1
|
Luo X, Luo B, Fei L, Zhang Q, Liang X, Chen Y, Zhou X. MS4A superfamily molecules in tumors, Alzheimer's and autoimmune diseases. Front Immunol 2024; 15:1481494. [PMID: 39717774 PMCID: PMC11663944 DOI: 10.3389/fimmu.2024.1481494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 11/12/2024] [Indexed: 12/25/2024] Open
Abstract
MS4A (membrane-spanning 4-domain, subfamily A) molecules are categorized into tetraspanins, which possess four-transmembrane structures. To date, eighteen MS4A members have been identified in humans, whereas twenty-three different molecules have been identified in mice. MS4A proteins are selectively expressed on the surfaces of various immune cells, such as B cells (MS4A1), mast cells (MS4A2), macrophages (MS4A4A), Foxp3+CD4+ regulatory T cells (MS4A4B), and type 3 innate lymphoid cells (TMEM176A and TMEM176B). Early research confirmed that most MS4A molecules function as ion channels that regulate the transport of calcium ions. Recent studies have revealed that some MS4A proteins also function as chaperones that interact with various immune molecules, such as pattern recognition receptors and/or immunoglobulin receptors, to form immune complexes and transmit downstream signals, leading to cell activation, growth, and development. Evidence from preclinical animal models and human genetic studies suggests that the MS4A superfamily plays critical roles in the pathogenesis of various diseases, including cancer, infection, allergies, neurodegenerative diseases and autoimmune diseases. We review recent progress in this field and focus on elucidating the molecular mechanisms by which different MS4A molecules regulate the progression of tumors, Alzheimer's disease, and autoimmune diseases. Therefore, in-depth research into MS4A superfamily members may clarify their ability to act as candidate biomarkers and therapeutic targets for these diseases. Eighteen distinct members of the MS4A (membrane-spanning four-domain subfamily A) superfamily of four-transmembrane proteins have been identified in humans, whereas the MS4A genes are translated into twenty-three different molecules in mice. These proteins are selectively expressed on the surface of various immune cells, such as B cells (MS4A1), macrophages (MS4A4A), mast cells (MS4A2), Foxp3+CD4+ regulatory T cells (MS4A4B), type 3 innate lymphoid cells (TMEM176A and TMEM176B) and colonic epithelial cells (MS4A12). Functionally, most MS4A molecules function as ion channels that regulate the flow of calcium ions [Ca2+] across cell membranes. Recent studies have revealed that some MS4A proteins also act as molecular chaperones and interact with various types of immune receptors, including pattern recognition receptors (PRRs) and immunoglobulin receptors (IgRs), to form signaling complexes, thereby modulating intracellular signaling and cellular activity. Evidence from preclinical animal models and human genetic studies suggests that MS4A proteins play critical roles in various diseases (2). Therefore, we reviewed the recent progress in understanding the role of the MS4A superfamily in diseases, particularly in elucidating its function as a candidate biomarker and therapeutic target for cancer.
Collapse
Affiliation(s)
- Xuejiao Luo
- Department of Dermatology, The Affiliated Hospital of the Non-Commissioned Officer (NCO) School, The Army Medical University, Shijiazhuang, Hebei, China
| | - Bin Luo
- Institute of Immunology, Department of Basic Medicine, The Army Military Medical University, Chongqing, China
| | - Lei Fei
- Institute of Immunology, Department of Basic Medicine, The Army Military Medical University, Chongqing, China
| | - Qinggao Zhang
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning, China
| | - Xinyu Liang
- Department of Otolaryngology, The Second Affiliated Hospital of the Army Military Medical University, Chongqing, China
| | - Yongwen Chen
- Institute of Immunology, Department of Basic Medicine, The Army Military Medical University, Chongqing, China
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning, China
| | - Xueqin Zhou
- Department of Otolaryngology, The Second Affiliated Hospital of the Army Military Medical University, Chongqing, China
| |
Collapse
|
2
|
López-Catalina A, Reverter A, Alexandre PA, Nguyen LT, González-Recio O. Stress-induced epigenetic effects driven by maternal lactation in dairy cattle: a comethylation network approach. Epigenetics 2024; 19:2381856. [PMID: 39044410 PMCID: PMC11271077 DOI: 10.1080/15592294.2024.2381856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 07/14/2024] [Indexed: 07/25/2024] Open
Abstract
Epigenetic marks do not follow the Mendelian laws of inheritance. The environment can alter the epigenotype of an individual when exposed to different external stressors. In lactating cows, the first stages of gestation overlap with the lactation peak, creating a negative energy balance that is difficult to overcome with diet. This negative energy balance could affect early embryo development that must compete with the mammary tissue for nutrients. We hypothesize that the methylation profiles of calves born to nonlactating heifers are different from those of calves born to lactating cows. We found 50,277 differentially methylated cytosines and 2,281 differentially methylated regions between these two groups of animals. A comethylation network was constructed to study the correlation between the phenotypes of the mothers and the epigenome of the calves, revealing 265 regions associated with the phenotypes. Our study revealed the presence of DMCs and DMRs in calves gestated by heifers and lactating cows, which were linked to the dam's lactation and the calves' ICAP and milk EBV. Gene-specific analysis highlighted associations with vasculature and organ morphogenesis and cell communication and signalling. These finding support the hypothesis that calves gestated by nonlactating mothers have a different methylation profile than those gestated by lactating cows.
Collapse
Affiliation(s)
- Adrián López-Catalina
- Departamento de Mejora Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), CSIC, Crta. de la Coruña km 7.5, Madrid, Spain
- Departamento de Producción Agraria, Escuela Técnica Superior de Ingeniería Agronómica, Alimentaria y de Biosistemas, Universidad Politécnica de Madrid, Ciudad Universitaria s/n, Madrid, Spain
- CSIRO Agriculture & Food, Queensland Bioscience Precinct, Brisbane, Queensland, Australia
| | - Antonio Reverter
- CSIRO Agriculture & Food, Queensland Bioscience Precinct, Brisbane, Queensland, Australia
| | - Pamela A. Alexandre
- CSIRO Agriculture & Food, Queensland Bioscience Precinct, Brisbane, Queensland, Australia
| | - Loan T. Nguyen
- Queensland Alliance for Agriculture and Food Innovation, University of Queensland, St Lucia, QLD, Australia
| | - Oscar González-Recio
- Departamento de Mejora Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), CSIC, Crta. de la Coruña km 7.5, Madrid, Spain
| |
Collapse
|
3
|
Singh S, Mahajan M, Kumar D, Singh K, Chowdhary M, Amit. An inclusive study of recent advancements in Alzheimer's disease: A comprehensive review. Neuropeptides 2023; 102:102369. [PMID: 37611472 DOI: 10.1016/j.npep.2023.102369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 08/08/2023] [Accepted: 08/11/2023] [Indexed: 08/25/2023]
Abstract
Alzheimer's disease (AD) has remained elusive in revealing its pathophysiology and mechanism of development. In this review paper, we attempt to highlight several theories that abound about the exact pathway of AD development. The number of cases worldwide has prompted a constant flow of research to detect high-risk patients, slow the progression of the disease and discover improved methods of treatment that may prove effective. We shall focus on the two main classes of drugs that are currently in use; and emerging ones with novel mechanisms that are under development. As of late there has also been increased attention towards factors that were previously thought to be unrelated to AD, such as the gut microbiome, lifestyle habits, and diet. Studies have now shown that all these factors make an impact on AD progression, thus bringing to our attention more areas that could hold the key to combating this disease. This paper covers all the aforementioned factors concisely. We also briefly explore the relationship between mental health and AD, both before and after the diagnosis of the disease.
Collapse
Affiliation(s)
- Sukanya Singh
- Department of Zoology, Hansraj College, University of Delhi, New Delhi, Delhi 110007, India
| | - Mitali Mahajan
- Department of Zoology, Hansraj College, University of Delhi, New Delhi, Delhi 110007, India
| | - Dhawal Kumar
- Department of Zoology, Hansraj College, University of Delhi, New Delhi, Delhi 110007, India
| | - Kunika Singh
- Department of Zoology, Hansraj College, University of Delhi, New Delhi, Delhi 110007, India
| | - Mehvish Chowdhary
- Department of Zoology, Hansraj College, University of Delhi, New Delhi, Delhi 110007, India
| | - Amit
- Department of Zoology, Hansraj College, University of Delhi, New Delhi, Delhi 110007, India.
| |
Collapse
|
4
|
KUROKAWA A, YAMAMOTO Y. Immunohistochemical identification of T and B lymphocytes in formalin-fixed, paraffin-embedded tissues of 53 avian species using commercial antibodies. J Vet Med Sci 2023; 85:1121-1130. [PMID: 37661384 PMCID: PMC10600538 DOI: 10.1292/jvms.23-0255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/16/2023] [Indexed: 09/05/2023] Open
Abstract
Providing a method to detect avian lymphocytes by immunohistochemistry (IHC) would be helpful for analyzing immune function and diagnosing diseases in birds. In this study, we comprehensively examined the immunohistochemical identification of avian T and B lymphocytes in formalin-fixed, paraffin-embedded tissues from 53 avian species across 15 orders, using eight commercially available lymphocyte markers. T lymphocytes from all 53 avian species tested were specifically detected by IHC using the anti-CD3 antibody (clone F7.2.38). The appropriate antibody for detecting avian B lymphocytes in IHC varied depending on the avian species. B lymphocytes were specifically labeled by IHC in 46 of 53 avian species (86.8%) using any of seven B cell markers. The anti-PAX5 antibody (clone SP34) immunohistochemically detected B lymphocytes from the majority of avian species (41 out of 53 species), excluding those in the orders Falconiformes (falcons) and Passeriformes (oscines). The anti-BAFF-R antibody (clone 2C4) proved suitable for detecting B lymphocytes in the orders Galliformes (landfowls) and Anseriformes (waterfowls) in IHC. Caution is advised when using the anti-BLA36 (clone A27-42) and two anti-CD20 (clone L26 and product No. PA5-16701) antibodies, which are commonly used as B cell markers in mammals, for detecting avian B lymphocytes. These antibodies reacted with cells located in both T and B cell areas in certain avian species. The anti-Bu-1a/b (clone AV20) and anti-CD79a (clone HM57) antibodies were found not to bind to B lymphocytes in various avian species in IHC.
Collapse
Affiliation(s)
- Aoi KUROKAWA
- National Institute of Animal Health, National Agriculture
and Food Research Organization, Ibaraki, Japan
| | - Yu YAMAMOTO
- National Institute of Animal Health, National Agriculture
and Food Research Organization, Ibaraki, Japan
| |
Collapse
|
5
|
Laghouaouta H, Fraile L, Suárez-Mesa R, Ros-Freixedes R, Estany J, Pena RN. A genome-wide screen for resilient responses in growing pigs. Genet Sel Evol 2022; 54:50. [PMID: 35787790 PMCID: PMC9251948 DOI: 10.1186/s12711-022-00739-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 06/21/2022] [Indexed: 11/18/2022] Open
Abstract
Background There is a growing interest to decipher the genetic background of resilience and its possible improvement through selective breeding. The objective of the present study was to provide new insights into the genetic make-up of resilience in growing pigs by identifying genomic regions and candidate genes associated with resilience indicators. Commercial Duroc pigs were challenged with an attenuated Aujeszky vaccine at 12 weeks of age. Two resilience indicators were used: deviation from the expected body weight at 16 weeks of age given the growth curve of non-vaccinated pigs (∆BW) and the increase in acute-phase protein haptoglobin at four days post-vaccination (∆HP). Genome-wide association analyses were carried out on 445 pigs, using genotypes at 41,165 single nucleotide polymorphisms (SNPs) and single-marker and Bayesian multiple-marker regression approaches. Results Genomic regions on pig chromosomes 2, 8, 9, 11 (∆BW) and 8, 9, 13 (∆HP) were found to be associated with the resilience indicators and explained high proportions of their genetic variance. The genomic regions that were associated explained 27 and 5% of the genetic variance of ∆BW and ∆HP, respectively. These genomic regions harbour promising candidate genes that are involved in pathways related to immune response, response to stress, or signal transduction (CD6, PTGDR2, IKZF1, RNASEL and MYD88), and growth (GRB10 and LCORL). Conclusions Our study identified novel genomic regions that are associated with two resilience indicators (∆BW and ∆HP) in pigs. These associated genomic regions harbour potential candidate genes involved in immune response and growth pathways, which emphasise the strong relationship between resilience and immune response. Supplementary Information The online version contains supplementary material available at 10.1186/s12711-022-00739-1.
