cancer (TRU/UFI 11/44), University of the Basque Country UPV/EHU, Leioa, Vizcaya, Spain
| | - Lucio Díaz-Flores
- Department of Anatomy, Pathology, Histology and Radiology, University of La Laguna, Tenerife, Spain
| | - Juan Francisco Madrid
- Department of Cell Biology and Histology, Regional Campus of International Excellence "Campus Mare Nostrum", IMIB-Arrixaca, University of Murcia, Espinardo, Murcia, Spain
| | - Francisco José Sáez
- Department of Cell Biology and Histology, Training and Research Unit: Reproduction, Development, Aging and Cancer (TRU/UFI 11/44), University of the Basque Country UPV/EHU, Leioa, Vizcaya, Spain.
| |
Collapse
|
24
|
The cytoprotective protein clusterin is overexpressed in hypergastrinemic rodent models of oxyntic preneoplasia and promotes gastric cancer cell survival. PLoS One 2017; 12:e0184514. [PMID: 28902909 PMCID: PMC5597207 DOI: 10.1371/journal.pone.0184514] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 08/27/2017] [Indexed: 02/07/2023] Open
Abstract
The cytoprotective protein clusterin is often dysregulated during tumorigenesis, and in the stomach, upregulation of clusterin marks emergence of the oxyntic atrophy (loss of acid-producing parietal cells)-associated spasmolytic polypeptide-expressing metaplasia (SPEM). The hormone gastrin is important for normal function and maturation of the gastric oxyntic mucosa and hypergastrinemia might be involved in gastric carcinogenesis. Gastrin induces expression of clusterin in adenocarcinoma cells. In the present study, we examined the expression patterns and gastrin-mediated regulation of clusterin in gastric tissue from: humans; rats treated with proton pump (H+/K+-ATPase) inhibitors and/or a gastrin receptor (CCK2R) antagonist; H+/K+-ATPase β-subunit knockout (H/K-β KO) mice; and Mongolian gerbils infected with Helicobacter pylori and given a CCK2R antagonist. Biological function of secretory clusterin was studied in human gastric cancer cells. Clusterin was highly expressed in neuroendocrine cells in normal oxyntic mucosa of humans and rodents. In response to hypergastrinemia, expression of clusterin increased significantly and its localization shifted to basal groups of proliferative cells in the mucous neck cell-chief cell lineage in all animal models. That shift was partially inhibited by antagonizing the CCK2R in rats and gerbils. The oxyntic mucosa of H/K-β KO mice contained areas with clusterin-positive mucous cells resembling SPEM. In gastric adenocarcinomas, clusterin mRNA expression was higher in diffuse tumors containing signet ring cells compared with diffuse tumors without signet ring cells, and clusterin seemed to be secreted by tumor cells. In gastric cancer cell lines, gastrin increased secretion of clusterin, and both gastrin and secretory clusterin promoted survival after starvation- and chemotherapy-induced stress. Overall, our results indicate that clusterin is overexpressed in hypergastrinemic rodent models of oxyntic preneoplasia and stimulates gastric cancer cell survival.
Collapse
|
25
|
Aouacheria A, Baghdiguian S, Lamb HM, Huska JD, Pineda FJ, Hardwick JM. Connecting mitochondrial dynamics and life-or-death events via Bcl-2 family proteins. Neurochem Int 2017; 109:141-161. [PMID: 28461171 DOI: 10.1016/j.neuint.2017.04.009] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 04/17/2017] [Indexed: 12/12/2022]
Abstract
The morphology of a population of mitochondria is the result of several interacting dynamical phenomena, including fission, fusion, movement, elimination and biogenesis. Each of these phenomena is controlled by underlying molecular machinery, and when defective can cause disease. New understanding of the relationships between form and function of mitochondria in health and disease is beginning to be unraveled on several fronts. Studies in mammals and model organisms have revealed that mitochondrial morphology, dynamics and function appear to be subject to regulation by the same proteins that regulate apoptotic cell death. One protein family that influences mitochondrial dynamics in both healthy and dying cells is the Bcl-2 protein family. Connecting mitochondrial dynamics with life-death pathway forks may arise from the intersection of Bcl-2 family proteins with the proteins and lipids that determine mitochondrial shape and function. Bcl-2 family proteins also have multifaceted influences on cells and mitochondria, including calcium handling, autophagy and energetics, as well as the subcellular localization of mitochondrial organelles to neuronal synapses. The remarkable range of physical or functional interactions by Bcl-2 family proteins is challenging to assimilate into a cohesive understanding. Most of their effects may be distinct from their direct roles in apoptotic cell death and are particularly apparent in the nervous system. Dual roles in mitochondrial dynamics and cell death extend beyond BCL-2 family proteins. In this review, we discuss many processes that govern mitochondrial structure and function in health and disease, and how Bcl-2 family proteins integrate into some of these processes.
Collapse
Affiliation(s)
- Abdel Aouacheria
- Institute of Evolutionary Sciences of Montpellier (ISEM), CNRS UMR 5554, University of Montpellier, Place Eugène Bataillon, 34095 Montpellier, France
| | - Stephen Baghdiguian
- Institute of Evolutionary Sciences of Montpellier (ISEM), CNRS UMR 5554, University of Montpellier, Place Eugène Bataillon, 34095 Montpellier, France
| | - Heather M Lamb
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, 615 North Wolfe St., Baltimore, MD 21205, USA
| | - Jason D Huska
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, 615 North Wolfe St., Baltimore, MD 21205, USA
| | - Fernando J Pineda
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, 615 North Wolfe St., Baltimore, MD 21205, USA; Department of Biostatistics, Johns Hopkins University, Bloomberg School of Public Health, 615 North Wolfe St., Baltimore, MD 21205, USA
| | - J Marie Hardwick
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, 615 North Wolfe St., Baltimore, MD 21205, USA.
| |
Collapse
|
26
|
Gómez-Santos L, Alonso E, Díaz-Flores L, Madrid JF, Sáez FJ. Characterization by Lectin Histochemistry of Two Subpopulations of Parietal Cells in the Rat Gastric Glands. J Histochem Cytochem 2017; 65:261-272. [PMID: 28438092 DOI: 10.1369/0022155417694871] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Parietal cells undergo a differentiation process while they move from the isthmus toward the pits and the base region of the gastric gland. The aim of this work was to analyze the rat gastric glands by lectin histochemistry to show the glycans expressed by upper (young) and lower (old) parietal cells. We used lectins recognizing the most frequent sugar moieties in mammals. Each lectin was assayed alone and in combination with several deglycosylation pretreatments: (1) β-elimination, which removes O-linked oligosaccharides; (2) incubation with Peptide-N-glycosidase F, to remove N-linked glycans; (3) acid hydrolysis, which removes terminal sialic acid moieties; (4) methylation-saponification, to remove sulfate groups from sugar residues; and (5) glucose oxidase, a technique carried out with the lectin concanavalin A to convert glucose into gluconic acid. The lectins from Helix pomatia, Dolichos biflorus (DBA), Glycine max (soybean), Maclura pomifera, Arachis hypogaea (peanut), Bandeiraea simplicifolia (lectin I-B4), and Datura stramonium showed a different glycan expression in the parietal cells throughout the gastric gland. This difference supports that parietal cells undergo a maturation/degeneration process while the cells descend along the gland. The role of DBA as a marker of parietal cells previously reported should be taken with caution because these cells showed different reactivity for the lectin, ranging from negative to strong labeling.
