1
|
Jung Y, Bang H, Kim YH, Park NE, Park YH, Park C, Lee SR, Lee JW, Song BS, Kim JS, Sim BW, Seol DW, Wee G, Kim S, Kim SU, Kim E. V-Set and Immunoglobulin Domain-Containing 1 (VSIG1), Predominantly Expressed in Testicular Germ Cells, Is Dispensable for Spermatogenesis and Male Fertility in Mice. Animals (Basel) 2021; 11:ani11041037. [PMID: 33916888 PMCID: PMC8067554 DOI: 10.3390/ani11041037] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 03/26/2021] [Indexed: 11/16/2022] Open
Abstract
To elucidate the functional role of V-set and immunoglobulin domain-containing 1 (VSIG1) in spermatogenesis and fertilization, we knocked out (KO) VSIG1 in a mouse embryo using CRISPR/Cas9 (Clustered regularly interspaced short palindromic repeat/CRISPR-associated protein 9) -mediated genome editing. Reverse transcription PCR was performed using cDNA synthesized from VSIG1 KO testis RNA. Although Western blot analysis using a specific antibody to VSIG1 confirmed VSIG1 protein defects in the KO mice, hematoxylin-eosin staining analysis was similar in the KO and wild-type mice. Additionally, computer-assisted sperm analysis and in vitro fertilization experiments were conducted to confirm the activity and fertilization ability of sperm derived from the KO mouse. Mice lacking VSIG1 were viable and had no serious developmental defects. As they got older, the KO mice showed slightly higher weight loss, male mice lacking VSIG1 had functional testes, including normal sperm number and motility, and both male and female mice lacking VSIG1 were fertile. Our results from VSIG1 KO mice suggest that VSIG1 may not play essential roles in spermatogenesis and normal testis development, function, and maintenance. VSIG1 in sperm is dispensable for spermatogenesis and male fertility in mice. As several genes are known to possess slightly different functions depending on the species, the importance and molecular mechanism of VSIG1 in tissues of other species needs further investigation.
Collapse
Affiliation(s)
- Yena Jung
- College of Pharmacy, Catholic University of Daegu, Gyeongsan-si 38430, Korea; (Y.J.); (H.B.); (N.-E.P.); (C.P.)
- Futuristic Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 28116, Korea; (Y.-H.P.); (B.-S.S.); (J.-S.K.); (B.-W.S.); (S.-U.K.)
| | - Hyewon Bang
- College of Pharmacy, Catholic University of Daegu, Gyeongsan-si 38430, Korea; (Y.J.); (H.B.); (N.-E.P.); (C.P.)
| | - Young-Hyun Kim
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 28116, Korea;
| | - Na-Eun Park
- College of Pharmacy, Catholic University of Daegu, Gyeongsan-si 38430, Korea; (Y.J.); (H.B.); (N.-E.P.); (C.P.)
| | - Young-Ho Park
- Futuristic Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 28116, Korea; (Y.-H.P.); (B.-S.S.); (J.-S.K.); (B.-W.S.); (S.-U.K.)
| | - Chaeli Park
- College of Pharmacy, Catholic University of Daegu, Gyeongsan-si 38430, Korea; (Y.J.); (H.B.); (N.-E.P.); (C.P.)
| | - Sang-Rae Lee
- Laboratory Animal Research Center, School of Medicine, Ajou University, Yeongtong-gu, Suwon 16499, Korea;
| | - Jeong-Woong Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Deajeon 34141, Korea;
| | - Bong-Seok Song
- Futuristic Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 28116, Korea; (Y.-H.P.); (B.-S.S.); (J.-S.K.); (B.-W.S.); (S.-U.K.)
| | - Ji-Su Kim
- Futuristic Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 28116, Korea; (Y.-H.P.); (B.-S.S.); (J.-S.K.); (B.-W.S.); (S.-U.K.)
| | - Bo-Woong Sim
- Futuristic Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 28116, Korea; (Y.-H.P.); (B.-S.S.); (J.-S.K.); (B.-W.S.); (S.-U.K.)
| | - Dong-Won Seol
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (D.-W.S.); (G.W.)
| | - Gabbine Wee
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (D.-W.S.); (G.W.)
| | - Sunhyung Kim
- Department of Environmental Horticulture, University of Seoul, Seoul 02504, Korea;
| | - Sun-Uk Kim
- Futuristic Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 28116, Korea; (Y.-H.P.); (B.-S.S.); (J.-S.K.); (B.-W.S.); (S.-U.K.)
| | - Ekyune Kim
- College of Pharmacy, Catholic University of Daegu, Gyeongsan-si 38430, Korea; (Y.J.); (H.B.); (N.-E.P.); (C.P.)