Collapse
Affiliation(s)
- Houda Laghouaouta
- Department of Animal Science, University of Lleida-Agrotecnio-CERCA Center, 25198, Lleida, Catalonia, Spain
| | - Lorenzo Fraile
- Department of Animal Science, University of Lleida-Agrotecnio-CERCA Center, 25198, Lleida, Catalonia, Spain
| | - Rafael Suárez-Mesa
- Department of Animal Science, University of Lleida-Agrotecnio-CERCA Center, 25198, Lleida, Catalonia, Spain
| | - Roger Ros-Freixedes
- Department of Animal Science, University of Lleida-Agrotecnio-CERCA Center, 25198, Lleida, Catalonia, Spain
| | - Joan Estany
- Department of Animal Science, University of Lleida-Agrotecnio-CERCA Center, 25198, Lleida, Catalonia, Spain
| | - Ramona Natacha Pena
- Department of Animal Science, University of Lleida-Agrotecnio-CERCA Center, 25198, Lleida, Catalonia, Spain.
| |
Collapse
|
6
|
Blood mRNA biomarkers distinguish variable systemic and sputum inflammation at treatment initiation of inhaled antibiotics in cystic fibrosis: A prospective non-randomized trial. PLoS One 2022; 17:e0267592. [PMID: 35511761 PMCID: PMC9070878 DOI: 10.1371/journal.pone.0267592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 04/07/2022] [Indexed: 11/19/2022] Open
Abstract
Inhaled antibiotics control chronic airway infection and maintain respiratory health in cystic fibrosis (CF). Given variation in patient responses to inhaled antibiotics, the ability to identify distinct responder phenotypes would facilitate the delivery of personalized care. Previously, a 10-gene panel was identified, measured directly from blood leukocytes, which predicted host response to intravenous antibiotic treatment during pulmonary exacerbations. In the current study, we tested whether the same panel predicted clinical response in subjects receiving a month of inhaled antibiotic therapy with aztreonam lysine (AZLI; Cayston®). A small cohort of CF subjects infected with Pseudomonas aeruginosa were enrolled at baseline health, prior to initiating one month’s treatment with AZLI using the Altera® nebulizer system. Eighteen CF subjects underwent blood leukocyte gene panel measurements, sputum quantitative microbiology, spirometry, and C-reactive protein (CRP) measurement prior to onset and at completion of 4 weeks of AZLI therapy. Mean absolute improvement in percent predicted Forced Expiratory Volume in one second (ppFEV1) was 3%. Significant reductions in sputum bacterial colony counts were detected with treatment. CRP increased following treatment. While single genes within the panel did not change significantly following treatment, the analysis of multigene panel data demonstrated that HCA112 gene predicted ppFEV1 improvement. Hierarchical clustering based on gene expression yielded two distinctive molecular clusters before and after AZLI therapy. In conclusion, peripheral blood leukocyte genes quantifying inflammation are associated with responses to inhaled antibiotic therapy. Molecular quantification of systemic inflammation may indicate subgroups of CF subjects with variations in underlying inflammation and with variable clinical responses to inhaled antibiotics. Given the size limitation of the study, larger studies are needed in order to evaluate whether molecular measures may add precision to the determination of infectious and inflammatory outcomes following courses of inhaled antimicrobial therapies. Clinical Trials.gov Identifier: NCT01736839.
Collapse
|
7
|
TMEM176B Regulates AKT/mTOR Signaling and Tumor Growth in Triple-Negative Breast Cancer. Cells 2021; 10:cells10123430. [PMID: 34943938 PMCID: PMC8700203 DOI: 10.3390/cells10123430] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/30/2021] [Accepted: 11/30/2021] [Indexed: 11/16/2022] Open
Abstract
TMEM176B is a member of the membrane spanning 4-domains (MS4) family of transmembrane proteins, and a putative ion channel that is expressed in immune cells and certain cancers. We aimed to understand the role of TMEM176B in cancer cell signaling, gene expression, cell proliferation, and migration in vitro, as well as tumor growth in vivo. We generated breast cancer cell lines with overexpressed and silenced TMEM176B, and a therapeutic antibody targeting TMEM176B. Proliferation and migration assays were performed in vitro, and tumor growth was evaluated in vivo. We performed gene expression and Western blot analyses to identify the most differentially regulated genes and signaling pathways in cells with TMEM176B overexpression and silencing. Silencing TMEM176B or inhibiting it with a therapeutic antibody impaired cell proliferation, while overexpression increased proliferation in vitro. Syngeneic and xenograft tumor studies revealed the attenuated growth of tumors with TMEM176B gene silencing compared with controls. We found that the AKT/mTOR signaling pathway was activated or repressed in cells overexpressing or silenced for TMEM176B, respectively. Overall, our results suggest that TMEM176B expression in breast cancer cells regulates key signaling pathways and genes that contribute to cancer cell growth and progression, and is a potential target for therapeutic antibodies.
Collapse
|
8
|
Mattiola I, Mantovani A, Locati M. The tetraspan MS4A family in homeostasis, immunity, and disease. Trends Immunol 2021; 42:764-781. [PMID: 34384709 DOI: 10.1016/j.it.2021.07.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/05/2021] [Accepted: 07/07/2021] [Indexed: 01/20/2023]
Abstract
The membrane-spanning 4A (MS4A) family includes 18 members with a tetraspan structure in humans. They are differentially and selectively expressed in immunocompetent cells, such as B cells (CD20/MS4A1) and macrophages (MS4A4A), and associate with, and modulate the signaling activity of, different classes of immunoreceptor, including pattern recognition receptors (PRRs) and Ig receptors. Evidence from preclinical models and genetic evidence from humans suggest that members of the MS4A family have key roles in different pathological settings, including cancer, infectious diseases, and neurodegeneration. Therefore, MS4A family members might serve as candidate biomarkers and therapeutic targets for various conditions.
Collapse
Affiliation(s)
- Irene Mattiola
- Humanitas Clinical and Research Center IRCCS, Rozzano, Italy; Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charitè - Universitätsmedizin Berlin, Germany; Berlin Institute of Health, Berlin, Germany; Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum, Berlin, Germany.
| | - Alberto Mantovani
- Humanitas Clinical and Research Center IRCCS, Rozzano, Italy; Department of Biomedical Sciences, Humanitas University, Rozzano, Italy; The William Harvey Research Institute, Queen Mary University of London, London, UK.
| | - Massimo Locati
- Humanitas Clinical and Research Center IRCCS, Rozzano, Italy; Department of Medical Biotechnologies and Translation Medicine, University of Milan, Italy.
| |
Collapse
|
9
|
Lancien M, Bienvenu G, Salle S, Gueno L, Feyeux M, Merieau E, Remy S, Even A, Moreau A, Molle A, Fourgeux C, Coulon F, Beriou G, Bouchet-Delbos L, Chiffoleau E, Kirstetter P, Chan S, Kerfoot SM, Abdu Rahiman S, De Simone V, Matteoli G, Boncompain G, Perez F, Josien R, Poschmann J, Cuturi MC, Louvet C. Dendritic Cells Require TMEM176A/B Ion Channels for Optimal MHC Class II Antigen Presentation to Naive CD4 + T Cells. THE JOURNAL OF IMMUNOLOGY 2021; 207:421-435. [PMID: 34233909 DOI: 10.4049/jimmunol.2000498] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 05/10/2021] [Indexed: 11/19/2022]
Abstract
Intracellular ion fluxes emerge as critical actors of immunoregulation but still remain poorly explored. In this study, we investigated the role of the redundant cation channels TMEM176A and TMEM176B (TMEM176A/B) in retinoic acid-related orphan receptor γt+ cells and conventional dendritic cells (DCs) using germline and conditional double knockout mice. Although Tmem176a/b appeared surprisingly dispensable for the protective function of Th17 and group 3 innate lymphoid cells in the intestinal mucosa, we found that they were required in conventional DCs for optimal Ag processing and presentation to CD4+ T cells. Using a real-time imaging method, we show that TMEM176A/B accumulate in dynamic post-Golgi vesicles preferentially linked to the late endolysosomal system and strongly colocalize with HLA-DM. Taken together, our results suggest that TMEM176A/B ion channels play a direct role in the MHC class II compartment of DCs for the fine regulation of Ag presentation and naive CD4+ T cell priming.
Collapse
Affiliation(s)
- Melanie Lancien
- Nantes Université, CHU Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Geraldine Bienvenu
- Nantes Université, CHU Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Sonia Salle
- Nantes Université, CHU Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Lucile Gueno
- Nantes Université, CHU Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Magalie Feyeux
- Nantes Université, CHU Nantes, INSERM, CNRS, SFR Santé, FED 4203, INSERM UMS 016, CNRS UMS 3556, Nantes, France
| | - Emmanuel Merieau
- Nantes Université, CHU Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Severine Remy
- Nantes Université, CHU Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Amandine Even
- Nantes Université, CHU Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Aurelie Moreau
- Nantes Université, CHU Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Alice Molle
- Nantes Université, CHU Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Cynthia Fourgeux
- Nantes Université, CHU Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Flora Coulon
- Nantes Université, CHU Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Gaelle Beriou
- Nantes Université, CHU Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Laurence Bouchet-Delbos
- Nantes Université, CHU Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Elise Chiffoleau
- Nantes Université, CHU Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Peggy Kirstetter
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, INSERM U1258, CNRS UMR 7104, Université de Strasbourg, Illkirch, France
| | - Susan Chan
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, INSERM U1258, CNRS UMR 7104, Université de Strasbourg, Illkirch, France
| | - Steven M Kerfoot
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Saeed Abdu Rahiman
- Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium; and
| | - Veronica De Simone
- Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium; and
| | - Gianluca Matteoli
- Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium; and
| | - Gaelle Boncompain
- Dynamique de l'Organisation Intra-Cellulaire, Institut Curie, PSL Research University, Sorbonne Université, Centre National de la Recherche Scientifique, UMR 144, Paris, France
| | - Franck Perez
- Dynamique de l'Organisation Intra-Cellulaire, Institut Curie, PSL Research University, Sorbonne Université, Centre National de la Recherche Scientifique, UMR 144, Paris, France
| | - Regis Josien
- Nantes Université, CHU Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Jeremie Poschmann
- Nantes Université, CHU Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Maria Cristina Cuturi
- Nantes Université, CHU Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Cedric Louvet
- Nantes Université, CHU Nantes, INSERM, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France;
| |
Collapse
|
10
|
Han JW, Heo W, Lee D, Kang C, Kim HY, Jun I, So I, Hur H, Lee MG, Jung M, Kim JY. Plasma Membrane Localized GCaMP-MS4A12 by Orai1 Co-Expression Shows Thapsigargin- and Ca 2+-Dependent Fluorescence Increases. Mol Cells 2021; 44:223-232. [PMID: 33935043 PMCID: PMC8112172 DOI: 10.14348/molcells.2021.2031] [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] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/04/2021] [Accepted: 03/10/2021] [Indexed: 11/27/2022] Open
Abstract
Uniquely expressed in the colon, MS4A12 exhibits store-operated Ca2+ entry (SOCE) activity. However, compared to MS4A1 (CD20), a Ca2+ channel and ideal target for successful leukaemia immunotherapy, MS4A12 has rarely been studied. In this study, we investigated the involvement of MS4A12 in Ca2+ influx and expression changes in MS4A12 in human colonic malignancy. Fluorescence of GCaMP-fused MS4A12 (GCaMP-M12) was evaluated to analyse MS4A12 activity in Ca2+ influx. Plasma membrane expression of GCaMP-M12 was achieved by homo- or hetero-complex formation with no-tagged MS4A12 (nt-M12) or Orai1, respectively. GCaMP-M12 fluorescence in plasma membrane increased only after thapsigargin-induced depletion of endoplasmic reticulum Ca2+ stores, and this fluorescence was inhibited by typical SOCE inhibitors and siRNA for Orai1. Furthermore, GCaMP-MS4A12 and Orai1 co-transfection elicited greater plasma membrane fluorescence than GCaMP-M12 co-transfected with nt-M12. Interestingly, the fluorescence of GCaMP-M12 was decreased by STIM1 over-expression, while increased by siRNA for STIM1 in the presence of thapsigargin and extracellular Ca2+. Moreover, immunoprecipitation assay revealed that Orai1 co-expression decreased protein interactions between MS4A12 and STIM1. In human colon tissue, MS4A12 was expressed in the apical region of the colonic epithelium, although its expression was dramatically decreased in colon cancer tissues. In conclusion, we propose that MS4A12 contributes to SOCE through complex formation with Orai1, but does not cooperate with STIM1. Additionally, we discovered that MS4A12 is expressed in the apical membrane of the colonic epithelium and that its expression is decreased with cancer progression.
Collapse
Affiliation(s)
- Jung Woo Han
- Department of Pharmacology and Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul 03080, Korea
| | - Woon Heo
- Department of Pharmacology and Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul 03080, Korea
| | - Donghyuk Lee
- Department of Pharmacology and Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul 03080, Korea
| | - Choeun Kang
- Department of Pharmacology and Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul 03080, Korea
| | - Hye-Yeon Kim
- Department of Pharmacology and Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul 03080, Korea
| | - Ikhyun Jun
- The Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Insuk So
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Hyuk Hur
- Department of Surgery, Yonsei University College of Medicine, Seoul 03080, Korea
| | - Min Goo Lee
- Department of Pharmacology and Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul 03080, Korea
| | - Minkyu Jung
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03080, Korea
| | - Joo Young Kim
- Department of Pharmacology and Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
11
|
Men X, Su M, Ma J, Mou Y, Dai P, Chen C, Cheng XA. Overexpression of TMEM47 Induces Tamoxifen Resistance in Human Breast Cancer Cells. Technol Cancer Res Treat 2021; 20:15330338211004916. [PMID: 33745390 PMCID: PMC7989118 DOI: 10.1177/15330338211004916] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background: Tamoxifen (TAM) is the eminent first-line drug for endocrine therapy of hormone receptor positive premenopausal breast cancer and reduces the risk of recurrence by ∼50%. However, many patients developed TAM resistance and their diseases recurred. Our previous study on transcriptome profile of TAM resistant breast cancer cells revealed that the TMEM47 is one of the most significantly differentially expressed genes. The mechanism of how TMEM47 is involved in TAM resistance was not known. Methods: We constructed a mammal breast cancer cell line, in which TMEM47 was stably overexpressed (TMEM47-OE/MCF-7), to further verify the role of TMEM47 in TAM resistance. siRNA targeting TMEM47 was transfected into TAMR / MCF-7 cells by Liposome. TMEM47 expression was validated on mRNA and protein level by qRT-PCR and western blotting. We tested the cytotoxicity of TAM in the cells. Apoptosis was detected by flow cytometry. Results: Compared to the MCF7 cells, TMEM47 mRNA was significantly up regulated more than 6 folds in the TAMR/MCF7 cells and so its protein. TMEM47 expression level in TMEM47-OE/MCF-7 was similar as in the TAMR/MCF-7 cells. The 50% inhibitory concentration (IC50) value (mean ± SD) of TAM in MCF-7, TAMR/MCF-7 and TMEM47-OE/MCF-7 cells was 1.58 ± 0.19, 2.74 ± 0.24 and 3.12 ± 0.32 µγ/mL, respectively. The apoptosis rates of TAMR/MCF-7 and TMEM47-OE/MCF-7 cell lines were significantly lower than that of MCF-7 cells. After 24 and 48 hours TAM treatments, cell viability was significantly inhibitied in TMEM47 knockdown TAMR/MCF7 cells (P < 0.01). Consistant with the decreased cell viability, the apoptosis rate in TMEM47 knockdown TAMR/MCF-7 cells was significantly increased. Conclusions: Our results suggest that overexpression of TMEM47 in MCF-7 cells acquired TAM resistance to those cells, and knockdown of TMEM47 in TAMR/MCF-7 cells reversed their resistance to TAM. TMEM47 might confer TAM resistance on MCF-7 cells through the inhibition of apoptosis.