Collapse
Affiliation(s)
- Laura Gómez-Santos
- Department of Cell Biology and Histology, Training and Research Unit: Reproduction, Development, Aging and Cancer (TRU/UFI 11/44), School of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Vizcaya, Spain (LG-S, EA, FJS)
| | - Edurne Alonso
- Department of Cell Biology and Histology, Training and Research Unit: Reproduction, Development, Aging and Cancer (TRU/UFI 11/44), School of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Vizcaya, Spain (LG-S, EA, FJS)
| | - Lucio Díaz-Flores
- Department of Anatomy, Pathology, Histology and Radiology, University of La Laguna, San Cristóbal de La Laguna, Tenerife, Spain (LD-F)
| | - Juan F Madrid
- Department of Cell Biology and Histology, Regional Campus of International Excellence "Campus Mare Nostrum," IMIB-Arrixaca, School of Medicine, University of Murcia, Espinardo, Murcia, Spain (JFM)
| | - Francisco J Sáez
- Department of Cell Biology and Histology, Training and Research Unit: Reproduction, Development, Aging and Cancer (TRU/UFI 11/44), School of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Vizcaya, Spain (LG-S, EA, FJS)
| |
Collapse
|
27
|
Garcia M, Chomel JC, Mustapha P, Tran CT, Garnier M, Paris I, Quellard N, Godet J, Cremniter J, Bennaceur-Griscelli A, Lecron JC, Turhan AG, Burucoa C, Bodet C. In vitro culture and phenotypic and molecular characterization of gastric stem cells from human stomach. Helicobacter 2017; 22. [PMID: 27592706 DOI: 10.1111/hel.12351] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND Human gastric mucosa shows continuous self-renewal via differentiation from stem cells that remain poorly characterized. METHODS We describe an original protocol for culture of gastric stem/progenitor cells from adult human stomach. The molecular characteristics of cells were studied using TaqMan low-density array and qRT-PCR analyses using the well-characterized H1 and H9 embryonic stem cells as reference. Epithelial progenitor cells were challenged with H. pylori to characterize their inflammatory response. RESULTS Resident gastric stem cells expressed specific molecular markers of embryonic stem cells (SOX2, NANOG, and OCT4), as well as others specific to adult stem cells, particularly LGR5 and CD44. We show that gastric stem cells spontaneously differentiate into epithelial progenitor cells that can be challenged with H. pylori. The epithelial progenitor response to H. pylori showed a cag pathogenicity island-dependent induction of matrix metalloproteinases 1 and 3, chemokine (CXCL1, CXCL5, CXCL8, CCL20) and interleukine 33 expression. CONCLUSION This study opens new outlooks for investigation of gastric stem cell biology and pathobiology as well as host-H. pylori interactions.
Collapse
Affiliation(s)
- Magali Garcia
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, Université de Poitiers, Poitiers, France.,Centre Hospitalier Universitaire de Poitiers, Poitiers, France
| | - Jean-Claude Chomel
- Centre Hospitalier Universitaire de Poitiers, Poitiers, France.,Inserm U935, Université de Poitiers, Poitiers, France
| | - Pascale Mustapha
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, Université de Poitiers, Poitiers, France
| | - Cong Tri Tran
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, Université de Poitiers, Poitiers, France.,Centre Hospitalier Universitaire de Poitiers, Poitiers, France
| | - Martine Garnier
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, Université de Poitiers, Poitiers, France.,Centre Hospitalier Universitaire de Poitiers, Poitiers, France
| | - Isabelle Paris
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, Université de Poitiers, Poitiers, France.,Centre Hospitalier Universitaire de Poitiers, Poitiers, France
| | | | - Julie Godet
- Centre Hospitalier Universitaire de Poitiers, Poitiers, France
| | - Julie Cremniter
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, Université de Poitiers, Poitiers, France.,Centre Hospitalier Universitaire de Poitiers, Poitiers, France
| | - Annelise Bennaceur-Griscelli
- Inserm U935, Université de Poitiers, Poitiers, France.,Inserm U935, Hôpitaux Universitaires Paris Sud, Hôpital Bicêtre & Paul Brousse, Université Paris-Sud, Villejuif, France
| | - Jean-Claude Lecron
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, Université de Poitiers, Poitiers, France.,Centre Hospitalier Universitaire de Poitiers, Poitiers, France
| | - Ali G Turhan
- Inserm U935, Université de Poitiers, Poitiers, France.,Inserm U935, Hôpitaux Universitaires Paris Sud, Hôpital Bicêtre & Paul Brousse, Université Paris-Sud, Villejuif, France
| | - Christophe Burucoa
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, Université de Poitiers, Poitiers, France.,Centre Hospitalier Universitaire de Poitiers, Poitiers, France
| | - Charles Bodet
- Laboratoire Inflammation, Tissus Epithéliaux et Cytokines, Université de Poitiers, Poitiers, France
| |
Collapse
|
28
|
IMI - Oral biopharmaceutics tools project - Evaluation of bottom-up PBPK prediction success part 2: An introduction to the simulation exercise and overview of results. Eur J Pharm Sci 2016; 96:610-625. [PMID: 27816631 DOI: 10.1016/j.ejps.2016.10.036] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 10/12/2016] [Accepted: 10/30/2016] [Indexed: 12/22/2022]
Abstract
Orally administered drugs are subject to a number of barriers impacting bioavailability (Foral), causing challenges during drug and formulation development. Physiologically-based pharmacokinetic (PBPK) modelling can help during drug and formulation development by providing quantitative predictions through a systems approach. The performance of three available PBPK software packages (GI-Sim, Simcyp®, and GastroPlus™) were evaluated by comparing simulated and observed pharmacokinetic (PK) parameters. Since the availability of input parameters was heterogeneous and highly variable, caution is required when interpreting the results of this exercise. Additionally, this prospective simulation exercise may not be representative of prospective modelling in industry, as API information was limited to sparse details. 43 active pharmaceutical ingredients (APIs) from the OrBiTo database were selected for the exercise. Over 4000 simulation output files were generated, representing over 2550 study arm-institution-software combinations and approximately 600 human clinical study arms simulated with overlap. 84% of the simulated study arms represented administration of immediate release formulations, 11% prolonged or delayed release, and 5% intravenous (i.v.). Higher percentages of i.v. predicted area under the curve (AUC) were within two-fold of observed (52.9%) compared to per oral (p.o.) (37.2%), however, Foral and relative AUC (Frel) between p.o. formulations and solutions were generally well predicted (64.7% and 75.0%). Predictive performance declined progressing from i.v. to solution and immediate release tablet, indicating the compounding error with each layer of complexity. Overall performance was comparable to previous large-scale evaluations. A general overprediction of AUC was observed with average fold error (AFE) of 1.56 over all simulations. AFE ranged from 0.0361 to 64.0 across the 43 APIs, with 25 showing overpredictions. Discrepancies between software packages were observed for a few APIs, the largest being 606, 171, and 81.7-fold differences in AFE between SimCYP and GI-Sim, however average performance was relatively consistent across the three software platforms.
Collapse
|
29
|
Roy SAB, Allaire JM, Ouellet C, Maloum-Rami F, Pomerleau V, Lemieux É, Babeu JP, Rousseau J, Paquet M, Garde-Granger P, Boudreau F, Perreault N. Loss of mesenchymal bone morphogenetic protein signaling leads to development of reactive stroma and initiation of the gastric neoplastic cascade. Sci Rep 2016; 6:32759. [PMID: 27609464 PMCID: PMC5016723 DOI: 10.1038/srep32759] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 08/02/2016] [Indexed: 02/07/2023] Open
Abstract
Bmps are morphogens involved in various gastric cellular functions. Studies in genetically-modified mice have shown that Bmp disruption in gastric epithelial and stromal cell compartments leads to the development of tumorigenesis. Our studies have demonstrated that abrogation of gastric epithelial Bmp signaling alone was not sufficient to recapitulate the neoplastic features associated with total gastric loss of Bmp signaling. Thus, epithelial Bmp signaling does not appear to be a key player in gastric tumorigenesis initiation. These observations suggest a greater role for stromal Bmp signaling in gastric polyposis initiation. In order to identify the specific roles played by mesenchymal Bmp signaling in gastric homeostasis, we generated a mouse model with abrogation of Bmp signaling exclusively in the gastro-intestinal mesenchyme (Bmpr1aΔMES). We were able to expose an unsuspected role for Bmp loss of signaling in leading normal gastric mesenchyme to adapt into reactive mesenchyme. An increase in the population of activated-fibroblasts, suggesting mesenchymal transdifferentiation, was observed in mutant stomach. Bmpr1aΔMES stomachs exhibited spontaneous benign polyps with presence of both intestinal metaplasia and spasmolytic-polypeptide-expressing metaplasia as early as 90 days postnatal. These results support the novel concept that loss of mesenchymal Bmp signaling cascade acts as a trigger in gastric polyposis initiation.