- Correspondence: ; Tel.: +82-53-850-3619; Fax: +82-53-850-3602
| |
Collapse
|
2
|
Ifergan I, Miller SD. Potential for Targeting Myeloid Cells in Controlling CNS Inflammation. Front Immunol 2020; 11:571897. [PMID: 33123148 PMCID: PMC7573146 DOI: 10.3389/fimmu.2020.571897] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 09/03/2020] [Indexed: 12/20/2022] Open
Abstract
Multiple Sclerosis (MS) is characterized by immune cell infiltration to the central nervous system (CNS) as well as loss of myelin. Characterization of the cells in lesions of MS patients revealed an important accumulation of myeloid cells such as macrophages and dendritic cells (DCs). Data from the experimental autoimmune encephalomyelitis (EAE) model of MS supports the importance of peripheral myeloid cells in the disease pathology. However, the majority of MS therapies focus on lymphocytes. As we will discuss in this review, multiple strategies are now in place to target myeloid cells in clinical trials. These strategies have emerged from data in both human and mouse studies. We discuss strategies targeting myeloid cell migration, growth factors and cytokines, biological functions (with a focus on miRNAs), and immunological activities (with a focus on nanoparticles).
Collapse
Affiliation(s)
- Igal Ifergan
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States.,Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Stephen D Miller
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States.,Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
3
|
Lam J, Bellayr IH, Marklein RA, Bauer SR, Puri RK, Sung KE. Functional Profiling of Chondrogenically Induced Multipotent Stromal Cell Aggregates Reveals Transcriptomic and Emergent Morphological Phenotypes Predictive of Differentiation Capacity. Stem Cells Transl Med 2018; 7:664-675. [PMID: 30084545 PMCID: PMC6127231 DOI: 10.1002/sctm.18-0065] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/07/2018] [Accepted: 05/17/2018] [Indexed: 12/20/2022] Open
Abstract
Multipotent stromal cells (MSCs) are an attractive cell source for bone and cartilage tissue repair strategies. However, the functional heterogeneity of MSCs derived from different donors and manufacturing conditions has limited clinical translation, emphasizing the need for improved methods to assess MSC chondrogenic capacity. We used functionally relevant morphological profiling to dynamically monitor emergent morphological phenotypes of chondrogenically induced MSC aggregates to identify morphological features indicative of MSC chondrogenesis. Toward this goal, we characterized the morphology of chondrogenically stimulated MSC aggregates from eight different human cell-lines at multiple passages and demonstrated that MSC aggregates exhibited unique morphological dynamics that were both cell line- and passage-dependent. This variation in 3D morphology was shown to be informative of long-term MSC chondrogenesis based on multiple quantitative functional assays. We found that the specific morphological features of spheroid area, radius, minimum feret diameter, and minor axis length to be strongly correlated with MSC chondrogenic synthetic activity but not gene expression as early as day 4 in 3D culture. Our high-throughput, nondestructive approach could potentially serve as a tool to identify MSC lines with desired chondrogenic capacity toward improving manufacturing strategies for MSC-based cellular products for cartilage tissue repair. Stem Cells Translational Medicine 2018;1-12.
Collapse
Affiliation(s)
- Johnny Lam
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Ian H Bellayr
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Ross A Marklein
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Steven R Bauer
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Raj K Puri
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Kyung E Sung
- Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
4
|
Rosager AM, Sørensen MD, Dahlrot RH, Boldt HB, Hansen S, Lathia JD, Kristensen BW. Expression and prognostic value of JAM-A in gliomas. J Neurooncol 2017; 135:107-117. [PMID: 28677106 PMCID: PMC5658466 DOI: 10.1007/s11060-017-2555-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 06/27/2017] [Indexed: 12/26/2022]
Abstract
Gliomas are among the most lethal cancers, being highly resistant to both chemo- and radiotherapy. The expression of junctional adhesion molecule-A (JAM-A) was recently identified on the surface of stem cell-like brain tumor-initiating cells and suggested to function as a unique glioblastoma niche adhesion factor influencing the tumorigenic potential of brain tumor-initiating cells. We have recently identified high JAM-A expression to be associated with poor outcome in glioblastomas, and our aim was to further investigate the expression of JAM-A in gliomas focusing especially on the prognostic value in WHO grade II and III gliomas. JAM-A protein expression was evaluated by immunohistochemistry and advanced quantitative image analysis with continuous estimates of staining intensity. The JAM-A antibody stained tumor cell membranes and cytoplasm to various extent in different glioma subtypes, and the intensity was higher in glioblastomas than low-grade gliomas. We could not detect an association with overall survival in patients with grade II and III tumors. Double-immunofluorescence stainings in glioblastomas revealed co-expression of JAM-A with CD133, SOX2, nestin, and GFAP in tumor cells as well as some co-expression with the microglial/macrophage marker IBA-1. In conclusion, JAM-A expression was higher in glioblastomas compared to low-grade gliomas and co-localized with recognized stem cell markers suggesting an association of JAM-A with glioma aggressiveness. No significant association between JAM-A expression and overall survival was found in grade II and III gliomas. Further research is needed to determine the function and clinical impact of JAM-A in gliomas.