Collapse
Affiliation(s)
- Xin Men
- School of Life Sciences, Northwest University, Xi'an, Shaanxi, China.,Microbiology Institute of Shaanxi, Xi'an, Shaanxi, China
| | - Mengyang Su
- School of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Jun Ma
- Shaanxi University of Science and Technology, Xi'an, Shaanxi, China
| | - Yueyang Mou
- School of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Penggao Dai
- School of Life Sciences, Northwest University, Xi'an, Shaanxi, China.,Lifegen Co. Ltd., Xi'an, Shaanxi, China
| | - Chao Chen
- School of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Xi An Cheng
- Tongchuan people's Hospital, Tongchuan, Shaanxi, China
| |
Collapse
|
12
|
Harjula SKE, Saralahti AK, Ojanen MJT, Rantapero T, Uusi-Mäkelä MIE, Nykter M, Lohi O, Parikka M, Rämet M. Characterization of immune response against Mycobacterium marinum infection in the main hematopoietic organ of adult zebrafish (Danio rerio). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 103:103523. [PMID: 31626817 DOI: 10.1016/j.dci.2019.103523] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 10/09/2019] [Accepted: 10/14/2019] [Indexed: 06/10/2023]
Abstract
Tuberculosis remains a major global health challenge. To gain information about genes important for defense against tuberculosis, we used a well-established tuberculosis model; Mycobacterium marinum infection in adult zebrafish. To characterize the immunological response to mycobacterial infection at 14 days post infection, we performed a whole-genome level transcriptome analysis using cells from kidney, the main hematopoietic organ of adult zebrafish. Among the upregulated genes, those associated with immune signaling and regulation formed the largest category, whereas the largest group of downregulated genes had a metabolic role. We also performed a forward genetic screen in adult zebrafish and identified a fish line with severely impaired survival during chronic mycobacterial infection. Based on transcriptome analysis, these fish have decreased expression of several immunological genes. Taken together, these results give new information about the genes involved in the defense against mycobacterial infection in zebrafish.
Collapse
Affiliation(s)
- Sanna-Kaisa E Harjula
- Laboratory of Experimental Immunology, BioMediTech, Faculty of Medicine and Health Technology, FI-33014, Tampere University, Finland.
| | - Anni K Saralahti
- Laboratory of Experimental Immunology, BioMediTech, Faculty of Medicine and Health Technology, FI-33014, Tampere University, Finland.
| | - Markus J T Ojanen
- Laboratory of Experimental Immunology, BioMediTech, Faculty of Medicine and Health Technology, FI-33014, Tampere University, Finland; Laboratory of Immunoregulation, BioMediTech, Faculty of Medicine and Health Technology, FI-33014, Tampere University, Finland.
| | - Tommi Rantapero
- Laboratory of Computational Biology, BioMediTech, Faculty of Medicine and Health Technology, FI-33014, Tampere University, Finland.
| | - Meri I E Uusi-Mäkelä
- Laboratory of Experimental Immunology, BioMediTech, Faculty of Medicine and Health Technology, FI-33014, Tampere University, Finland.
| | - Matti Nykter
- Laboratory of Computational Biology, BioMediTech, Faculty of Medicine and Health Technology, FI-33014, Tampere University, Finland.
| | - Olli Lohi
- Tampere Center for Child Health Research, Tampere University and Tays Cancer Center, Tampere University Hospital, FI-33014, Tampere University, Finland.
| | - Mataleena Parikka
- Laboratory of Infection Biology, BioMediTech, Faculty of Medicine and Health Technology, FI-33014, Tampere University, Finland; Oral and Maxillofacial Unit, Tampere University Hospital, P.O. Box 2000, FI-33521, Tampere, Finland.
| | - Mika Rämet
- Laboratory of Experimental Immunology, BioMediTech, Faculty of Medicine and Health Technology, FI-33014, Tampere University, Finland; Department of Pediatrics, Tampere University Hospital, P.O. Box 2000, FI-33521, Tampere, Finland; PEDEGO Research Unit, Medical Research Center Oulu, P.O. Box 8000, FI-90014, University of Oulu, Finland; Department of Children and Adolescents, Oulu University Hospital, P.O. Box 10, FI-90029, OYS, Finland.
| |
Collapse
|
13
|
Lowe M, Lage J, Paatela E, Munson D, Hostager R, Yuan C, Katoku-Kikyo N, Ruiz-Estevez M, Asakura Y, Staats J, Qahar M, Lohman M, Asakura A, Kikyo N. Cry2 Is Critical for Circadian Regulation of Myogenic Differentiation by Bclaf1-Mediated mRNA Stabilization of Cyclin D1 and Tmem176b. Cell Rep 2019; 22:2118-2132. [PMID: 29466738 PMCID: PMC5849083 DOI: 10.1016/j.celrep.2018.01.077] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 01/09/2018] [Accepted: 01/25/2018] [Indexed: 12/30/2022] Open
Abstract
Circadian rhythms regulate cell proliferation and differentiation; however, little is known about their roles in myogenic differentiation. Our synchronized differentiation studies demonstrate that myoblast proliferation and subsequent myotube formation by cell fusion occur in circadian manners. We found that one of the core regulators of circadian rhythms, Cry2, but not Cry1, is critical for the circadian patterns of these two critical steps in myogenic differentiation. This is achieved through the specific interaction between Cry2 and Bclaf1, which stabilizes mRNAs encoding cyclin D1, a G1/S phase transition regulator, and Tmem176b, a transmembrane regulator for myogenic cell fusion. Myoblasts lacking Cry2 display premature cell cycle exit and form short myotubes because of inefficient cell fusion. Consistently, muscle regeneration is impaired in Cry2-/- mice. Bclaf1 knockdown recapitulated the phenotypes of Cry2 knockdown: early cell cycle exit and inefficient cell fusion. This study uncovers a post-transcriptional regulation of myogenic differentiation by circadian rhythms.
Collapse
Affiliation(s)
- Matthew Lowe
- Stem Cell Institute , University of Minnesota, Minneapolis, MN 55455, USA; Department of Genetics, Cell Biology, and Development , University of Minnesota, Minneapolis, MN 55455, USA
| | - Jacob Lage
- Stem Cell Institute , University of Minnesota, Minneapolis, MN 55455, USA; Department of Genetics, Cell Biology, and Development , University of Minnesota, Minneapolis, MN 55455, USA
| | - Ellen Paatela
- Stem Cell Institute , University of Minnesota, Minneapolis, MN 55455, USA
| | - Dane Munson
- Stem Cell Institute , University of Minnesota, Minneapolis, MN 55455, USA
| | - Reilly Hostager
- Stem Cell Institute , University of Minnesota, Minneapolis, MN 55455, USA
| | - Ce Yuan
- Stem Cell Institute , University of Minnesota, Minneapolis, MN 55455, USA; Bioinformatics and Computational Biology Graduate Program , University of Minnesota, Minneapolis, MN 55455, USA
| | - Nobuko Katoku-Kikyo
- Stem Cell Institute , University of Minnesota, Minneapolis, MN 55455, USA; Department of Neurology , University of Minnesota, Minneapolis, MN 55455, USA
| | - Mercedes Ruiz-Estevez
- Stem Cell Institute , University of Minnesota, Minneapolis, MN 55455, USA; Department of Genetics, Cell Biology, and Development , University of Minnesota, Minneapolis, MN 55455, USA
| | - Yoko Asakura
- Stem Cell Institute , University of Minnesota, Minneapolis, MN 55455, USA; Department of Genetics, Cell Biology, and Development , University of Minnesota, Minneapolis, MN 55455, USA
| | - James Staats
- Stem Cell Institute , University of Minnesota, Minneapolis, MN 55455, USA; Department of Genetics, Cell Biology, and Development , University of Minnesota, Minneapolis, MN 55455, USA
| | - Mulan Qahar
- Stem Cell Institute , University of Minnesota, Minneapolis, MN 55455, USA; Department of Neurology , University of Minnesota, Minneapolis, MN 55455, USA
| | - Michaela Lohman
- Stem Cell Institute , University of Minnesota, Minneapolis, MN 55455, USA
| | - Atsushi Asakura
- Stem Cell Institute , University of Minnesota, Minneapolis, MN 55455, USA; Department of Neurology , University of Minnesota, Minneapolis, MN 55455, USA.
| | - Nobuaki Kikyo
- Stem Cell Institute , University of Minnesota, Minneapolis, MN 55455, USA; Department of Genetics, Cell Biology, and Development , University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
14
|
Chang YT, Mori E, Suzuki M, Ikeda M, Huang CW, Lee JJ, Chang WN, Chang CC. APOE-MS4A genetic interactions are associated with executive dysfunction and network abnormality in clinically mild Alzheimer's disease. Neuroimage Clin 2018; 21:101621. [PMID: 30528368 PMCID: PMC6411654 DOI: 10.1016/j.nicl.2018.101621] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/17/2018] [Accepted: 12/01/2018] [Indexed: 11/09/2022]
Abstract
PURPOSE OF THE RESEARCH Although single nucleotide polymorphisms of membrane-spanning 4A (MS4A) (rs670139) and several other susceptibility genes have shown interaction effects on the risk of Alzheimer's disease (AD), little is known about the interaction effects of apolipoprotein E (APOE) with MS4A (rs670139) on cognitive performances, and the underlying pathogenesis is unclear. The study aimed to investigate the APOE-MS4A (rs670139) interaction effects on cognitive performances, cortical volumes, and functional connectivity (FC) in brain networks. PRINCIPAL RESULTS Cognitive performances were characterized in each genotypic group, and were compared between normal controls and patients in each genotypic group. APOE-MS4A interaction effects on memory and executive function scores, cortical volumes, and FC in brain networks were demonstrated. Significant effects of APOE-MS4A interactions on FC were observed in executive control network (ECN) (T maxima = 4.99, false discovery rate-corrected p < .001), the calculation score (F3, 87 = 6.218; p = .015), and the volume in prefrontal (F3, 87 = 4.374; p = .039) and orbitofrontal cortices (F3, 87 = 6.022; p = .016). The calculation score was correlated with each frontal volume (cc) (ρ = 0.304; p = .004) and genetic interaction-associated FC in ECN (ρ = 0.282; p = .008). Variations in genotypes affected the relationship between the calculation score and each frontal volume (cc). MAJOR CONCLUSIONS These findings indicate that the genetic interaction effects on FC in ECN might contribute to pathogenic mechanisms underlying the interaction effects of APOE-MS4A on calculation ability in AD.
Collapse
Affiliation(s)
- Ya-Ting Chang
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Etsuro Mori
- Department of Behavioral Neurology and Neuropsychiatry, Osaka University United Graduate School of Child Development, Suita, Osaka, Japan
| | - Maki Suzuki
- Department of Behavioral Neurology and Neuropsychiatry, Osaka University United Graduate School of Child Development, Suita, Osaka, Japan
| | - Manabu Ikeda
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Chi-Wei Huang
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Jun-Jun Lee
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Wen-Neng Chang
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
| | - Chiung-Chih Chang
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan.
| |
Collapse
|
15
|
Zhao J, Zhu D, Zhang X, Zhang Y, Zhou J, Dong M. TMEM206 promotes the malignancy of colorectal cancer cells by interacting with AKT and extracellular signal‐regulated kinase signaling pathways. J Cell Physiol 2018; 234:10888-10898. [DOI: 10.1002/jcp.27751] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 10/24/2018] [Indexed: 12/24/2022]
Affiliation(s)
- Jinbo Zhao
- Department of Gastrointestinal Surgery The First Affiliated Hospital of China Medical University Shenyang China
| | - Dehua Zhu
- Department of Gastrointestinal Surgery The First Affiliated Hospital of China Medical University Shenyang China
| | - Xiupeng Zhang
- Department of Pathology The First Affiliated Hospital of China Medical University Shenyang China
| | - Yong Zhang
- Department of Pathology Liaoning Provincial People's Hospital China Medical University Shenyang China
| | - Jianping Zhou
- Department of Gastrointestinal Surgery The First Affiliated Hospital of China Medical University Shenyang China
| | - Ming Dong
- Department of Gastrointestinal Surgery The First Affiliated Hospital of China Medical University Shenyang China
| |
Collapse
|
16
|
Ma J, Zhang W, Tan L, Wang HF, Wan Y, Sun FR, Tan CC, Yu JT, Tan L, Alzheimer's Disease Neuroimaging Initiative. MS4A6A genotypes are associated with the atrophy rates of Alzheimer's disease related brain structures. Oncotarget 2018; 7:58779-58788. [PMID: 27244883 PMCID: PMC5312275 DOI: 10.18632/oncotarget.9563] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Accepted: 04/26/2016] [Indexed: 11/25/2022] Open
Abstract
Membrane-spanning 4-domains, subfamily A, member 6A (MS4A6A) has been identified as susceptibility loci of Alzheimer's disease (AD) by several recent genome-wide association studies (GWAS), whereas little is known about the potential roles of these variants in the brain structure and function of AD. In this study, we included a total of 812 individuals from the Alzheimer's disease Neuroimaging Initiative (ADNI) database. Using multiple linear regression models, we found MS4A6A genotypes were strongly related to atrophy rate of left middle temporal (rs610932: Pc = 0.017, rs7232: Pc = 0.022), precuneus (rs610932: Pc = 0.015) and entorhinal (rs610932, Pc = 0.022) on MRI in the entire group. In the subgroup analysis, MS4A6A SNPs were significantly accelerated the percentage of volume loss of middle temporal, precuneus and entorhinal, especially in the MCI subgroup. These findings reveal that MS4A6A genotypes affect AD specific brain structures which supported the possible role of MS4A6A polymorphisms in influencing AD-related neuroimaging phenotypes.