Collapse
Affiliation(s)
- Sébastien A B Roy
- Département d'Anatomie et Biologie Cellulaire, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Joannie M Allaire
- Département d'Anatomie et Biologie Cellulaire, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Camille Ouellet
- Département d'Anatomie et Biologie Cellulaire, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Faiza Maloum-Rami
- Département d'Anatomie et Biologie Cellulaire, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Véronique Pomerleau
- Département d'Anatomie et Biologie Cellulaire, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Étienne Lemieux
- Département d'Anatomie et Biologie Cellulaire, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Jean-Philippe Babeu
- Département d'Anatomie et Biologie Cellulaire, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Jasmin Rousseau
- Département d'Anatomie et Biologie Cellulaire, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Marilène Paquet
- Département de pathologie et de microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, St-Hyacinthe, QC, Canada
| | - Perrine Garde-Granger
- Département de Pathologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - François Boudreau
- Département d'Anatomie et Biologie Cellulaire, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Nathalie Perreault
- Département d'Anatomie et Biologie Cellulaire, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
30
|
Niwa O, Barcellos-Hoff MH, Globus RK, Harrison JD, Hendry JH, Jacob P, Martin MT, Seed TM, Shay JW, Story MD, Suzuki K, Yamashita S. ICRP Publication 131: Stem Cell Biology with Respect to Carcinogenesis Aspects of Radiological Protection. Ann ICRP 2016; 44:7-357. [PMID: 26637346 DOI: 10.1177/0146645315595585] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
This report provides a review of stem cells/progenitor cells and their responses to ionising radiation in relation to issues relevant to stochastic effects of radiation that form a major part of the International Commission on Radiological Protection's system of radiological protection. Current information on stem cell characteristics, maintenance and renewal, evolution with age, location in stem cell 'niches', and radiosensitivity to acute and protracted exposures is presented in a series of substantial reviews as annexes concerning haematopoietic tissue, mammary gland, thyroid, digestive tract, lung, skin, and bone. This foundation of knowledge of stem cells is used in the main text of the report to provide a biological insight into issues such as the linear-no-threshold (LNT) model, cancer risk among tissues, dose-rate effects, and changes in the risk of radiation carcinogenesis by age at exposure and attained age. Knowledge of the biology and associated radiation biology of stem cells and progenitor cells is more developed in tissues that renew fairly rapidly, such as haematopoietic tissue, intestinal mucosa, and epidermis, although all the tissues considered here possess stem cell populations. Important features of stem cell maintenance, renewal, and response are the microenvironmental signals operating in the niche residence, for which a well-defined spatial location has been identified in some tissues. The identity of the target cell for carcinogenesis continues to point to the more primitive stem cell population that is mostly quiescent, and hence able to accumulate the protracted sequence of mutations necessary to result in malignancy. In addition, there is some potential for daughter progenitor cells to be target cells in particular cases, such as in haematopoietic tissue and in skin. Several biological processes could contribute to protecting stem cells from mutation accumulation: (a) accurate DNA repair; (b) rapidly induced death of injured stem cells; (c) retention of the DNA parental template strand during divisions in some tissue systems, so that mutations are passed to the daughter differentiating cells and not retained in the parental cell; and (d) stem cell competition, whereby undamaged stem cells outcompete damaged stem cells for residence in the niche. DNA repair mainly occurs within a few days of irradiation, while stem cell competition requires weeks or many months depending on the tissue type. The aforementioned processes may contribute to the differences in carcinogenic radiation risk values between tissues, and may help to explain why a rapidly replicating tissue such as small intestine is less prone to such risk. The processes also provide a mechanistic insight relevant to the LNT model, and the relative and absolute risk models. The radiobiological knowledge also provides a scientific insight into discussions of the dose and dose-rate effectiveness factor currently used in radiological protection guidelines. In addition, the biological information contributes potential reasons for the age-dependent sensitivity to radiation carcinogenesis, including the effects of in-utero exposure.
Collapse
|
31
|
KHODER GHALIA, AL-MENHALI ASMAA, AL-YASSIR FARAH, KARAM SHERIFM. Potential role of probiotics in the management of gastric ulcer. Exp Ther Med 2016; 12:3-17. [PMID: 27347010 PMCID: PMC4906699 DOI: 10.3892/etm.2016.3293] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Accepted: 03/03/2016] [Indexed: 02/07/2023] Open
Abstract
Gastric ulcer is one of the most common chronic gastrointestinal diseases characterized by a significant defect in the mucosal barrier. Helicobacter pylori (H. pylori) infection and the frequent long-term use of non-steroidal anti-inflammatory drugs are major factors involved in gastric ulcer development. Acid inhibitors and antibiotics are commonly used to treat gastric ulcer. However, in the last few decades, the accumulating evidence for resistance to antibiotics and the side effects of antibiotics and acid inhibitors have drawn attention to the possible use of probiotics in the prevention and treatment of gastric ulcer. Probiotics are live microorganisms that when administered in adequate amounts confer health benefits on the host. Currently, the available experimental and clinical studies indicate that probiotics are promising for future applications in the management of gastric ulcers. This review aims to provide an overview of the general health benefits of probiotics on various systemic and gastrointestinal disorders with a special focus on gastric ulcer and the involved cellular and molecular mechanisms: i) Protection of gastric mucosal barrier; ii) upregulation of prostaglandins, mucus, growth factors and anti-inflammatory cytokines; iii) increased cell proliferation to apoptosis ratio; and iv) induction of angiogenesis. Finally, some of the available data on the possible use of probiotics in H. pylori eradication are discussed.
Collapse
Affiliation(s)
- GHALIA KHODER
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - ASMA A. AL-MENHALI
- Department of Biology, College of Science, United Arab Emirates University, Al-Ain 17666, United Arab Emirates
| | - FARAH AL-YASSIR
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain 17666, United Arab Emirates
| | - SHERIF M. KARAM
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain 17666, United Arab Emirates
| |
Collapse
|
32
|
Noreldin AE, Sogabe M, Yamano Y, Uehara M, Mahdy MAA, Elnasharty MA, Sayed-Ahmed A, Warita K, Hosaka YZ. Spatial distribution of osteoblast activating peptide in the rat stomach. Acta Histochem 2016; 118:109-17. [PMID: 26686297 DOI: 10.1016/j.acthis.2015.12.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 11/30/2015] [Accepted: 12/02/2015] [Indexed: 11/18/2022]
Abstract
Osteoblast activating peptide (OBAP) was previously reported to be expressed in the rat stomach and to have a vital role in osteogenesis, but its distribution in rat stomach has not been determined. Thus, the aim of the present study was to identify the cell types expressing OBAP in the rat stomach. The stomachs of twelve 10-to-11-week-old male Jc1:SD rats were used. Samples were collected for immunohistochemistry, immunoelectron microscopy and dot blot assay. Immunohistochemical investigation revealed that OBAP was distributed mainly in parietal cells without any expression in chief cells, X/A-like cells or enterochromaffin-like cells. Moreover, OBAP-immunopositive cells were observed mainly in the upper and lower parts of the gastric gland. Significantly high optical density of immunopositive cells was observed in the upper and lower gastric gland regions. The dot blot assay confirmed that OBAP is secreted by parietal cells and that it is present in the gastric gland lumen. Immunoelectron microscopy demonstrated that OBAP was confined to the mitochondrial inner membrane within parietal cells and that the number of mitochondria in the upper and lower parts of the gastric epithelium was significantly larger than the number in the middle part of the gastric epithelium. Based on the results, it was concluded that OBAP is mainly produced by mitochondria of parietal cells in the upper and lower parts of the gastric epithelium. Moreover, the presence of OBAP in the gastric gland lumen suggests an exocrine mechanism of release.