Collapse
Affiliation(s)
- Jing Ma
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China
| | - Wei Zhang
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China
| | - Lin Tan
- College of Medicine and Pharmaceutics, Ocean University of China, Qingdao, China
| | - Hui-Fu Wang
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China
| | - Yu Wan
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China
| | - Fu-Rong Sun
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China
| | - Chen-Chen Tan
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China.,College of Medicine and Pharmaceutics, Ocean University of China, Qingdao, China
| | | |
Collapse
|
17
|
|
18
|
Kaneko T, Toshimori K, Iida H. Subcellular localization of MS4A13 isoform 2 in mouse spermatozoa. Reproduction 2017; 154:843-857. [PMID: 28971897 DOI: 10.1530/rep-17-0477] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/11/2017] [Accepted: 10/02/2017] [Indexed: 01/02/2023]
Abstract
To identify upregulated genes during the development of spermatozoa, we performed PCR-selected subtraction analysis of testes RNA samples from 10-day-old and 12-week-old shrews. A transcript, highly homologous to two mouse transcripts, Ms4a13-1 and Ms4a13-2, was differentially regulated. Ms4a13-2, but not Ms4a13-1, was shown to be primarily expressed in mouse testes in an age-dependent manner. Ms4a13-2 cDNA contains an open-reading frame of 522 nucleotides, encoding a protein of 174 amino acids, with predicted molecular mass, 19,345 Da. MS4A13-2 protein was expressed along the periphery of nuclei of round and elongated spermatids (steps 3-16) in adult mouse testes, and in the equatorial region of the heads of fresh mature mouse spermatozoa. In addition, MS4A13-2 was found to localize to the outer acrosomal membrane in the equatorial region of heads in fresh spermatozoa. In acrosome-reacted spermatozoa, the MS4A13-2 expression extended to the entire sperm head including the postacrosomal region and acrosomal cap. MS4A family proteins are known to facilitate intracellular protein-protein interactions as ion channel/adaptor proteins by oligomerization, and have important regulatory roles in cellular growth, survival and activation. We report that the MS4A family member, MS4A13-2, may form oligomers in sperm membranes, which may be involved in an interaction with the zona pellucida or cumulus during fertilization.
Collapse
Affiliation(s)
- Takane Kaneko
- Laboratory of ZoologyGraduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Kiyotaka Toshimori
- Future Medicine Research Center and Department of Reproductive Biology and MedicineGraduate School of Medicine, Chiba University, Chiba, Japan
| | - Hiroshi Iida
- Laboratory of ZoologyGraduate School of Agriculture, Kyushu University, Fukuoka, Japan
| |
Collapse
|
19
|
Genetics of Alzheimer's disease: From pathogenesis to clinical usage. J Clin Neurosci 2017; 45:1-8. [PMID: 28869135 DOI: 10.1016/j.jocn.2017.06.074] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 06/19/2017] [Indexed: 01/27/2023]
Abstract
Alzheimer's disease (AD) is the most common type of dementia and has caused a major global health concern. Understanding the etiology of AD can be beneficial for the diagnosis and intervention of this disease. Genetics plays a vital role in the pathogenesis of AD. Research methods in genetics such as the linkage analysis, study of candidate genes, genome-wide association study (GWAS), and next-generation sequencing (NGS) technology help us map the genetic information in AD, which can not only provide a new insight into the pathogenesis of AD but also be beneficial for early targeted intervention of AD. This review summarizes the pathogenesis as well as the diagnostic and therapeutic value of genetics in AD.
Collapse
|
20
|
Sanyal R, Polyak MJ, Zuccolo J, Puri M, Deng L, Roberts L, Zuba A, Storek J, Luider JM, Sundberg EM, Mansoor A, Baigorri E, Chu MP, Belch AR, Pilarski LM, Deans JP. MS4A4A: a novel cell surface marker for M2 macrophages and plasma cells. Immunol Cell Biol 2017; 95:611-619. [PMID: 28303902 DOI: 10.1038/icb.2017.18] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 02/24/2017] [Accepted: 03/13/2017] [Indexed: 01/05/2023]
Abstract
MS4A4A is a member of the membrane-spanning, four domain family, subfamily A (MS4A) that includes CD20 (MS4A1), FcRβ (MS4A2) and Htm4 (MS4A3). Like the first three members of this family, transcription of MS4A4A appears to be limited to hematopoietic cells. To evaluate expression of the MS4A4A protein in hematopoietic cell lineages and subsets we generated monoclonal antibodies against extracellular epitopes for use in flow cytometry. In human peripheral blood we found that MS4A4A is expressed at the plasma membrane in monocytes but not in granulocytes or lymphocytes. In vitro differentiation of monocytes demonstrated that MS4A4A is expressed in immature but not activated dendritic cells, and in macrophages generated in the presence of interleukin-4 ('alternatively activated' or M2 macrophages) but not by interferon-γ and lipopolysaccharide ('classically' activated or M1 macrophages). MS4A4A was expressed in the U937 monocytic cell line only after differentiation. In normal bone marrow, MS4A4A was expressed in mature monocytes but was undetected, or detected at only a low level, in myeloid/monocytic precursors, as well as their malignant counterparts in patients with various subtypes of myeloid leukemia. Although MS4A4A was not expressed in healthy B lymphocytes, it was highly expressed in normal plasma cells, CD138+ cells from multiple myeloma patients, and bone marrow B cells from a patient with mantle cell lymphoma. These findings suggest immunotherapeutic potential for MS4A4A antibodies in targeting alternatively activated macrophages such as tumor-associated macrophages, and in the treatment of multiple myeloma and mantle cell lymphoma.
Collapse
Affiliation(s)
- Ratna Sanyal
- Department of Biochemistry and Molecular Biology, and Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Maria J Polyak
- Department of Biochemistry and Molecular Biology, and Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Jonathan Zuccolo
- Department of Biochemistry and Molecular Biology, and Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Mandip Puri
- Department of Biochemistry and Molecular Biology, and Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Lili Deng
- Department of Biochemistry and Molecular Biology, and Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Luc Roberts
- Department of Biochemistry and Molecular Biology, and Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Ania Zuba
- Department of Biochemistry and Molecular Biology, and Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Jan Storek
- Departments of Medicine and Oncology, University of Calgary, Calgary, Alberta, Canada
| | - Joanne M Luider
- Calgary Laboratory Services, Foothills Medical Centre, Calgary, Alberta, Canada
| | - Ellen M Sundberg
- Calgary Laboratory Services, Foothills Medical Centre, Calgary, Alberta, Canada
| | - Adnan Mansoor
- Calgary Laboratory Services, Foothills Medical Centre, Calgary, Alberta, Canada.,Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Eva Baigorri
- Department of Oncology, University of Alberta and Cross Cancer Institute, Edmonton, Alberta, Canada
| | - Michael P Chu
- Department of Oncology, University of Alberta and Cross Cancer Institute, Edmonton, Alberta, Canada
| | - Andrew R Belch
- Department of Oncology, University of Alberta and Cross Cancer Institute, Edmonton, Alberta, Canada
| | - Linda M Pilarski
- Department of Oncology, University of Alberta and Cross Cancer Institute, Edmonton, Alberta, Canada
| | - Julie P Deans
- Department of Biochemistry and Molecular Biology, and Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
21
|
Pozhitkov AE, Neme R, Domazet-Lošo T, Leroux BG, Soni S, Tautz D, Noble PA. Tracing the dynamics of gene transcripts after organismal death. Open Biol 2017; 7:160267. [PMID: 28123054 PMCID: PMC5303275 DOI: 10.1098/rsob.160267] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 12/12/2016] [Indexed: 12/13/2022] Open
Abstract
In life, genetic and epigenetic networks precisely coordinate the expression of genes-but in death, it is not known if gene expression diminishes gradually or abruptly stops or if specific genes and pathways are involved. We studied this by identifying mRNA transcripts that apparently increase in relative abundance after death, assessing their functions, and comparing their abundance profiles through postmortem time in two species, mouse and zebrafish. We found mRNA transcript profiles of 1063 genes became significantly more abundant after death of healthy adult animals in a time series spanning up to 96 h postmortem. Ordination plots revealed non-random patterns in the profiles by time. While most of these transcript levels increased within 0.5 h postmortem, some increased only at 24 and 48 h postmortem. Functional characterization of the most abundant transcripts revealed the following categories: stress, immunity, inflammation, apoptosis, transport, development, epigenetic regulation and cancer. The data suggest a step-wise shutdown occurs in organismal death that is manifested by the apparent increase of certain transcripts with various abundance maxima and durations.
Collapse
Affiliation(s)
- Alex E Pozhitkov
- Department of Oral Health Sciences, University of Washington, PO Box 357444, Seattle, WA 98195, USA
- Max Planck Institute for Evolutionary Biology, August-Thienemann-Strasse 2, 24306 Ploen, Germany
| | - Rafik Neme
- Max Planck Institute for Evolutionary Biology, August-Thienemann-Strasse 2, 24306 Ploen, Germany
| | - Tomislav Domazet-Lošo
- Laboratory of Evolutionary Genetics, Division of Molecular Biology, Ruđer Bošković Institute, 10002 Zagreb, Croatia
- Catholic University of Croatia, Ilica 242, Zagreb, Croatia
| | - Brian G Leroux
- Department of Oral Health Sciences, University of Washington, PO Box 357444, Seattle, WA 98195, USA
| | - Shivani Soni
- Department of Biological Sciences, Alabama State University, Montgomery, AL 36101-0271, USA
| | - Diethard Tautz
- Max Planck Institute for Evolutionary Biology, August-Thienemann-Strasse 2, 24306 Ploen, Germany
| | - Peter A Noble
- Department of Periodontics, University of Washington, PO Box 357444, Seattle, WA 98195, USA
- Department of Biological Sciences, Alabama State University, Montgomery, AL 36101-0271, USA
- PhD Program in Microbiology, Alabama State University, Montgomery, AL 36101-0271, USA
| |
Collapse
|
22
|
Grunin M, Hagbi-Levi S, Rinsky B, Smith Y, Chowers I. Transcriptome Analysis on Monocytes from Patients with Neovascular Age-Related Macular Degeneration. Sci Rep 2016; 6:29046. [PMID: 27374485 PMCID: PMC4931446 DOI: 10.1038/srep29046] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 06/10/2016] [Indexed: 01/09/2023] Open
Abstract
Mononuclear phagocytes (MPs), including monocytes/macrophages, play complex roles in age-related macular degeneration (AMD) pathogenesis. We reported altered gene-expression signature in peripheral blood mononuclear cells from AMD patients, and a chemokine receptor signature on AMD monocytes. To obtain comprehensive understanding of MP involvement, particularly in peripheral circulation in AMD, we performed global gene expression analysis in monocytes. We separated monocytes from treatment-naïve neovascular AMD (nvAMD) patients (n = 14) and age-matched controls (n = 15), and performed microarray and bioinformatics analysis. Quantitative real-time PCR was performed on other sets of nvAMD (n = 25), atrophic AMD (n = 21), and controls (n = 28) for validation. This validated microarray genes (like TMEM176A/B and FOSB) tested, including differences between nvAMD and atrophic AMD. We identified 2,165 differentially-expressed genes (P < 0.05), including 79 genes with log2 fold change ≥1.5 between nvAMD and controls. Functional annotation using DAVID and TANGO demonstrated immune response alterations in AMD monocytes (FDR-P <0.05), validated by randomized data comparison (P < 0.0001). GSEA, ISMARA, and MEME analysis found immune enrichment and specific involved microRNAs. Enrichment of differentially-expressed genes in monocytes was found in retina via SAGE data-mining. These genes were enriched in non-classical vs. classical monocyte subsets (P < 0.05). Therefore, global gene expression analysis in AMD monocytes reveals an altered immune-related signature, further implicating systemic MP activation in AMD.
Collapse
Affiliation(s)
- Michelle Grunin
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Shira- Hagbi-Levi
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Batya Rinsky
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Yoav Smith
- Genomic Data Analysis Unit, Hebrew University, Jerusalem, Israel
| | - Itay Chowers
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
23
|
Wu D, Xi QY, Cheng X, Dong T, Zhu XT, Shu G, Wang LN, Jiang QY, Zhang YL. miR-146a-5p inhibits TNF-α-induced adipogenesis via targeting insulin receptor in primary porcine adipocytes. J Lipid Res 2016; 57:1360-72. [PMID: 27324794 PMCID: PMC4959853 DOI: 10.1194/jlr.m062497] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Indexed: 02/05/2023] Open
Abstract
TNF-α is a multifunctional cytokine participating in immune disorders, inflammation, and tumor development with regulatory effects on energy metabolism. Our work focused on the function of TNF-α in adipogenesis of primary porcine adipocytes. TNF-α could suppress the insulin receptor (IR) at the mRNA and protein levels. Microarray analysis of TNF-α-treated porcine adipocytes was used to screen out 29 differentially expressed microRNAs (miRNAs), 13 of which were remarkably upregulated and 16 were intensely downregulated. These 29 differentially expressed miRNAs were predicted to mainly participate in the insulin signaling pathway, adipocytokine signaling pathway, and type 2 diabetes mellitus pathway by Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses. miR-146a-5p, reportedly involved in immunity and cancer relevant processes, was one of the most highly differentially expressed miRNAs after TNF-α treatment. Red Oil O staining and TG assay revealed that miR-146a-5p suppressed adipogenesis. A dual-luciferase reporter and siRNA assay verified that miR-146a-5p targeted IR and could inhibit its protein expression. miR-146a-5p was also validated to be involved in the insulin signaling pathway by reducing tyrosine phosphorylation of insulin receptor substrate-1. Our study provides the first evidence of miR-146a-5p targeting IR, which facilitates future studies related to obesity and diabetes using pig models.