Collapse
Affiliation(s)
- Ahmed E Noreldin
- Department of Veterinary Anatomy, Faculty of Agriculture, Tottori University, Tottori, Japan; Department of Histology and Cytology, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| | - Maina Sogabe
- Department of Veterinary Anatomy, Faculty of Agriculture, Tottori University, Tottori, Japan
| | - Yoshiaki Yamano
- Department of Veterinary Biochemistry, Faculty of Agriculture, Tottori University, Tottori, Japan; Department of Basic Veterinary Science, United Graduate School of Veterinary Science, Yamaguchi University, Yamaguchi, Japan
| | - Masato Uehara
- Department of Veterinary Anatomy, Faculty of Agriculture, Tottori University, Tottori, Japan
| | - Mohamed A A Mahdy
- Department of Basic Veterinary Science, United Graduate School of Veterinary Science, Yamaguchi University, Yamaguchi, Japan; Department of Anatomy and Embryology, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Mohamed A Elnasharty
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| | - Ahmed Sayed-Ahmed
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| | - Katsuhiko Warita
- Department of Veterinary Anatomy, Faculty of Agriculture, Tottori University, Tottori, Japan
| | - Yoshinao Z Hosaka
- Department of Veterinary Anatomy, Faculty of Agriculture, Tottori University, Tottori, Japan; Department of Basic Veterinary Science, United Graduate School of Veterinary Science, Yamaguchi University, Yamaguchi, Japan.
| |
Collapse
|
33
|
Qiu LX, Cheng L, He J, Zhou ZR, Wang MY, Zhou F, Guo WJ, Li J, Sun MH, Zhou XY, Wang YN, Yang YJ, Wang JC, Jin L, Zhu XD, Wei QY. PSCA polymorphisms and gastric cancer susceptibility in an eastern Chinese population. Oncotarget 2016; 7:9420-8. [PMID: 26848528 PMCID: PMC4891049 DOI: 10.18632/oncotarget.7137] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 01/19/2016] [Indexed: 02/07/2023] Open
Abstract
The prostate stem cell antigen (PSCA) gene, which encodes a prostate-specific antigen (PSA), was identified as a gene involved in cell adhesion and proliferation. The associations between the PSCA rs2294008 and rs2976392 single nucleotide polymorphisms (SNPs) and gastric cancer (GCa) susceptibility were still controversial. To derive a more precise estimation of the associations, we conducted a case-control study of 1,124 cases and 1,192 controls in an eastern Chinese population. We found that the rs2294008T variant genotypes were associated with an increased GCa risk in this study population (CT vs CC, OR=1.59, 95% CI=1.33-1.89 and CT+TT vs CC, OR=1.38, 95% CI=1.17-1.62). For SNP rs2976392, the variant A genotypes were also associated with an increased GCa risk (AG vs GG, OR=1.61, 95% CI=1.35-1.91 and AG+AA vs GG, OR=1.47, 95% CI=1.25-1.74). The results were further validated by a meta-analysis. In conclusion, the results indicated that the PSCA rs2294008 T and rs2976392 A alleles were low-penetrate risk factors for GCa in this study population. However, large and well-designed studies are warranted to validate our findings.
Collapse
Affiliation(s)
- Li-Xin Qiu
- 1 Department of Medical Oncology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- 2 Cancer Institute, Collaborative Innovation Center for Cancer Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Lei Cheng
- 2 Cancer Institute, Collaborative Innovation Center for Cancer Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jing He
- 3 Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Zhi-Rui Zhou
- 4 Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Meng-Yun Wang
- 2 Cancer Institute, Collaborative Innovation Center for Cancer Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Fei Zhou
- 2 Cancer Institute, Collaborative Innovation Center for Cancer Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Wei-Jian Guo
- 1 Department of Medical Oncology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jin Li
- 1 Department of Medical Oncology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Meng-Hong Sun
- 5 Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xiao-Yan Zhou
- 5 Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ya-Nong Wang
- 6 Department of Gastric Cancer & Soft Tissue Sarcoma Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ya-Jun Yang
- 7 Ministry of Education Key Laboratory of Contemporary Anthropology and State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
- 8 Fudan-Taizhou Institute of Health Sciences, Taizhou, China
| | - Jiu-Cun Wang
- 7 Ministry of Education Key Laboratory of Contemporary Anthropology and State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
- 8 Fudan-Taizhou Institute of Health Sciences, Taizhou, China
| | - Li Jin
- 7 Ministry of Education Key Laboratory of Contemporary Anthropology and State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
- 8 Fudan-Taizhou Institute of Health Sciences, Taizhou, China
| | - Xiao-Dong Zhu
- 1 Department of Medical Oncology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qing-Yi Wei
- 2 Cancer Institute, Collaborative Innovation Center for Cancer Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
- 9 Duke Cancer Institute, Duke University Medical Center, and Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
34
|
Amieva M, Peek RM. Pathobiology of Helicobacter pylori-Induced Gastric Cancer. Gastroenterology 2016; 150:64-78. [PMID: 26385073 PMCID: PMC4691563 DOI: 10.1053/j.gastro.2015.09.004] [Citation(s) in RCA: 598] [Impact Index Per Article: 66.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 09/01/2015] [Accepted: 09/03/2015] [Indexed: 02/07/2023]
Abstract
Colonization of the human stomach by Helicobacter pylori and its role in causing gastric cancer is one of the richest examples of a complex relationship among human cells, microbes, and their environment. It is also a puzzle of enormous medical importance given the incidence and lethality of gastric cancer worldwide. We review recent findings that have changed how we view these relationships and affected the direction of gastric cancer research. For example, recent data have indicated that subtle mismatches between host and microbe genetic traits greatly affect the risk of gastric cancer. The ability of H pylori and its oncoprotein CagA to reprogram epithelial cells and activate properties of stemness show the sophisticated relationship between H pylori and progenitor cells in the gastric mucosa. The observation that cell-associated H pylori can colonize the gastric glands and directly affect precursor and stem cells supports these observations. The ability to mimic these interactions in human gastric organoid cultures as well as animal models will allow investigators to more fully unravel the extent of H pylori control on the renewing gastric epithelium. Finally, our realization that external environmental factors, such as dietary components and essential micronutrients, as well as the gastrointestinal microbiota, can change the balance between H pylori's activity as a commensal or a pathogen has provided direction to studies aimed at defining the full carcinogenic potential of this organism.
Collapse
Affiliation(s)
- Manuel Amieva
- Department of Microbiology and Immunology, Stanford University, Palo Alto, California; Department of Pediatrics, Stanford University, Palo Alto, California
| | - Richard M Peek
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University, Nashville, Tennessee; Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee.
| |
Collapse
|
35
|
McClatchey PM, Keller AC, Bouchard R, Knaub LA, Reusch JEB. Fully automated software for quantitative measurements of mitochondrial morphology. Mitochondrion 2016; 26:58-71. [PMID: 26688338 PMCID: PMC5891219 DOI: 10.1016/j.mito.2015.12.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 11/09/2015] [Accepted: 12/09/2015] [Indexed: 10/22/2022]
Abstract
Mitochondria undergo dynamic changes in morphology in order to adapt to changes in nutrient and oxygen availability, communicate with the nucleus, and modulate intracellular calcium dynamics. Many recent papers have been published assessing mitochondrial morphology endpoints. Although these studies have yielded valuable insights, contemporary assessment of mitochondrial morphology is typically subjective and qualitative, precluding direct comparison of outcomes between different studies and likely missing many subtle effects. In this paper, we describe a novel software technique for measuring the average length, average width, spatial density, and intracellular localization of mitochondria from a fluorescent microscope image. This method was applied to distinguish baseline characteristics of Human Umbilical Vein Endothelial Cells (HUVECs), primary Goto-Kakizaki rat aortic smooth muscle cells (GK SMCs), primary Wistar rat aortic smooth muscle cells (Wistar SMCs), and SH-SY5Ys (human neuroblastoma cell line). Consistent with direct observation, our algorithms found SH-SY5Ys to have the greatest mitochondrial density, while HUVECs were found to have the longest mitochondria. Mitochondrial morphology responses to temperature, nutrient, and oxidative stressors were characterized to test algorithm performance. Large morphology changes recorded by the software agreed with direct observation, and subtle but consistent morphology changes were found that would not otherwise have been detected. Endpoints were consistent between experimental repetitions (R=0.93 for length, R=0.93 for width, R=0.89 for spatial density, and R=0.74 for localization), and maintained reasonable agreement even when compared to images taken with compromised microscope resolution or in an alternate imaging plane. These results indicate that the automated software described herein allows quantitative and objective characterization of mitochondrial morphology from fluorescent microscope images.
Collapse
Affiliation(s)
- P Mason McClatchey
- Division of Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Medicine, Denver VA Medical Center, Denver, CO, USA; Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Amy C Keller
- Division of Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Medicine, Denver VA Medical Center, Denver, CO, USA
| | - Ron Bouchard
- Department of Medicine, Denver VA Medical Center, Denver, CO, USA
| | - Leslie A Knaub
- Division of Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Medicine, Denver VA Medical Center, Denver, CO, USA
| | - Jane E B Reusch
- Division of Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Medicine, Denver VA Medical Center, Denver, CO, USA; Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Center for Women's Health Research, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
36
|
Yoshida S, Yamamoto H, Tetsui T, Kobayakawa Y, Hatano R, Mukaisho KI, Hattori T, Sugihara H, Asano S. Effects of ezrin knockdown on the structure of gastric glandular epithelia. J Physiol Sci 2016; 66:53-65. [PMID: 26329936 PMCID: PMC10717290 DOI: 10.1007/s12576-015-0393-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 08/18/2015] [Indexed: 10/23/2022]
Abstract
Ezrin, an adaptor protein that cross-links plasma membrane-associated proteins with the actin cytoskeleton, is concentrated on apical surfaces of epithelial cells, especially in microvilli of the small intestine and stomach. In the stomach, ezrin is predominantly expressed on the apical canalicular membrane of parietal cells. Transgenic ezrin knockdown mice in which the expression level of ezrin was reduced to <7% compared with the wild-type suffered from achlorhydria because of impairment of membrane fusion between tubulovesicles and apical membranes. We observed, for the first time, hypergastrinemia and foveolar hyperplasia in the gastric fundic region of the knockdown mice. Dilation of fundic glands was observed, the percentage of parietal and chief cells was reduced, and that of mucous-secreting cells was increased. The parietal cells of knockdown mice contained dilated tubulovesicles and abnormal mitochondria, and subsets of these cells contained abnormal vacuoles and multilamellar structures. Therefore, lack of ezrin not only causes achlorhydria and hypergastrinemia but also changes the structure of gastric glands, with severe perturbation of the secretory membranes of parietal cells.
Collapse
Affiliation(s)
- Saori Yoshida
- Department of Molecular Physiology, College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan
| | - Hiroto Yamamoto
- Department of Molecular Physiology, College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan
- Department of Pathology, Shiga University of Medical Sciences, Seta Tsukinowa-cho, Otsu, Shiga, 520-2192, Japan
| | - Takahito Tetsui
- Department of Molecular Physiology, College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan
| | - Yuka Kobayakawa
- Department of Molecular Physiology, College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan
| | - Ryo Hatano
- Department of Molecular Physiology, College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan
| | - Ken-ichi Mukaisho
- Department of Pathology, Shiga University of Medical Sciences, Seta Tsukinowa-cho, Otsu, Shiga, 520-2192, Japan
| | - Takanori Hattori
- Department of Pathology, Shiga University of Medical Sciences, Seta Tsukinowa-cho, Otsu, Shiga, 520-2192, Japan
| | - Hiroyuki Sugihara
- Department of Pathology, Shiga University of Medical Sciences, Seta Tsukinowa-cho, Otsu, Shiga, 520-2192, Japan
| | - Shinji Asano
- Department of Molecular Physiology, College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan.
| |
Collapse
|
37
|
Vange P, Bruland T, Beisvag V, Erlandsen SE, Flatberg A, Doseth B, Sandvik AK, Bakke I. Genome-wide analysis of the oxyntic proliferative isthmus zone reveals ASPM as a possible gastric stem/progenitor cell marker over-expressed in cancer. J Pathol 2015; 237:447-59. [PMID: 26178168 PMCID: PMC5049620 DOI: 10.1002/path.4591] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 06/22/2015] [Accepted: 07/13/2015] [Indexed: 12/20/2022]
Abstract
The oxyntic proliferative isthmus zone contains the main stem/progenitor cells that provide for physiological renewal of the distinct mature cell lineages in the oxyntic epithelium of the stomach. These cells are also proposed to be the potential cells-of-origin of gastric cancer, although little is known about their molecular characteristics and specific biological markers are lacking. In this study, we developed a method for serial section-navigated laser microdissection to isolate cells from the proliferative isthmus zone of rat gastric oxyntic mucosa for genome-wide microarray gene expression analysis. Enrichment analysis showed a distinct gene expression profile for the isthmus zone, with genes regulating intracellular processes such as the cell cycle and ribosomal activity. The profile was also related to stem cell transcriptional networks and stomach neoplasia. Genes expressed uniquely in the isthmus zone were associated with E2F transcription factor 1 (E2F1), which participates in the self-renewal of stem cells and in gastric carcinogenesis. One of the unique genes was Aspm [Asp (abnormal spindle) homologue, microcephaly-associated (Drosophila)]. Here we show ASPM in single scattered epithelial cells located in the proliferative isthmus zone of rat, mouse and human oxyntic mucosa, which do not seem to be actively dividing. The ASPM-expressing cells are mainly mature cell marker-deficient, except for a limited overlap with cells with neuroendocrine and tuft cell features. Further, both ASPM and E2F1 were expressed in human gastric cancer cell lines and increased and correlated in human gastric adenocarcinomas compared to non-tumour mucosa, as shown by expression profile analyses and immunohistochemistry. The association between ASPM and the transcription factor E2F1 in gastric tissue is relevant, due to their common involvement in crucial cell fate-regulatory mechanisms. Our results thus introduce ASPM as a novel possible oxyntic stem/progenitor cell marker that may be involved in both normal gastric physiology and gastric carcinogenesis.
Collapse
Affiliation(s)
- Pål Vange
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Central Norway Regional Health Authority (RHA), Stjørdal, Norway
| | - Torunn Bruland
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Central Norway Regional Health Authority (RHA), Stjørdal, Norway
| | - Vidar Beisvag
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Sten Even Erlandsen
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Arnar Flatberg
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Berit Doseth
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Central Norway Regional Health Authority (RHA), Stjørdal, Norway
| | - Arne K Sandvik
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Central Norway Regional Health Authority (RHA), Stjørdal, Norway.,Department of Gastroenterology and Hepatology, St. Olav's University Hospital, Trondheim, Norway.,Centre of Molecular Inflammation Research (CEMIR), NTNU, Trondheim, Norway
| | - Ingunn Bakke
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Central Norway Regional Health Authority (RHA), Stjørdal, Norway
| |
Collapse
|
38
|
Hoffmann W. [Continual self-renewal of the gastric epithelium by cell differentiation: implications for carcinogenesis]. DER PATHOLOGE 2015; 35 Suppl 2:202-6. [PMID: 25394968 DOI: 10.1007/s00292-014-1996-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND The gastric mucosa and its glands represent a close interactive barrier to the outside world. This delicate surface is protected by a multilayered mucus barrier which contains among others the mucins MUC5AC and MUC6 and the trefoil factor family peptide TFF2. Furthermore, two types of gastric glands form delicate homeostatic systems, i.e. the fundic and antral glands, which show continual bidirectional self-renewal via differentiation from stem and progenitor cells. It was the aim of this study to analyze the self-renewal of these gastric units. MATERIAL AND METHODS Three characteristic regions (i.e. foveolar, proliferative zone and lower gland regions) were isolated from fundic and antral units by the use of laser microdissection and expression profiles concerning known marker genes were generated by reverse transcription polymerase chain reaction (RT-PCR) analysis. RESULTS The surface mucous cells (SMCs) of fundic and antral units characteristically differed in the expression of certain secretory genes. Furthermore, the maturation of mucous neck cells and their trans-differentiation into chief cells as well as the maturation of antral SMCs and antral gland cells occurred in a stepwise manner. DISCUSSION The correct maturation particularly of mucous neck cells and their trans-differentiation into chief cells is critical for homeostatic self-renewal of fundic units. Dysregulation of this multistep process can result in generation of the spasmolytic polypeptide-expressing metaplasia (SPEM) lineage which is characterized by its strong ectopic TFF2 expression. Chronic inflammation is known to support SPEM formation. The SPEM lineage is a precancerous lesion which can further differentiate into intestinal metaplasia.