Collapse
Affiliation(s)
- Di Wu
- National Engineering Research Center for Swine Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agriculture University, Guangzhou, China, 510642
| | - Qian-Yun Xi
- National Engineering Research Center for Swine Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agriculture University, Guangzhou, China, 510642
| | - Xiao Cheng
- National Engineering Research Center for Swine Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agriculture University, Guangzhou, China, 510642
| | - Tao Dong
- National Engineering Research Center for Swine Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agriculture University, Guangzhou, China, 510642
| | - Xiao-Tong Zhu
- National Engineering Research Center for Swine Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agriculture University, Guangzhou, China, 510642
| | - Gang Shu
- National Engineering Research Center for Swine Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agriculture University, Guangzhou, China, 510642
| | - Li-Na Wang
- National Engineering Research Center for Swine Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agriculture University, Guangzhou, China, 510642
| | - Qing-Yan Jiang
- National Engineering Research Center for Swine Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agriculture University, Guangzhou, China, 510642
| | - Yong-Liang Zhang
- National Engineering Research Center for Swine Breeding Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agriculture University, Guangzhou, China, 510642
| |
Collapse
|
24
|
Greer PL, Bear DM, Lassance JM, Bloom ML, Tsukahara T, Pashkovski SL, Masuda FK, Nowlan AC, Kirchner R, Hoekstra HE, Datta SR. A Family of non-GPCR Chemosensors Defines an Alternative Logic for Mammalian Olfaction. Cell 2016; 165:1734-1748. [PMID: 27238024 DOI: 10.1016/j.cell.2016.05.001] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 03/14/2016] [Accepted: 04/28/2016] [Indexed: 10/21/2022]
Abstract
Odor perception in mammals is mediated by parallel sensory pathways that convey distinct information about the olfactory world. Multiple olfactory subsystems express characteristic seven-transmembrane G-protein-coupled receptors (GPCRs) in a one-receptor-per-neuron pattern that facilitates odor discrimination. Sensory neurons of the "necklace" subsystem are nestled within the recesses of the olfactory epithelium and detect diverse odorants; however, they do not express known GPCR odor receptors. Here, we report that members of the four-pass transmembrane MS4A protein family are chemosensors expressed within necklace sensory neurons. These receptors localize to sensory endings and confer responses to ethologically relevant ligands, including pheromones and fatty acids, in vitro and in vivo. Individual necklace neurons co-express many MS4A proteins and are activated by multiple MS4A ligands; this pooling of information suggests that the necklace is organized more like subsystems for taste than for smell. The MS4As therefore define a distinct mechanism and functional logic for mammalian olfaction.
Collapse
Affiliation(s)
- Paul L Greer
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Daniel M Bear
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Jean-Marc Lassance
- Departments of Molecular and Cellular Biology and Organismic and Evolutionary Biology, Center for Brain Science, Harvard University, Howard Hughes Medical Institute, Cambridge, MA 02138, USA
| | | | - Tatsuya Tsukahara
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Stan L Pashkovski
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Francis Kei Masuda
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Alexandra C Nowlan
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Rory Kirchner
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Hopi E Hoekstra
- Departments of Molecular and Cellular Biology and Organismic and Evolutionary Biology, Center for Brain Science, Harvard University, Howard Hughes Medical Institute, Cambridge, MA 02138, USA
| | | |
Collapse
|
25
|
Lemoine A, Chauveau-Le Friec G, Langa F, Louvet C. Generation of a Double KO Mouse by Simultaneous Targeting of the Neighboring Genes Tmem176a and Tmem176b Using CRISPR/Cas9: Key Steps from Design to Genotyping. J Genet Genomics 2016; 43:329-40. [DOI: 10.1016/j.jgg.2016.04.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 03/26/2016] [Accepted: 04/04/2016] [Indexed: 01/16/2023]
|
26
|
RORγt+ cells selectively express redundant cation channels linked to the Golgi apparatus. Sci Rep 2016; 6:23682. [PMID: 27009467 PMCID: PMC4806298 DOI: 10.1038/srep23682] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 03/08/2016] [Indexed: 01/11/2023] Open
Abstract
Retinoid-related orphan receptor gamma t (RORγt) is a master transcription factor central to type 17 immunity involving cells such as T helper 17, group 3 innate lymphoid cells or IL-17-producing γδ T cells. Here we show that the intracellular ion channel TMEM176B and its homologue TMEM176A are strongly expressed in these RORγt+ cells. We demonstrate that TMEM176A and B exhibit a similar cation channel activity and mainly colocalise in close proximity to the trans-Golgi network. Strikingly, in the mouse, the loss of Tmem176b is systematically associated with a strong upregulation of Tmem176a. While Tmem176b single-deficiency has no effect on the course of experimental autoimmune encephalomyelitis, T cell or DSS-induced colitis, it significantly reduces imiquimod-induced psoriasis-like skin inflammation. These findings shed light on a potentially novel specific process linked to post-Golgi trafficking for modulating the function of RORγt+ cells and indicate that both homologues should be simultaneously targeted to clearly elucidate the role of this intracellular ion flow.
Collapse
|
27
|
Khan N. Comparative and evolutionary insights into CD4 gene across mammalian and avian taxa. Interv Med Appl Sci 2016; 7:152-60. [PMID: 26767121 DOI: 10.1556/1646.7.2015.4.4] [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: 11/19/2022] Open
Abstract
The present day genetic architecture of a species bears much significance to its closely related species which is due to species-specific differences, shaped by different evolutionary forces across time scale. With the availability of whole genome sequence of several closely related species, it is now possible to infer evolutionary patterns of genes and genomes in specific lineages. To this respect, CD4 gene, primarily responsible for defensive mechanism in human, is conserved across a few taxa, and thus, comparative genomic studies could be useful for better understanding of host-pathogen biology. Comparative and evolutionary analyses were performed in eleven taxa (10 mammalian and avian) with different statistical algorithms. Phylogenetic inferences revealed recent divergence of human and chimpanzee, and pig was found to be diverged from rest of the taxa significantly. Additionally, gene length, microsatellites, and secondary structures were observed across taxa. The genetic architecture of CD4 gene and its evolutionary history in different mammalian taxa provide crucial evidence in support of the fact that this gene might have been evolving at a similar rate to other human immune system genes. Future population-based study and structural modeling would unravel the differential ability to interact with HIV virus and influence immune system in humans.
Collapse
Affiliation(s)
- Naazneen Khan
- Department of Biotechnology, Jamia Millia Islamia University , Jamia Nagar, Delhi , India
| |
Collapse
|
28
|
A novel Alzheimer disease locus located near the gene encoding tau protein. Mol Psychiatry 2016; 21:108-17. [PMID: 25778476 PMCID: PMC4573764 DOI: 10.1038/mp.2015.23] [Citation(s) in RCA: 203] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Revised: 12/05/2014] [Accepted: 01/08/2015] [Indexed: 12/11/2022]
Abstract
APOE ɛ4, the most significant genetic risk factor for Alzheimer disease (AD), may mask effects of other loci. We re-analyzed genome-wide association study (GWAS) data from the International Genomics of Alzheimer's Project (IGAP) Consortium in APOE ɛ4+ (10 352 cases and 9207 controls) and APOE ɛ4- (7184 cases and 26 968 controls) subgroups as well as in the total sample testing for interaction between a single-nucleotide polymorphism (SNP) and APOE ɛ4 status. Suggestive associations (P<1 × 10(-4)) in stage 1 were evaluated in an independent sample (stage 2) containing 4203 subjects (APOE ɛ4+: 1250 cases and 536 controls; APOE ɛ4-: 718 cases and 1699 controls). Among APOE ɛ4- subjects, novel genome-wide significant (GWS) association was observed with 17 SNPs (all between KANSL1 and LRRC37A on chromosome 17 near MAPT) in a meta-analysis of the stage 1 and stage 2 data sets (best SNP, rs2732703, P=5·8 × 10(-9)). Conditional analysis revealed that rs2732703 accounted for association signals in the entire 100-kilobase region that includes MAPT. Except for previously identified AD loci showing stronger association in APOE ɛ4+ subjects (CR1 and CLU) or APOE ɛ4- subjects (MS4A6A/MS4A4A/MS4A6E), no other SNPs were significantly associated with AD in a specific APOE genotype subgroup. In addition, the finding in the stage 1 sample that AD risk is significantly influenced by the interaction of APOE with rs1595014 in TMEM106B (P=1·6 × 10(-7)) is noteworthy, because TMEM106B variants have previously been associated with risk of frontotemporal dementia. Expression quantitative trait locus analysis revealed that rs113986870, one of the GWS SNPs near rs2732703, is significantly associated with four KANSL1 probes that target transcription of the first translated exon and an untranslated exon in hippocampus (P ⩽ 1.3 × 10(-8)), frontal cortex (P ⩽ 1.3 × 10(-9)) and temporal cortex (P⩽1.2 × 10(-11)). Rs113986870 is also strongly associated with a MAPT probe that targets transcription of alternatively spliced exon 3 in frontal cortex (P=9.2 × 10(-6)) and temporal cortex (P=2.6 × 10(-6)). Our APOE-stratified GWAS is the first to show GWS association for AD with SNPs in the chromosome 17q21.31 region. Replication of this finding in independent samples is needed to verify that SNPs in this region have significantly stronger effects on AD risk in persons lacking APOE ɛ4 compared with persons carrying this allele, and if this is found to hold, further examination of this region and studies aimed at deciphering the mechanism(s) are warranted.
Collapse
|
29
|
Villegas-Llerena C, Phillips A, Garcia-Reitboeck P, Hardy J, Pocock JM. Microglial genes regulating neuroinflammation in the progression of Alzheimer's disease. Curr Opin Neurobiol 2015; 36:74-81. [PMID: 26517285 DOI: 10.1016/j.conb.2015.10.004] [Citation(s) in RCA: 191] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 09/24/2015] [Accepted: 10/07/2015] [Indexed: 01/09/2023]
Abstract
Neuroinflammation is a pathological hallmark of Alzheimer's disease (AD), and microglia, the brain's resident phagocyte, are pivotal for the immune response observed in AD. Microglia act as sentinel and protective cells, but may become inappropriately reactive in AD to drive neuropathology. Recent Genome Wide Association Studies (GWAS) have identified more than 20 gene variants associated with an increased risk of late-onset AD (LOAD), the most prevalent form of AD [1]. The findings strongly implicate genes related to the immune response (CR1, CD33, MS4A, CLU, ABCA7, EPHA1 and HLA-DRB5-HLA-DRB1), endocytosis (BIN1, PICALM, CD2AP, EPHA1 and SORL1) and lipid biology (CLU, ABCA7 and SORL1) [2-8], and many encode proteins which are highly expressed in microglia [1]. Furthermore, recent identification of a low frequency mutation in the gene encoding the triggering receptor expressed in myeloid cells 2 protein (TREM2) confers increased risk of AD in LOAD cohorts with an effect size similar to that for APOE, until recently the only identified genetic risk factor associated with LOAD [9,10(••)] (Figure 1). The present review summarises our current understanding of the probable roles of microglial genes in the regulation of neuroinflammatory processes in AD and their relation to other processes affecting the disease's progression.
Collapse
Affiliation(s)
- Claudio Villegas-Llerena
- Department of Neuroinflammation, University College London, Institute of Neurology, 1 Wakefield Street, London WC1 N 1PK, UK; Department of Molecular Neuroscience, University College London, Institute of Neurology, 1 Wakefield Street, London WC1 N 1PK, UK
| | - Alexandra Phillips
- Department of Neuroinflammation, University College London, Institute of Neurology, 1 Wakefield Street, London WC1 N 1PK, UK
| | - Pablo Garcia-Reitboeck
- Department of Neuroinflammation, University College London, Institute of Neurology, 1 Wakefield Street, London WC1 N 1PK, UK; Department of Molecular Neuroscience, University College London, Institute of Neurology, 1 Wakefield Street, London WC1 N 1PK, UK
| | - John Hardy
- Department of Molecular Neuroscience, University College London, Institute of Neurology, 1 Wakefield Street, London WC1 N 1PK, UK
| | - Jennifer M Pocock
- Department of Neuroinflammation, University College London, Institute of Neurology, 1 Wakefield Street, London WC1 N 1PK, UK.
| |
Collapse
|
30
|
Cuajungco MP, Silva J, Habibi A, Valadez JA. The mucolipin-2 (TRPML2) ion channel: a tissue-specific protein crucial to normal cell function. Pflugers Arch 2015; 468:177-92. [PMID: 26336837 DOI: 10.1007/s00424-015-1732-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 08/25/2015] [Accepted: 08/27/2015] [Indexed: 12/26/2022]
Abstract
The discovery of the TRPML subfamily of ion channels has created an exciting niche in the fields of membrane trafficking, signal transduction, autophagy, and metal homeostasis. The TRPML protein subfamily consists of three members, TRPML1, TRPML2, and TRPML3, which are encoded by MCOLN1, MCOLN2, and MCOLN3 genes, respectively. They are non-selective cation channels with six predicted transmembrane domains and intracellular amino- and carboxyl-terminus regions. They localize to the plasma membrane, endosomes, and lysosomes of cells. TRPML1 is associated with the human lysosomal storage disease known as mucolipidosis type IV (MLIV), but TRPML2 and TRPML3 have not been linked with a human disease. Although TRPML1 is expressed in many tissues, TRPML3 is expressed in a varied but limited set of tissues, while TRPML2 has a more limited expression pattern where it is mostly detected in lymphoid and myeloid tissues. This review focuses on TRPML2 because it appears to play an important, yet unrecognized role in the immune system. While the evidence has been mostly indirect, we present and discuss relevant data that strengthen the connection of TRPML2 with cellular immunity. We also discuss the functional redundancy between the TRPML proteins, and how such features could be exploited as a potential therapeutic strategy for MLIV disease. We present evidence that TRPML2 expression may complement certain phenotypic alterations in MLIV cells and briefly examine the challenges of functional complementation. In conclusion, the function of TRPML2 still remains obscure, but emerging data show that it may serve a critical role in immune cell development and inflammatory responses.