Collapse
Affiliation(s)
- W Hoffmann
- Institut für Molekularbiologie und Medizinische Chemie, Medizinische Fakultät, Otto-von-Guericke-Universität, Leipziger Str. 44, 39120, Magdeburg, Deutschland,
| |
Collapse
|
39
|
Current Status on Stem Cells and Cancers of the Gastric Epithelium. Int J Mol Sci 2015; 16:19153-69. [PMID: 26287172 PMCID: PMC4581291 DOI: 10.3390/ijms160819153] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 07/28/2015] [Accepted: 08/06/2015] [Indexed: 12/13/2022] Open
Abstract
Gastric cancer is still a leading cause of cancer-related mortality worldwide in spite of declining incidence. Gastric cancers are, essentially, adenocarcinomas and one of the strongest risk factors is still infection with Helicobacter pylori. Within the last years, it became clear that gastric self-renewal and carcinogenesis are intimately linked, particularly during chronic inflammatory conditions. Generally, gastric cancer is now regarded as a disease resulting from dysregulated differentiation of stem and progenitor cells, mainly due to an inflammatory environment. However, the situation in the stomach is rather complex, consisting of two types of gastric units which show bidirectional self-renewal from an unexpectedly large variety of progenitor/stem cell populations. As in many other tumors, cancer stem cells have also been characterized for gastric cancer. This review focuses on the various gastric epithelial stem cells, how they contribute to self-renewal and which routes are known to gastric adenocarcinomas, including their stem cells.
Collapse
|
40
|
Arapis K, Cavin JB, Gillard L, Cluzeaud F, Lettéron P, Ducroc R, Le Beyec J, Hourseau M, Couvelard A, Marmuse JP, Le Gall M, Bado A. Remodeling of the residual gastric mucosa after roux-en-y gastric bypass or vertical sleeve gastrectomy in diet-induced obese rats. PLoS One 2015; 10:e0121414. [PMID: 25822172 PMCID: PMC4379088 DOI: 10.1371/journal.pone.0121414] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 01/31/2015] [Indexed: 01/06/2023] Open
Abstract
Whereas the remodeling of intestinal mucosa after bariatric surgeries has been the matter of numerous studies to our knowledge, very few reported on the remodeling of the residual gastric mucosa. In this study, we analyzed remodeling of gastric mucosa after Roux-en-Y gastric bypass (RYGB) and vertical sleeve gastrectomy (VSG) in rats. Diet-induced obese rats were subjected to RYGB, VSG or sham surgical procedures. All animals were assessed for food intake, body-weight, fasting blood, metabolites and hormones profiling, as well as insulin and glucose tolerance tests before and up to 5 weeks post-surgery. Remodeling of gastric tissues was analyzed by routine histology and immunohistochemistry studies, and qRT-PCR analyses of ghrelin and gastrin mRNA levels. In obese rats with impaired glucose tolerance, VSG and RYGB caused substantial weight loss and rats greatly improved their oral glucose tolerance. The remaining gastric mucosa after VSG and gastric pouch (GP) after RYGB revealed a hyperplasia of the mucous neck cells that displayed a strong immunoreactivity for parietal cell H+/K+-ATPase. Ghrelin mRNA levels were reduced by 2-fold in remaining fundic mucosa after VSG and 10-fold in GP after RYGB. In the antrum, gastrin mRNA levels were reduced after VSG in line with the reduced number of gastrin positive cells. This study reports novel and important observations dealing with the remaining gastric mucosa after RYGB and VSG. The data demonstrate, for the first time, a hyperplasia of the mucous neck cells, a transit cell population of the stomach bearing differentiating capacities into zymogenic and peptic cells.
Collapse
Affiliation(s)
- Konstantinos Arapis
- Inserm UMR 1149, UFR de Médecine Paris Diderot, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- DHU Unity, Bichat-Beaujon AP-HP, Paris, France
- Service de Chirurgie Générale et Digestive; Hôpital Bichat—Claude Bernard. Paris, France
| | - Jean Baptiste Cavin
- Inserm UMR 1149, UFR de Médecine Paris Diderot, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- DHU Unity, Bichat-Beaujon AP-HP, Paris, France
| | - Laura Gillard
- Inserm UMR 1149, UFR de Médecine Paris Diderot, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- DHU Unity, Bichat-Beaujon AP-HP, Paris, France
| | - Françoise Cluzeaud
- Inserm UMR 1149, UFR de Médecine Paris Diderot, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- DHU Unity, Bichat-Beaujon AP-HP, Paris, France
| | - Philippe Lettéron
- Inserm UMR 1149, UFR de Médecine Paris Diderot, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- DHU Unity, Bichat-Beaujon AP-HP, Paris, France
| | - Robert Ducroc
- Inserm UMR 1149, UFR de Médecine Paris Diderot, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- DHU Unity, Bichat-Beaujon AP-HP, Paris, France
| | - Johanne Le Beyec
- Inserm UMR 1149, UFR de Médecine Paris Diderot, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- DHU Unity, Bichat-Beaujon AP-HP, Paris, France
| | - Muriel Hourseau
- DHU Unity, Bichat-Beaujon AP-HP, Paris, France
- Département de Pathologie; Hôpital Bichat—Claude Bernard, Paris, France
| | - Anne Couvelard
- Inserm UMR 1149, UFR de Médecine Paris Diderot, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- DHU Unity, Bichat-Beaujon AP-HP, Paris, France
- Département de Pathologie; Hôpital Bichat—Claude Bernard, Paris, France
| | - Jean-Pierre Marmuse
- Inserm UMR 1149, UFR de Médecine Paris Diderot, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- DHU Unity, Bichat-Beaujon AP-HP, Paris, France
- Service de Chirurgie Générale et Digestive; Hôpital Bichat—Claude Bernard. Paris, France
| | - Maude Le Gall
- Inserm UMR 1149, UFR de Médecine Paris Diderot, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- DHU Unity, Bichat-Beaujon AP-HP, Paris, France
| | - André Bado
- Inserm UMR 1149, UFR de Médecine Paris Diderot, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- DHU Unity, Bichat-Beaujon AP-HP, Paris, France
- * E-mail:
| |
Collapse
|
41
|
Alfazari AS, Al-Dabbagh B, Al-Dhaheri W, Taha MS, Chebli AA, Fontagnier EM, Koutoubi Z, Kochiyi J, Karam SM, Souid AK. Profiling cellular bioenergetics, glutathione levels, and caspase activities in stomach biopsies of patients with upper gastrointestinal symptoms. World J Gastroenterol 2015. [PMID: 25593494 DOI: 10.3748/wjg.v21.i2.635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM To measure biochemical parameters in stomach biopsies and test their suitability as diagnostic biomarkers for gastritis and precancerous lesions. METHODS Biopsies were obtained from the stomachs of two groups of patients (n = 40) undergoing fiber-optic endoscopy due to upper gastrointestinal symptoms. In the first group (n = 17), only the corpus region was examined. Biopsies were processed for microscopic examination and measurement of mitochondrial O2 consumption (cellular respiration), cellular adenosine triphosphate (ATP), glutathione (GSH), and caspase activity. In the second group of patients (n = 23), both corpus and antral regions were studied. Some biopsies were processed for microscopic examination, while the others were used for measurements of cellular respiration and GSH level. RESULTS Microscopic examinations of gastric corpus biopsies from 17 patients revealed normal mucosae in 8 patients, superficial gastritis in 7 patients, and chronic atrophic gastritis in 1 patient. In patients with normal histology, the rate (mean ± SD) of cellular respiration was 0.17 ± 0.02 μmol/L O2 min(-1) mg(-1), ATP content was 487 ± 493 pmol/mg, and GSH was 469 ± 98 pmol/mg. Caspase activity was detected in 3 out of 8 specimens. The values of ATP and caspase activity were highly variable. The presence of superficial gastritis had insignificant effects on the measured biomarkers. In the patient with atrophic gastritis, cellular respiration was high and ATP was relatively low, suggesting uncoupling oxidative phosphorylation. In the second cohort of patients, the examined biopsies showed either normal or superficial gastritis. The rate of cellular respiration (O2. μmol/L min(-1) mg(-1)) was slightly higher in the corpus than the antrum (0.18 ± 0.05 vs 0.15 ± 0.04, P = 0.019). The value of GSH was about the same in both tissues (310 ± 135 vs 322 ± 155, P = 0.692). CONCLUSION The corpus mucosa was metabolically more active than the antrum tissue. The data in this study will help in understanding the pathophysiology of gastric mucosa.