Collapse
Affiliation(s)
- Math P Cuajungco
- Department of Biological Science, California State University Fullerton, 800 N. State College Blvd., Fullerton, CA, 92831, USA. .,Center for Applied Biotechnology Studies, California State University Fullerton, Fullerton, CA, 92831, USA.
| | - Joshua Silva
- Department of Biological Science, California State University Fullerton, 800 N. State College Blvd., Fullerton, CA, 92831, USA
| | - Ania Habibi
- Department of Biological Science, California State University Fullerton, 800 N. State College Blvd., Fullerton, CA, 92831, USA
| | - Jessica A Valadez
- Department of Biological Science, California State University Fullerton, 800 N. State College Blvd., Fullerton, CA, 92831, USA
| |
Collapse
|
31
|
Abstract
Alzheimer's disease (AD) represents the main form of dementia, and is a major public health problem. Despite intensive research efforts, current treatments have only marginal symptomatic benefits and there are no effective disease-modifying or preventive interventions. AD has a strong genetic component, so much research in AD has focused on identifying genetic causes and risk factors. This chapter will cover genetic discoveries in AD and their consequences in terms of improved knowledge regarding the disease and the identification of biomarkers and drug targets. First, we will discuss the study of the rare early-onset, autosomal dominant forms of AD that led to the discovery of mutations in three major genes, APP, PSEN1, and PSEN2. These discoveries have shaped our current understanding of the pathophysiology and natural history of AD as well as the development of therapeutic targets and the design of clinical trials. Then, we will explore linkage analysis and candidate gene approaches, which identified variants in Apolipoprotein E (APOE) as the major genetic risk factor for late-onset, "sporadic" forms of AD (LOAD), but failed to robustly identify other genetic risk factors, with the exception of variants in SORL1. The main focus of this chapter will be on recent genome-wide association studies that have successfully identified common genetic variations at over 20 loci associated with LOAD outside of the APOE locus. These loci are in or near-novel AD genes including BIN1, CR1, CLU, phosphatidylinositol-binding clathrin assembly protein (PICALM), CD33, EPHA1, MS4A4/MS4A6, ABCA7, CD2AP, SORL1, HLA-DRB5/DRB1, PTK2B, SLC24A4-RIN3, INPP5D, MEF2C, NME8, ZCWPW1, CELF1, FERMT2, CASS4, and TRIP4 and each has small effects on risk of AD (relative risks of 1.1-1.3). Finally, we will touch upon the ongoing effort to identify less frequent and rare variants through whole exome and whole genome sequencing. This effort has identified two novel genes, TREM2 and PLD3, and shown a role for APP in LOAD. The identification of these recently identified genes has implicated previously unsuspected biological pathways in the pathophysiology of AD.
Collapse
Affiliation(s)
- Vincent Chouraki
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA; Framingham Heart Study, Framingham, MA, USA
| | - Sudha Seshadri
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA; Framingham Heart Study, Framingham, MA, USA
| |
Collapse
|
32
|
The MS4A family: counting past 1, 2 and 3. Immunol Cell Biol 2015; 94:11-23. [PMID: 25835430 DOI: 10.1038/icb.2015.48] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 03/27/2015] [Accepted: 03/28/2015] [Indexed: 02/01/2023]
Abstract
The MS4A (membrane-spanning 4-domain family, subfamily A) family of proteins contains some well-known members including MS4A1 (CD20), MS4A2 (FcɛRIβ) and MS4A3 (HTm4). These three MS4A family members are expressed on the cell surface of specific leukocyte subsets and have been well characterized as having key roles in regulating cell activation, growth and development. However, beyond MS4A1-3 there are a large number of related molecules (18 to date in humans) where our understanding of their biological roles is at a relatively nascent stage. This review examines the larger MS4A family focusing on their structure, expression, regulation and characterized and/or emerging biological roles. Our own work on one family member MS4A8B, and its possible role in epithelial cell regulation, is also highlighted.
Collapse
|
33
|
Karch CM, Goate AM. Alzheimer's disease risk genes and mechanisms of disease pathogenesis. Biol Psychiatry 2015; 77:43-51. [PMID: 24951455 PMCID: PMC4234692 DOI: 10.1016/j.biopsych.2014.05.006] [Citation(s) in RCA: 905] [Impact Index Per Article: 90.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 04/30/2014] [Accepted: 05/05/2014] [Indexed: 01/18/2023]
Abstract
We review the genetic risk factors for late-onset Alzheimer's disease (AD) and their role in AD pathogenesis. More recent advances in understanding of the human genome-technologic advances in methods to analyze millions of polymorphisms in thousands of subjects-have revealed new genes associated with AD risk, including ABCA7, BIN1, CASS4, CD33, CD2AP, CELF1, CLU, CR1, DSG2, EPHA1, FERMT2, HLA-DRB5-DBR1, INPP5D, MS4A, MEF2C, NME8, PICALM, PTK2B, SLC24H4-RIN3, SORL1, and ZCWPW1. Emerging technologies to analyze the entire genome in large data sets have also revealed coding variants that increase AD risk: PLD3 and TREM2. We review the relationship between these AD risk genes and the cellular and neuropathologic features of AD. Understanding the mechanisms underlying the association of these genes with risk for disease will provide the most meaningful targets for therapeutic development to date.
Collapse
Affiliation(s)
| | - Alison M. Goate
- Corresponding author Contact information: Department of Psychiatry, Washington University School of Medicine, 425 S. Euclid Ave, Campus Box 8134, St. Louis, MO 63110, phone: 314-362-8691, fax: 314-747-2983,
| |
Collapse
|
34
|
Ma J, Yu JT, Tan L. MS4A Cluster in Alzheimer's Disease. Mol Neurobiol 2014; 51:1240-8. [PMID: 24981432 DOI: 10.1007/s12035-014-8800-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Accepted: 06/22/2014] [Indexed: 01/13/2023]
Abstract
Several variants within membrane-spanning 4-domains subfamily A (MS4A) gene cluster have recently been implicated the association of Alzheimer's disease (AD) by serial recent genome-wide association studies (GWAS). As cell membrane proteins, MS4A family members are found to participate in the regulation of calcium signaling which have been widely discussed in neurodegeneration and AD. Besides, although the MS4A family members are poorly characterized, an important role in immunity has already been identified for several members of this cluster (such as MS4A1, MS4A2, and MS4A4B), indicating the possible involvement of MS4A gene cluster in AD pathogenesis. In this article, we briefly summarize the structure, localization, and function of MS4A gene cluster, review recent genetic and expression findings concerning the association of MS4A gene cluster with AD pathogenesis, and also speculate the possible roles of MS4A gene cluster in this disease. Based on the contributing effects of MS4A gene cluster in AD pathogenesis, targeting MS4A gene cluster might provide new opportunities for AD treatment.
Collapse
Affiliation(s)
- Jing Ma
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China
| | | | | |
Collapse
|
35
|
Ye L, Yao XD, Wan FN, Qu YY, Liu ZY, Shen XX, Li S, Liu XJ, Yue F, Wang N, Dai B, Ye DW. MS4A8B promotes cell proliferation in prostate cancer. Prostate 2014; 74:911-22. [PMID: 24789009 DOI: 10.1002/pros.22802] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 02/18/2014] [Indexed: 01/12/2023]
Abstract
BACKGROUND Prostate cancer cells must maintain or achieve the further ability of proliferation during the progression. The molecular mechanisms, however, remain poorly understood. We identified a novel oncogene, termed membrane-spanning 4-domains, subfamily A, member 8B (MS4A8B), over-expressed in prostate cancer. METHODS We firstly detected MS4A8B mRNA in 13 types of paired human normal and cancer tissues by real-time polymerase chain reaction (RT-PCR). In 140 clinically localized prostate cancer samples from radical prostatectomy, immunohistochemical staining was performed to study MS4A8B and PCNA protein level as an index of proliferative activity, TUNEL staining as an index of apoptosis. As MS4A8B RNAi and cDNA transfection technologies were used, the effect of MS4A8B on cellular vitality was determined in vitro and in vivo. RESULTS MS4A8B mRNA was over-expressed specifically in prostate cancer. Positive ratios of MS4A8B protein expression were 1.94%, 5.92%, and 62.8% in benign, HPIN and prostate cancer, respectively. Moreover, MS4A8B was positively associated with Gleason score, the proliferation index. In vitro, MS4A8B knockdown resulted in G1 -S cell cycle arrest and descended vitality, MS4A8B over-expression with accelerated S phase entry, elevated vitality in prostate cancer cells. Moreover, it was also found that expression of MS4A8B led to changes of Cyclin D1 , Cyclin E1 and PCNA. LNCaP cells transfected with sh-MS4A8B lentivirus particles grew more slowly when subcutaneously injected into the flanks of nude mice. CONCLUSIONS We conclude that the expression of MS4A8B expression promotes cell proliferation and plays an important role in carcinogenesis and progression of prostate cancer.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Animals
- Apoptosis/physiology
- Cell Cycle Checkpoints/physiology
- Cell Growth Processes/physiology
- Cell Line, Tumor
- Flow Cytometry
- Humans
- Immunohistochemistry
- In Situ Nick-End Labeling
- Kallikreins/metabolism
- Male
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice
- Mice, Nude
- Neoplasms, Hormone-Dependent/genetics
- Neoplasms, Hormone-Dependent/metabolism
- Neoplasms, Hormone-Dependent/pathology
- Prostate-Specific Antigen/metabolism
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/metabolism
- Prostatic Neoplasms/pathology
- RNA, Neoplasm/chemistry
- RNA, Neoplasm/genetics
- RNA, Neoplasm/metabolism
- Real-Time Polymerase Chain Reaction
- Statistics, Nonparametric
Collapse
Affiliation(s)
- Lin Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Segovia M, Louvet C, Charnet P, Savina A, Tilly G, Gautreau L, Carretero-Iglesia L, Beriou G, Cebrian I, Cens T, Hepburn L, Chiffoleau E, Floto RA, Anegon I, Amigorena S, Hill M, Cuturi MC. Autologous dendritic cells prolong allograft survival through Tmem176b-dependent antigen cross-presentation. Am J Transplant 2014; 14:1021-1031. [PMID: 24731243 PMCID: PMC4629416 DOI: 10.1111/ajt.12708] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 12/18/2013] [Accepted: 01/07/2014] [Indexed: 01/25/2023]
Abstract
The administration of autologous (recipient-derived) tolerogenic dendritic cells (ATDCs) is under clinical evaluation. However, the molecular mechanisms by which these cells prolong graft survival in a donor-specific manner is unknown. Here, we tested mouse ATDCs for their therapeutic potential in a skin transplantation model. ATDC injection in combination with anti-CD3 treatment induced the accumulation of CD8(+) CD11c(+) T cells and significantly prolonged allograft survival. TMEM176B is an intracellular protein expressed in ATDCs and initially identified in allograft tolerance. We show that Tmem176b(-/-) ATDCs completely failed to trigger both phenomena but recovered their effect when loaded with donor peptides before injection. These results strongly suggested that ATDCs require TMEM176B to cross-present antigens in a tolerogenic fashion. In agreement with this, Tmem176b(-/-) ATDCs specifically failed to cross-present male antigens or ovalbumin to CD8(+) T cells. Finally, we observed that a Tmem176b-dependent cation current controls phagosomal pH, a critical parameter in cross-presentation. Thus, ATDCs require TMEM176B to cross-present donor antigens to induce donor-specific CD8(+) CD11c(+) T cells with regulatory properties and prolong graft survival.