Collapse
Affiliation(s)
- Ali S Alfazari
- Ali S Alfazari, Bayan Al-Dabbagh, Department of Medicine, United Arab Emirates University, PO Box 17666, Al Ain, United Arab Emirates
| | - Bayan Al-Dabbagh
- Ali S Alfazari, Bayan Al-Dabbagh, Department of Medicine, United Arab Emirates University, PO Box 17666, Al Ain, United Arab Emirates
| | - Wafa Al-Dhaheri
- Ali S Alfazari, Bayan Al-Dabbagh, Department of Medicine, United Arab Emirates University, PO Box 17666, Al Ain, United Arab Emirates
| | - Mazen S Taha
- Ali S Alfazari, Bayan Al-Dabbagh, Department of Medicine, United Arab Emirates University, PO Box 17666, Al Ain, United Arab Emirates
| | - Ahmad A Chebli
- Ali S Alfazari, Bayan Al-Dabbagh, Department of Medicine, United Arab Emirates University, PO Box 17666, Al Ain, United Arab Emirates
| | - Eva M Fontagnier
- Ali S Alfazari, Bayan Al-Dabbagh, Department of Medicine, United Arab Emirates University, PO Box 17666, Al Ain, United Arab Emirates
| | - Zaher Koutoubi
- Ali S Alfazari, Bayan Al-Dabbagh, Department of Medicine, United Arab Emirates University, PO Box 17666, Al Ain, United Arab Emirates
| | - Jose Kochiyi
- Ali S Alfazari, Bayan Al-Dabbagh, Department of Medicine, United Arab Emirates University, PO Box 17666, Al Ain, United Arab Emirates
| | - Sherif M Karam
- Ali S Alfazari, Bayan Al-Dabbagh, Department of Medicine, United Arab Emirates University, PO Box 17666, Al Ain, United Arab Emirates
| | - Abdul-Kader Souid
- Ali S Alfazari, Bayan Al-Dabbagh, Department of Medicine, United Arab Emirates University, PO Box 17666, Al Ain, United Arab Emirates
| |
Collapse
|
42
|
Alfazari AS, Al-Dabbagh B, Al-Dhaheri W, Taha MS, Chebli AA, Fontagnier EM, Koutoubi Z, Kochiyi J, Karam SM, Souid AK. Profiling cellular bioenergetics, glutathione levels, and caspase activities in stomach biopsies of patients with upper gastrointestinal symptoms. World J Gastroenterol 2015; 21:644-652. [PMID: 25593494 PMCID: PMC4292300 DOI: 10.3748/wjg.v21.i2.644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 06/30/2014] [Accepted: 07/30/2014] [Indexed: 02/07/2023] Open
Abstract
AIM: To measure biochemical parameters in stomach biopsies and test their suitability as diagnostic biomarkers for gastritis and precancerous lesions.
METHODS: Biopsies were obtained from the stomachs of two groups of patients (n = 40) undergoing fiber-optic endoscopy due to upper gastrointestinal symptoms. In the first group (n = 17), only the corpus region was examined. Biopsies were processed for microscopic examination and measurement of mitochondrial O2 consumption (cellular respiration), cellular adenosine triphosphate (ATP), glutathione (GSH), and caspase activity. In the second group of patients (n = 23), both corpus and antral regions were studied. Some biopsies were processed for microscopic examination, while the others were used for measurements of cellular respiration and GSH level.
RESULTS: Microscopic examinations of gastric corpus biopsies from 17 patients revealed normal mucosae in 8 patients, superficial gastritis in 7 patients, and chronic atrophic gastritis in 1 patient. In patients with normal histology, the rate (mean ± SD) of cellular respiration was 0.17 ± 0.02 μmol/L O2 min-1 mg-1, ATP content was 487 ± 493 pmol/mg, and GSH was 469 ± 98 pmol/mg. Caspase activity was detected in 3 out of 8 specimens. The values of ATP and caspase activity were highly variable. The presence of superficial gastritis had insignificant effects on the measured biomarkers. In the patient with atrophic gastritis, cellular respiration was high and ATP was relatively low, suggesting uncoupling oxidative phosphorylation. In the second cohort of patients, the examined biopsies showed either normal or superficial gastritis. The rate of cellular respiration (O2.μmol/L min-1 mg-1) was slightly higher in the corpus than the antrum (0.18 ± 0.05 vs 0.15 ± 0.04, P = 0.019). The value of GSH was about the same in both tissues (310 ± 135 vs 322 ± 155, P = 0.692).
CONCLUSION: The corpus mucosa was metabolically more active than the antrum tissue. The data in this study will help in understanding the pathophysiology of gastric mucosa.
Collapse
|
43
|
Zhao Y, Feng F, Zhou YN. Stem cells in gastric cancer. World J Gastroenterol 2015; 21:112-123. [PMID: 25574084 PMCID: PMC4284326 DOI: 10.3748/wjg.v21.i1.112] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 09/19/2014] [Accepted: 10/21/2014] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer (GC) is one of the leading causes of cancer-related mortality worldwide. Cancer stem cells (CSCs), which were first identified in acute myeloid leukemia and subsequently in a large array of solid tumors, play important roles in cancer initiation, dissemination and recurrence. CSCs are often transformed tissue-specific stem cells or de-differentiated transit amplifying progenitor cells. Several populations of multipotent gastric stem cells (GSCs) that reside in the stomach have been determined to regulate physiological tissue renewal and injury repair. These populations include the Villin+ and Lgr5+ GSCs in the antrum, the Troy+ chief cells in the corpus, and the Sox2+ GSCs that are found in both the antrum and the corpus. The disruption of tumor suppressors in Villin+ or Lgr5+ GSCs leads to GC in mouse models. In addition to residing GSCs, bone marrow-derived cells can initiate GC in a mouse model of chronic Helicobacter infection. Furthermore, expression of the cell surface markers CD133 or CD44 defines gastric CSCs in mouse models and in human primary GC tissues and cell lines. Targeted elimination of CSCs effectively reduces tumor size and grade in mouse models. In summary, the recent identification of normal GSCs and gastric CSCs has greatly improved our understanding of the molecular and cellular etiology of GC and will aid in the development of effective therapies to treat patients.
Collapse
|
44
|
McDonald SA, Graham TA, Lavery DL, Wright NA, Jansen M. The Barrett's Gland in Phenotype Space. Cell Mol Gastroenterol Hepatol 2015; 1:41-54. [PMID: 28247864 PMCID: PMC5301147 DOI: 10.1016/j.jcmgh.2014.10.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 10/15/2014] [Indexed: 02/06/2023]
Abstract
Barrett's esophagus is characterized by the erosive replacement of esophageal squamous epithelium by a range of metaplastic glandular phenotypes. These glandular phenotypes likely change over time, and their distribution varies along the Barrett's segment. Although much recent work has addressed Barrett's esophagus from the genomic viewpoint-its genotype space-the fact that the phenotype of Barrett's esophagus is nonstatic points to conversion between phenotypes and suggests that Barrett's esophagus also exists in phenotype space. Here we explore this latter concept, investigating the scope of glandular phenotypes in Barrett's esophagus and how they exist in physical and temporal space as well as their evolution and their life history. We conclude that individual Barrett's glands are clonal units; because of this important fact, we propose that it is the Barrett's gland that is the unit of selection in phenotypic and indeed neoplastic progression. Transition between metaplastic phenotypes may be governed by neutral drift akin to niche turnover in normal and dysplastic niches. In consequence, the phenotype of Barrett's glands assumes considerable importance, and we make a strong plea for the integration of the Barrett's gland in both genotype and phenotype space in future work.