Collapse
Affiliation(s)
- M. Segovia
- ITUN, INSERM UMR_S 1064, Center for Research in Transplantation and Immunology, Nantes, France
| | - C. Louvet
- ITUN, INSERM UMR_S 1064, Center for Research in Transplantation and Immunology, Nantes, France
| | - P. Charnet
- CRBM, CNRS UMR 5237, Montpellier, France
| | - A. Savina
- Institut Curie, Paris, France
,INSERM U932, Paris, France
| | - G. Tilly
- ITUN, INSERM UMR_S 1064, Center for Research in Transplantation and Immunology, Nantes, France
| | - L. Gautreau
- ITUN, INSERM UMR_S 1064, Center for Research in Transplantation and Immunology, Nantes, France
| | - L. Carretero-Iglesia
- ITUN, INSERM UMR_S 1064, Center for Research in Transplantation and Immunology, Nantes, France
| | - G. Beriou
- ITUN, INSERM UMR_S 1064, Center for Research in Transplantation and Immunology, Nantes, France
| | - I. Cebrian
- Institut Curie, Paris, France
,INSERM U932, Paris, France
| | - T. Cens
- CRBM, CNRS UMR 5237, Montpellier, France
| | - L. Hepburn
- Department of Medicine, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - E. Chiffoleau
- ITUN, INSERM UMR_S 1064, Center for Research in Transplantation and Immunology, Nantes, France
| | - R. A. Floto
- Department of Medicine, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - I. Anegon
- ITUN, INSERM UMR_S 1064, Center for Research in Transplantation and Immunology, Nantes, France
| | - S. Amigorena
- Institut Curie, Paris, France
,INSERM U932, Paris, France
| | - M. Hill
- ITUN, INSERM UMR_S 1064, Center for Research in Transplantation and Immunology, Nantes, France
,Corresponding authors: Marcelo Hill, , and Maria Cristina Cuturi,
| | - M. C. Cuturi
- ITUN, INSERM UMR_S 1064, Center for Research in Transplantation and Immunology, Nantes, France
,Corresponding authors: Marcelo Hill, , and Maria Cristina Cuturi,
| |
Collapse
|
37
|
Otsubo T, Hida Y, Ohga N, Sato H, Kai T, Matsuki Y, Takasu H, Akiyama K, Maishi N, Kawamoto T, Shinohara N, Nonomura K, Hida K. Identification of novel targets for antiangiogenic therapy by comparing the gene expressions of tumor and normal endothelial cells. Cancer Sci 2014; 105:560-7. [PMID: 24602018 PMCID: PMC4317838 DOI: 10.1111/cas.12394] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 03/02/2014] [Accepted: 03/03/2014] [Indexed: 01/25/2023] Open
Abstract
Targeting tumor angiogenesis is an established strategy for cancer therapy. Because angiogenesis is not limited to pathological conditions such as cancer, molecular markers that can distinguish between physiological and pathological angiogenesis are required to develop more effective and safer approaches for cancer treatment. To identify such molecules, we determined the gene expression profiles of murine tumor endothelial cells (mTEC) and murine normal endothelial cells using DNA microarray analysis followed by quantitative reverse transcription–polymerase chain reaction analysis. We identified 131 genes that were differentially upregulated in mTEC. Functional analysis using siRNA-mediated gene silencing revealed five novel tumor endothelial cell markers that were involved in the proliferation or migration of mTEC. The expression of DEF6 and TMEM176B was upregulated in tumor vessels of human renal cell carcinoma specimens, suggesting that they are potential targets for antiangiogenic intervention for renal cell carcinoma. Comparative gene expression analysis revealed molecular differences between tumor endothelial cells and normal endothelial cells and identified novel tumor endothelial cell markers that may be exploited to target tumor angiogenesis for cancer treatment.
Collapse
Affiliation(s)
- Tsuguteru Otsubo
- Drug Discovery II, DSP Cancer Institute, Dainippon Sumitomo Pharma Co., Ltd, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abós B, Castro R, Pignatelli J, Luque A, González L, Tafalla C. Transcriptional heterogeneity of IgM+ cells in rainbow trout (Oncorhynchus mykiss) tissues. PLoS One 2013; 8:e82737. [PMID: 24324826 PMCID: PMC3855791 DOI: 10.1371/journal.pone.0082737] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 10/28/2013] [Indexed: 12/24/2022] Open
Abstract
Two major classes of B lymphocytes have been described to date in rainbow trout: IgM+ and IgT+ cells. IgM+ cells are mainly localized in the spleen, peripheral blood and kidney but are also found in other tissues. However, differences among IgM+ cell populations attending to its location are poorly defined in fish. Thus, the aim of this work was to characterize the expression of different immune molecules such as chemokine receptors, Toll-like receptors (TLRs) and transcription factors on sorted IgM+ lymphocytes from different rainbow trout tissues. IgM+ populations from blood, spleen, kidney, gills, intestine and liver were isolated by cell sorting and the constitutive levels of transcription of these genes evaluated by real-time PCR. To further characterize B cells, we identified an MS4A sequence. In humans, the MS4A family includes several genes with immune functions, such as the B cell marker CD20 or FcRβ. Subsequently, we have also evaluated the mRNA levels of this MS4A gene in the different IgM+ populations. The relevant differences in transcriptional patterns observed for each of these IgM+ populations analyzed, point to the presence of functionally different tissue-specific B cell populations in rainbow trout. The data shown provides a pattern of genes transcribed in IgM+ B cells not previously revealed in teleost fish. Furthermore, the constitutive expression of all the TLR genes analyzed in IgM+ cells suggests an important role for these cells in innate immunity.
Collapse
Affiliation(s)
- Beatriz Abós
- Centro de Investigación en Sanidad Animal (CISA-INIA), Valdeolmos, Madrid, Spain
| | - Rosario Castro
- Centro de Investigación en Sanidad Animal (CISA-INIA), Valdeolmos, Madrid, Spain
| | - Jaime Pignatelli
- Centro de Investigación en Sanidad Animal (CISA-INIA), Valdeolmos, Madrid, Spain
| | - Alfonso Luque
- Centro de Investigación en Sanidad Animal (CISA-INIA), Valdeolmos, Madrid, Spain
| | - Lucia González
- Centro de Investigación en Sanidad Animal (CISA-INIA), Valdeolmos, Madrid, Spain
| | - Carolina Tafalla
- Centro de Investigación en Sanidad Animal (CISA-INIA), Valdeolmos, Madrid, Spain
- * E-mail:
| |
Collapse
|
39
|
Philips ST, Hildenbrand ZL, Oravecz-Wilson KI, Foley SB, Mgbemena VE, Ross TS. Toward a therapeutic reduction of imatinib refractory myeloproliferative neoplasm-initiating cells. Oncogene 2013; 33:5379-90. [PMID: 24240679 PMCID: PMC4025985 DOI: 10.1038/onc.2013.484] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2013] [Revised: 08/27/2013] [Accepted: 09/24/2013] [Indexed: 12/13/2022]
Abstract
Myeloproliferative neoplasms (MPNs) such as chronic myelogenous (CML) and chronic myelomonocytic leukemias (CMML) are frequently induced by tyrosine kinase oncogenes. Although these MPNs are sensitive to tyrosine kinase inhibitors such as imatinib, patients often relapse upon withdrawal of therapy. We used a model of MPN, which is induced by co-expression of the oncoproteins HIP1/PDGFβR (H/P) and AML1/ETO (A/E) from their endogenous loci, to examine the mechanisms of disease development and recurrence following imatinib withdrawal. Although the MPN displayed a full hematologic response to imatinib, 100% of the diseased mice relapsed upon drug withdrawal. MPN persistence was not due to imatinib resistance mutations in the H/P oncogene or massive gene expression changes. Within one week of imatinib treatment, more than 98% of gene expression changes induced by the oncogenes in isolated hematopoietic stem and progenitor cells (LSKs) normalized. Supplementation of imatinib with G-CSF or arsenic trioxide reduced MPN-initiating cell frequencies and the combination of imatinib with arsenic trioxide cured a large fraction of mice with MPNs. In contrast, no mice in the imatinib-treated control cohorts were cured. These data suggest that treatment with a combination of arsenic trioxide and imatinib can eliminate refractory MPN-initiating cells and reduce disease relapse.
Collapse
Affiliation(s)
- S T Philips
- Division of Hematology/Oncology, Department of Internal Medicine, Southwestern Medical Center, University of Texas, Dallas, TX, USA
| | - Z L Hildenbrand
- Division of Hematology/Oncology, Department of Internal Medicine, Southwestern Medical Center, University of Texas, Dallas, TX, USA
| | | | - S B Foley
- Division of Hematology/Oncology, Department of Internal Medicine, Southwestern Medical Center, University of Texas, Dallas, TX, USA
| | - V E Mgbemena
- Division of Hematology/Oncology, Department of Internal Medicine, Southwestern Medical Center, University of Texas, Dallas, TX, USA
| | - T S Ross
- Division of Hematology/Oncology, Department of Internal Medicine, Southwestern Medical Center, University of Texas, Dallas, TX, USA
| |
Collapse
|
40
|
Zuccolo J, Deng L, Unruh TL, Sanyal R, Bau JA, Storek J, Demetrick DJ, Luider JM, Auer-Grzesiak IA, Mansoor A, Deans JP. Expression of MS4A and TMEM176 Genes in Human B Lymphocytes. Front Immunol 2013; 4:195. [PMID: 23874341 PMCID: PMC3711070 DOI: 10.3389/fimmu.2013.00195] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 07/02/2013] [Indexed: 11/13/2022] Open
Abstract
The MS4A gene family in humans includes CD20 and at least 15 other genes. CD20 exists as homo-oligomers in the plasma membrane, however different MS4A proteins expressed in the same cell may hetero-oligomerize. Given the importance of CD20 in B-cell function and as a therapeutic target, we sought to explore the potential for CD20 hetero-oligomerization with other MS4A proteins. We investigated expression in primary human B-cells of the four MS4A genes previously shown to be expressed in human B-cell lines (MS4A4A, MS4A6A, MS4A7, MS4A8B), as well as two genes comprising the closely related TMEM176 gene family, with a view to identifying candidates for future investigation at the protein level. TMEM176A and TMEM176B transcripts were either not detected, or were detected at relatively low levels in a minority of donor B-cell samples. MS4A4A and MS4A8B transcripts were not detected in any normal B-cell sample. MS4A6A and MS4A7 transcripts were detected at low levels in most samples, however the corresponding proteins were not at the plasma membrane when expressed as GFP conjugates in BJAB cells. We also examined expression of these genes in chronic lymphocytic leukemia (CLL), and found that it was similar to normal B-cells with two exceptions. First, whereas MS4A4A expression was undetected in normal B-cells, it was expressed in 1/14 CLL samples. Second, compared to expression levels in normal B-cells, MS4A6A transcripts were elevated in 4/14 CLL samples. In summary, none of the MS4A/TMEM176 genes tested was expressed at high levels in normal or in most CLL B-cells. MS4A6A and MS4A7 were expressed at low levels in most B-cell samples, however the corresponding proteins may not be positioned at the plasma membrane. Altogether, these data suggest that CD20 normally does not form hetero-oligomers with other MS4A proteins and that there are unlikely to be other MS4A proteins in CLL that might provide useful alternate therapeutic targets.
Collapse
Affiliation(s)
- Jonathan Zuccolo
- Department of Biochemistry and Molecular Biology, Snyder Institute for Chronic Diseases, University of Calgary , Calgary, AB , Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Holton P, Ryten M, Nalls M, Trabzuni D, Weale ME, Hernandez D, Crehan H, Gibbs JR, Mayeux R, Haines JL, Farrer LA, Pericak-Vance MA, Schellenberg GD, Ramirez-Restrepo M, Engel A, Myers AJ, Corneveaux JJ, Huentelman MJ, Dillman A, Cookson MR, Reiman EM, Singleton A, Hardy J, Guerreiro R. Initial assessment of the pathogenic mechanisms of the recently identified Alzheimer risk Loci. Ann Hum Genet 2013; 77:85-105. [PMID: 23360175 PMCID: PMC3578142 DOI: 10.1111/ahg.12000] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Accepted: 03/05/2012] [Indexed: 12/27/2022]
Abstract
Recent genome wide association studies have identified CLU, CR1, ABCA7 BIN1, PICALM and MS4A6A/MS4A6E in addition to the long established APOE, as loci for Alzheimer's disease. We have systematically examined each of these loci to assess whether common coding variability contributes to the risk of disease. We have also assessed the regional expression of all the genes in the brain and whether there is evidence of an eQTL explaining the risk. In agreement with other studies we find that coding variability may explain the ABCA7 association, but common coding variability does not explain any of the other loci. We were not able to show that any of the loci had eQTLs within the power of this study. Furthermore the regional expression of each of the loci did not match the pattern of brain regional distribution in Alzheimer pathology. Although these results are mainly negative, they allow us to start defining more realistic alternative approaches to determine the role of all the genetic loci involved in Alzheimer's disease.