Collapse
Affiliation(s)
- Stuart A.C. McDonald
- Centre for Tumour Biology, Barts Cancer Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Trevor A. Graham
- Centre for Tumour Biology, Barts Cancer Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Danielle L. Lavery
- Centre for Tumour Biology, Barts Cancer Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Nicholas A. Wright
- Centre for Tumour Biology, Barts Cancer Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Marnix Jansen
- Centre for Tumour Biology, Barts Cancer Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Department of Pathology, Academic Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
45
|
Naruki S, Fujino T, Ohnuma S, Endo A, Koizumi H, Kato Y, Takagi M. Histopathologic and Immunohistochemical Characterization of Human Gastric Oxyntic Mucosa with Parietal Cell Protrusions and Investigation into the Association Between Such Mucosal Changes of the Stomach and Use of Proton Pump Inhibitors. ACTA ACUST UNITED AC 2015. [DOI: 10.17264/stmarieng.6.119] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Saeko Naruki
- Department of Pathology, St. Marianna University School of Medicine
| | - Takashi Fujino
- Department of Pathology, St. Marianna University School of Medicine
| | - Shigeko Ohnuma
- Department of Pathology, St. Marianna University School of Medicine
| | - Akira Endo
- Department of Pathology, St. Marianna University School of Medicine
| | - Hirotaka Koizumi
- Department of Pathology, St. Marianna University School of Medicine
| | - Yo Kato
- Department of Pathology, Nikko Medical Center, Dokkyo Medical University
| | - Masayuki Takagi
- Department of Pathology, St. Marianna University School of Medicine
| |
Collapse
|
46
|
Pulikkot S, Greish YE, Mourad AI, Karam SM. Establishment of a three-dimensional culture system of gastric stem cells supporting mucous cell differentiation using microfibrous polycaprolactone scaffolds. Cell Prolif 2014; 47:553-63. [PMID: 25345659 PMCID: PMC6495834 DOI: 10.1111/cpr.12141] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Accepted: 08/02/2014] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVES To generate various polycaprolactone (PCL) scaffolds and test their suitability for growth and differentiation of immortalized mouse gastric stem (mGS) cells. MATERIALS AND METHODS Non-porous, microporous and three-dimensional electrospun microfibrous PCL scaffolds were prepared and characterized for culture of mGS cells. First, growth of mGS cells was compared on these different scaffolds after 3 days culture, using viability assay and microscopy. Secondly, growth pattern of the cells on microfibrous scaffolds was studied after 3, 6, 9 and 12 days culture using DNA PicoGreen assay and scanning electron microscopy. Thirdly, differentiation of the cells grown on microfibrous scaffolds for 3 and 9 days was analysed using lectin/immunohistochemistry. RESULTS The mGS cells grew preferentially on microfibrous scaffolds. From 3 to 6 days, there was increase in cell number, followed by reduction by days 9 and 12. To test whether the reduction in cell number was associated with cell differentiation, cryosections of cell-containing scaffolds cultured for 3 and 9 days were probed with gastric epithelial cell differentiation markers. On day 3, none of the markers examined bound to the cells. However by day 9, approximately, 50% of them bound to N-acetyl-d-glucosamine-specific lectin and anti-trefoil factor 2 antibodies, indicating their differentiation into glandular mucus-secreting cells. CONCLUSIONS Microfibrous PCL scaffolds supported growth and differentiation of mGS cells into mucus-secreting cells. These data will help lay groundwork for future experiments to explore use of gastric stem cells and PCL scaffolds in stomach tissue engineering.
Collapse
Affiliation(s)
- S. Pulikkot
- Department of AnatomyCollege of Medicine and Health SciencesUnited Arab Emirates UniversityAl AinUnited Arab Emirates
- Department of ChemistryCollege of ScienceUnited Arab Emirates UniversityAl AinUnited Arab Emirates
| | - Y. E. Greish
- Department of ChemistryCollege of ScienceUnited Arab Emirates UniversityAl AinUnited Arab Emirates
| | - A‐H. I. Mourad
- Department of Mechanical EngineeringCollege of EngineeringUnited Arab Emirates UniversityAl AinUnited Arab Emirates
| | - S. M. Karam
- Department of AnatomyCollege of Medicine and Health SciencesUnited Arab Emirates UniversityAl AinUnited Arab Emirates
| |
Collapse
|
47
|
Mihi B, Van Meulder F, Rinaldi M, Van Coppernolle S, Chiers K, Van den Broeck W, Goddeeris B, Vercruysse J, Claerebout E, Geldhof P. Analysis of cell hyperplasia and parietal cell dysfunction induced by Ostertagia ostertagi infection. Vet Res 2013; 44:121. [PMID: 24330735 PMCID: PMC3878833 DOI: 10.1186/1297-9716-44-121] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 11/29/2013] [Indexed: 02/07/2023] Open
Abstract
Infections in cattle with the gastric nematode Ostertagia ostertagi are associated with decreased acid secretion and profound physio-morphological changes of the gastric mucosa. The purpose of the current study was to investigate the mechanisms triggering these pathophysiological changes. O. ostertagi infection resulted in a marked cellular hyperplasia, which can be explained by increased transcriptional levels of signaling molecules related to the homeostasis of gastric epithelial cells such as HES1, WNT5A, FGF10, HB-EGF, AREG, ADAM10 and ADAM17. Intriguingly, histological analysis indicated that the rapid rise in the gastric pH, observed following the emergence of adult worms, cannot be explained by a loss of parietal cells, as a decrease in the number of parietal cells was only observed following a long term infection of several weeks, but is likely to be caused by an inhibition of parietal cell activity. To investigate whether this inhibition is caused by a direct effect of the parasites, parietal cells were co-cultured with parasite Excretory/Secretory products (ESP) and subsequently analyzed for acid production. The results indicate that adult ESP inhibited acid secretion, whereas ESP from the L4 larval stages did not alter parietal cell function. In addition, our data show that the inhibition of parietal cell activity could be mediated by a marked upregulation of inflammatory factors, which are partly induced by adult ESP in abomasal epithelial cells. In conclusion, this study shows that the emergence of adult O. ostertagi worms is associated with marked cellular changes that can be partly triggered by the worm’s Excretory/secretory antigens.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Peter Geldhof
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium.
| |
Collapse
|
48
|
Singh SR. Gastric cancer stem cells: a novel therapeutic target. Cancer Lett 2013; 338:110-9. [PMID: 23583679 DOI: 10.1016/j.canlet.2013.03.035] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 03/25/2013] [Accepted: 03/30/2013] [Indexed: 12/14/2022]
Abstract
Gastric cancer remains one of the leading causes of global cancer mortality. Multipotent gastric stem cells have been identified in both mouse and human stomachs, and they play an essential role in the self-renewal and homeostasis of gastric mucosa. There are several environmental and genetic factors known to promote gastric cancer. In recent years, numerous in vitro and in vivo studies suggest that gastric cancer may originate from normal stem cells or bone marrow-derived mesenchymal cells, and that gastric tumors contain cancer stem cells. Cancer stem cells are believed to share a common microenvironment with normal niche, which play an important role in gastric cancer and tumor growth. This mini-review presents a brief overview of the recent developments in gastric cancer stem cell research. The knowledge gained by studying cancer stem cells in gastric mucosa will support the development of novel therapeutic strategies for gastric cancer.
Collapse
Affiliation(s)
- Shree Ram Singh
- Mouse Cancer Genetics Program, National Cancer Institute, Frederick, MD 21702, USA.
| |
Collapse
|
49
|
Kopic S, Geibel JP. Gastric acid, calcium absorption, and their impact on bone health. Physiol Rev 2013; 93:189-268. [PMID: 23303909 DOI: 10.1152/physrev.00015.2012] [Citation(s) in RCA: 110] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Calcium balance is essential for a multitude of physiological processes, ranging from cell signaling to maintenance of bone health. Adequate intestinal absorption of calcium is a major factor for maintaining systemic calcium homeostasis. Recent observations indicate that a reduction of gastric acidity may impair effective calcium uptake through the intestine. This article reviews the physiology of gastric acid secretion, intestinal calcium absorption, and their respective neuroendocrine regulation and explores the physiological basis of a potential link between these individual systems.
Collapse
Affiliation(s)
- Sascha Kopic
- Department of Surgery and Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut, USA
| | | |
Collapse
|
50
|
Irie-Maezono R, Tsuyama S. Immunohistochemical Analysis of the Acid Secretion Potency in Gastric Parietal Cells. Cell 2013. [DOI: 10.4236/cellbio.2013.24020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|