Collapse
Affiliation(s)
- Patrick Holton
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Mina Ryten
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Michael Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD
| | - Daniah Trabzuni
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, PO Box 3354, Riyadh 11211, Saudi Arabia
| | - Michael E. Weale
- Department of Medical & Molecular Genetics, King’s College London, Guy’s Hospital, London, UK
| | - Dena Hernandez
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD
| | - Helen Crehan
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - J. Raphael Gibbs
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD
| | - Richard Mayeux
- Gertrude H. Sergievsky Center and Taub Institute on Alzheimer's Disease and the Aging Brain, Department of Neurology, Columbia University, New York, NY
| | - Jonathan L. Haines
- Department of Molecular Physiology and Biophysics and Vanderbilt Center for Human Genetics Research, Vanderbilt University, Nashville, TN
| | - Lindsay A. Farrer
- Departments of Medicine (Biomedical Genetics), Biostatistics, Ophthalmology, Epidemiology, and Neurology, Boston University Schools of Medicine and Public Health, Boston, MA
| | - Margaret A. Pericak-Vance
- The John P. Hussman Institute for Human Genomics and Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miami, FL
| | - Gerard D. Schellenberg
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA
| | | | - Manuel Ramirez-Restrepo
- Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, FL
- Johnnie B. Byrd Sr. Alzheimer's Center and Research Institute, Tampa, FL
| | - Anzhelika Engel
- Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, FL
- Johnnie B. Byrd Sr. Alzheimer's Center and Research Institute, Tampa, FL
| | - Amanda J. Myers
- Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, FL
- Johnnie B. Byrd Sr. Alzheimer's Center and Research Institute, Tampa, FL
| | - Jason J. Corneveaux
- Neurogenomics Division, Translational Genomics Research Institute and Arizona Alzheimer's Consortium, Phoenix, AZ
| | - Matthew J. Huentelman
- Neurogenomics Division, Translational Genomics Research Institute and Arizona Alzheimer's Consortium, Phoenix, AZ
| | - Allissa Dillman
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Mark R. Cookson
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD
| | - Eric M. Reiman
- Neurogenomics Division, Translational Genomics Research Institute and Arizona Alzheimer's Consortium, Phoenix, AZ
- Banner Alzheimer's Institute and Department of Psychiatry, University of Arizona, Phoenix, AZ
- Department of Psychiatry, University of Arizona, Tucson, AZ
| | - Andrew Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD
| | - John Hardy
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
- Reta Lila Weston Laboratories and Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Rita Guerreiro
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| |
Collapse
|
42
|
Tan L, Yu JT, Zhang W, Wu ZC, Zhang Q, Liu QY, Wang W, Wang HF, Ma XY, Cui WZ. Association of GWAS-linked loci with late-onset Alzheimer's disease in a northern Han Chinese population. Alzheimers Dement 2012; 9:546-53. [PMID: 23232270 DOI: 10.1016/j.jalz.2012.08.007] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2012] [Revised: 07/18/2012] [Accepted: 08/29/2012] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Five genomewide association studies (GWAS) in white populations have recently identified and confirmed 9 novel Alzheimer's disease (AD) susceptibility loci (CLU, CR1, PICALM, BIN1, ABCA7, MS4A gene cluster, CD2AP, CD33, and EPHA1). These studies have been conducted almost exclusively in white populations and it is unclear whether these observations generalize to populations with different ethnicities. METHODS We recruited 1224 unrelated northern Han Chinese subjects comprising 612 patients with a clinical diagnosis of late-onset AD (LOAD) according to the criteria of the National Institute of Neurological and Communicative Diseases and Stroke/Alzheimer's Disease and Related Disorders Association and 612 healthy age- and sex-matched control subjects. Because of our previous study investigating CLU, CR1, and PICALM in the Han population, we limited the current analysis to BIN1, ABCA7, MS4A gene cluster, CD2AP, CD33, and EPHA1. RESULTS In a multivariate analysis, associations of MS4A6A (rs610932; odds ratio = 0.632, Bonferroni corrected P = .019) and CD33 (rs3865444; odds ratio = 1.492, Bonferroni corrected P = .017) with LOAD were replicated successfully. When these data were stratified by apolipoprotein E (APOE) ε4 status, both rs610932 and rs610932 were evident only among subjects without the APOE ε4 allele. For BIN1, assuming a dominant model of inheritance, a positive association for rs7561528 in APOE ε4 carriers was observed. This association, however, did not remain significant after Bonferroni correction. As for ABCA7, CD2AP, and EPHA1 single nucleotide polymorphisms from recent GWAS, despite the similar directional effects, no significant differences in genotype and estimated allele frequency distribution between patients and control subjects were observed. CONCLUSIONS This study provides the first independent evidence that MS4A and CD33 loci are associated with the risk of LOAD in northern Han Chinese population. Genotypes at the two loci confer risk predominantly in APOE ε4-negative subjects.
Collapse
Affiliation(s)
- Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, PR China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Karch CM, Jeng AT, Nowotny P, Cady J, Cruchaga C, Goate AM. Expression of novel Alzheimer's disease risk genes in control and Alzheimer's disease brains. PLoS One 2012; 7:e50976. [PMID: 23226438 PMCID: PMC3511432 DOI: 10.1371/journal.pone.0050976] [Citation(s) in RCA: 242] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Accepted: 10/29/2012] [Indexed: 01/18/2023] Open
Abstract
Late onset Alzheimer’s disease (LOAD) etiology is influenced by complex interactions between genetic and environmental risk factors. Large-scale genome wide association studies (GWAS) for LOAD have identified 10 novel risk genes: ABCA7, BIN1, CD2AP, CD33, CLU, CR1, EPHA1, MS4A6A, MS4A6E, and PICALM. We sought to measure the influence of GWAS single nucleotide polymorphisms (SNPs) and gene expression levels on clinical and pathological measures of AD in brain tissue from the parietal lobe of AD cases and age-matched, cognitively normal controls. We found that ABCA7, CD33, and CR1 expression levels were associated with clinical dementia rating (CDR), with higher expression being associated with more advanced cognitive decline. BIN1 expression levels were associated with disease progression, where higher expression was associated with a delayed age at onset. CD33, CLU, and CR1 expression levels were associated with disease status, where elevated expression levels were associated with AD. Additionally, MS4A6A expression levels were associated with Braak tangle and Braak plaque scores, with elevated expression levels being associated with more advanced brain pathology. We failed to detect an association between GWAS SNPs and gene expression levels in our brain series. The minor allele of rs3764650 in ABCA7 is associated with age at onset and disease duration, and the minor allele of rs670139 in MS4A6E was associated with Braak tangle and Braak plaque score. These findings suggest that expression of some GWAS genes, namely ABCA7, BIN1, CD33, CLU, CR1 and the MS4A family, are altered in AD brains.
Collapse
Affiliation(s)
- Celeste M Karch
- Department of Psychiatry and Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, MO, USA
| | | | | | | | | | | |
Collapse
|
44
|
Cuajungco MP, Podevin W, Valluri VK, Bui Q, Nguyen VH, Taylor K. Abnormal accumulation of human transmembrane (TMEM)-176A and 176B proteins is associated with cancer pathology. Acta Histochem 2012; 114:705-12. [PMID: 22244448 DOI: 10.1016/j.acthis.2011.12.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Revised: 12/08/2011] [Accepted: 12/11/2011] [Indexed: 12/31/2022]
Abstract
Transmembrane (TMEM)-176A and 176B proteins belong to the MS4A family of proteins whose function in the immune system remains unclear. TMEM176A transcripts were previously shown to be elevated in liver cancer or kidney tissue with proteinuria, while marked changes in TMEM176B transcripts have been found in tolerated tissue allografts and neoplastic fibroblasts. To study the functional relationship between human TMEM176A and 176B and their putative link to cancer, we used polymerase chain reaction and biochemical assays. Here, we show that TMEM176A and 176B are widely expressed in all human tissues examined. Co-immunoprecipitation of heterologously expressed TMEM176A and 176B revealed direct physical interaction. To determine the relevance of such interaction to cancer pathology, we analyzed biopsied tissue samples from a variety of normal and cancer tissues. Our data reveal that human TMEM176A and 176B protein levels are significantly elevated in lymphoma, but not in normal tissues. The protein levels of TMEM176A are also significantly increased in lung carcinoma. Finally, analysis of the protein expression ratio of TMEM176A over 176B showed significant differences between normal and cancer tissues of the breast, lymph, skin, and liver, which indicates that both TMEM proteins could be potential useful markers for certain human cancers.
Collapse
|
45
|
Further insights into Alzheimer's disease. QUALITY IN AGEING AND OLDER ADULTS 2012. [DOI: 10.1108/14717791211264043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
46
|
Deng YL, Liu LH, Wang Y, Tang HD, Ren RJ, Xu W, Ma JF, Wang LL, Zhuang JP, Wang G, Chen SD. The prevalence of CD33 and MS4A6A variant in Chinese Han population with Alzheimer's disease. Hum Genet 2012; 131:1245-9. [PMID: 22382309 DOI: 10.1007/s00439-012-1154-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Accepted: 02/19/2012] [Indexed: 11/24/2022]
Abstract
CD33 and MS4A6A genes play potential key roles in the pathogenesis of Alzheimer's disease (AD). One recent genome-wide association study has revealed that the rs3865444 polymorphism in the CD33 gene and rs610932 polymorphism in the MS4A6A gene are associated with susceptibility to AD in Caucasians. To evaluate the relationship between the polymorphism of the CD33, MS4A6A gene and AD in the ethnic Chinese Han, we conducted a case-control study (n = 383, age > 54) to determine the prevalence of single-nucleotide polymorphism of two genes in patients with AD in Chinese population of Mainland, and clarified whether these polymorphisms are risk factors for AD. The prevalence of the allele (T) in the rs3865444 polymorphism of the CD33 gene and allele (C) in rs610932 polymorphism of the MS4A6A gene was significantly different in AD patients and control subjects (P < 0.001, respectively), and the results were not influenced by age, gender, or APOE status. Our data revealed the allele (T) of the rs3865444 polymorphism of the CD33 gene and the allele (C) of the rs610932 polymorphism of the MS4A6A gene may contribute to AD risk in the Chinese Han population.
Collapse
Affiliation(s)
- Yu-Lei Deng
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
In the present review, we look back at the recent history of GWAS (genome-wide association studies) in AD (Alzheimer's disease) and integrate the major findings with current knowledge of biological processes and pathways. These topics are essential for the development of animal models, which will be fundamental to our complete understanding of AD.
Collapse
|
48
|
The membrane-spanning 4-domains, subfamily A (MS4A) gene cluster contains a common variant associated with Alzheimer's disease. Genome Med 2011; 3:33. [PMID: 21627779 PMCID: PMC3219074 DOI: 10.1186/gm249] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2011] [Revised: 05/19/2011] [Accepted: 05/31/2011] [Indexed: 12/24/2022] Open
Abstract
Background In order to identify novel loci associated with Alzheimer's disease (AD), we conducted a genome-wide association study (GWAS) in the Spanish population. Methods We genotyped 1,128 individuals using the Affymetrix Nsp I 250K chip. A sample of 327 sporadic AD patients and 801 controls with unknown cognitive status from the Spanish general population were included in our initial study. To increase the power of the study, we combined our results with those of four other public GWAS datasets by applying identical quality control filters and the same imputation methods, which were then analyzed with a global meta-GWAS. A replication sample with 2,200 sporadic AD patients and 2,301 controls was genotyped to confirm our GWAS findings. Results Meta-analysis of our data and independent replication datasets allowed us to confirm a novel genome-wide significant association of AD with the membrane-spanning 4-domains subfamily A (MS4A) gene cluster (rs1562990, P = 4.40E-11, odds ratio = 0.88, 95% confidence interval 0.85 to 0.91, n = 10,181 cases and 14,341 controls). Conclusions Our results underscore the importance of international efforts combining GWAS datasets to isolate genetic loci for complex diseases.
Collapse
|
49
|
Klimcakova E, Roussel B, Kovacova Z, Kovacikova M, Siklova-Vitkova M, Combes M, Hejnova J, Decaunes P, Maoret JJ, Vedral T, Viguerie N, Bourlier V, Bouloumié A, Stich V, Langin D. Macrophage gene expression is related to obesity and the metabolic syndrome in human subcutaneous fat as well as in visceral fat. Diabetologia 2011; 54:876-87. [PMID: 21267541 DOI: 10.1007/s00125-010-2014-3] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2010] [Accepted: 11/25/2010] [Indexed: 12/28/2022]
Abstract
AIMS/HYPOTHESIS Our goal was to identify a set of human adipose tissue macrophage (ATM)-specific markers and investigate whether their gene expression in subcutaneous adipose tissue (SAT) as well as in visceral adipose tissue (VAT) is related to obesity and to the occurrence of the metabolic syndrome. METHODS ATM-specific markers were identified by DNA microarray analysis of adipose tissue cell types isolated from SAT of lean and obese individuals. We then analysed gene expression of these markers by reverse transcription quantitative PCR in paired samples of SAT and VAT from 53 women stratified into four groups (lean, overweight, obese and obese with the metabolic syndrome). Anthropometric measurements, euglycaemic-hyperinsulinaemic clamp, blood analysis and computed tomography scans were performed. RESULTS A panel of 24 genes was selected as ATM-specific markers based on overexpression in ATM compared with other adipose tissue cell types. In SAT and VAT, gene expression of ATM markers was lowest in lean and highest in the metabolic syndrome group. mRNA levels in the two fat depots were negatively correlated with glucose disposal rate and positively associated with indices of adiposity and the metabolic syndrome. CONCLUSIONS/INTERPRETATION In humans, expression of ATM-specific genes increases with the degree of adiposity and correlates with markers of insulin resistance and the metabolic syndrome to a similar degree in SAT and in VAT.
Collapse
Affiliation(s)
- E Klimcakova
- Franco-Czech Laboratory for Clinical Research on Obesity, Department of Sports Medicine, 3rd Faculty of Medicine, Charles University, Prague, Czech Republic.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
López-Muñoz A, Sepulcre MP, Roca FJ, Figueras A, Meseguer J, Mulero V. Evolutionary conserved pro-inflammatory and antigen presentation functions of zebrafish IFNγ revealed by transcriptomic and functional analysis. Mol Immunol 2011; 48:1073-83. [PMID: 21354627 DOI: 10.1016/j.molimm.2011.01.015] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2010] [Revised: 01/26/2011] [Accepted: 01/31/2011] [Indexed: 11/17/2022]
Abstract
In mammals, IFNγ is the only type II IFN member, whereas most bony fish species have two IFNγ genes, namely IFNγ1 and IFNγ2. We report that both zebrafish IFNγ genes were unable to protect zebrafish larvae against viral infection, despite the fact that they moderately induced the expression of antiviral genes, strongly induced pro-inflammatory and antigen processing and presentation genes, and increased neutrophil numbers. Although both zebrafish IFNγs induced a similar set of immune genes, IFNγ1 was more powerful at inducing pro-inflammatory genes than IFNγ2, which correlated with its ability to promote larval death. Strikingly, IFNγ1-induced larval death was prevented by genetic ablation of the myeloid transcription factor SPI1 but not IL-1β or TNFα, suggesting that professional phagocytes are also one of the main targets of IFNγ in fish. In addition, the usefulness of the zebrafish for the identification of IFNγ-target genes is illustrated by the identification of several genes whose expression is also regulated in murine macrophages by IFNγ, namely two membrane-spanning 4-domain family members and the opioid growth factor receptor. Finally, we found for the first time that the thymic specific proteasome subunit PSMB11/β5t is regulated by IFNγ. Collectively, our data throw light on partially redundant functions of fish IFNγ genes, demonstrate that the pro-inflammatory and antigen presentation functions of IFNγ have been conserved during vertebrate evolution, and highlight the fact that zebrafish is an excellent model for studying IFNγ biology.
Collapse
Affiliation(s)
- Azucena López-Muñoz
- Department of Cell Biology and Histology, Faculty of Biology, University of Murcia, 30100 Murcia, Spain
| | | | | | | | | | | |
Collapse
|