Collapse
Affiliation(s)
- Ann Mari Rosager
- Department of Pathology, Odense University Hospital, Winsløwparken 15, 3rd floor, 5000, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Winsløwparken 19, 3rd floor, 5000, Odense, Denmark
| | - Mia D Sørensen
- Department of Pathology, Odense University Hospital, Winsløwparken 15, 3rd floor, 5000, Odense, Denmark.
- Department of Clinical Research, University of Southern Denmark, Winsløwparken 19, 3rd floor, 5000, Odense, Denmark.
| | - Rikke H Dahlrot
- Department of Oncology, Odense University Hospital, Sdr. Boulevard 29, 5000, Odense, Denmark
| | - Henning B Boldt
- Department of Pathology, Odense University Hospital, Winsløwparken 15, 3rd floor, 5000, Odense, Denmark
| | - Steinbjørn Hansen
- Department of Clinical Research, University of Southern Denmark, Winsløwparken 19, 3rd floor, 5000, Odense, Denmark
- Department of Oncology, Odense University Hospital, Sdr. Boulevard 29, 5000, Odense, Denmark
| | - Justin D Lathia
- Department of Cellular and Molecular Medicine, Lerner Research Institute, 9500 Euclid Avenue, NC10, Cleveland, OH, 44195, USA
| | - Bjarne W Kristensen
- Department of Pathology, Odense University Hospital, Winsløwparken 15, 3rd floor, 5000, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Winsløwparken 19, 3rd floor, 5000, Odense, Denmark
| |
Collapse
|
5
|
Redmond SA, Mei F, Eshed-Eisenbach Y, Osso LA, Leshkowitz D, Shen YAA, Kay JN, Aurrand-Lions M, Lyons DA, Peles E, Chan JR. Somatodendritic Expression of JAM2 Inhibits Oligodendrocyte Myelination. Neuron 2016; 91:824-836. [PMID: 27499083 DOI: 10.1016/j.neuron.2016.07.021] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 06/08/2016] [Accepted: 07/01/2016] [Indexed: 02/05/2023]
Abstract
Myelination occurs selectively around neuronal axons to increase the efficiency and velocity of action potentials. While oligodendrocytes are capable of myelinating permissive structures in the absence of molecular cues, structurally permissive neuronal somata and dendrites remain unmyelinated. Utilizing a purified spinal cord neuron-oligodendrocyte myelinating co-culture system, we demonstrate that disruption of dynamic neuron-oligodendrocyte signaling by chemical cross-linking results in aberrant myelination of the somatodendritic compartment of neurons. We hypothesize that an inhibitory somatodendritic cue is necessary to prevent non-axonal myelination. Using next-generation sequencing and candidate profiling, we identify neuronal junction adhesion molecule 2 (JAM2) as an inhibitory myelin-guidance molecule. Taken together, our results demonstrate that the somatodendritic compartment directly inhibits myelination and suggest a model in which broadly indiscriminate myelination is tailored by inhibitory signaling to meet local myelination requirements.
Collapse
Affiliation(s)
- Stephanie A Redmond
- Department of Neurology and Program in Neuroscience, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Feng Mei
- Department of Neurology and Program in Neuroscience, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Yael Eshed-Eisenbach
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Lindsay A Osso
- Department of Neurology and Program in Neuroscience, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Dena Leshkowitz
- Bioinformatics Unit, Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Yun-An A Shen
- Department of Neurology and Program in Neuroscience, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jeremy N Kay
- Departments of Neurobiology and Ophthalmology, Duke University School of Medicine, Durham, NC 27703, USA
| | - Michel Aurrand-Lions
- Centre de Recherche en Cancérologie de Marseille, Inserm, CNRS, Aix-Marseille University, UMR1068, 13284 Marseille, France
| | - David A Lyons
- Centre for Neuroregeneration, Centre for Multiple Sclerosis Research, Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Elior Peles
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel.
| | - Jonah R Chan
- Department of Neurology and Program in Neuroscience, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
6
|
Chang CH, Hale SJ, Cox CV, Blair A, Kronsteiner B, Grabowska R, Zhang Y, Cook D, Khoo CP, Schrader JB, Kabuga SB, Martin-Rendon E, Watt SM. Junctional Adhesion Molecule-A Is Highly Expressed on Human Hematopoietic Repopulating Cells and Associates with the Key Hematopoietic Chemokine Receptor CXCR4. Stem Cells 2016; 34:1664-78. [PMID: 26866290 DOI: 10.1002/stem.2340] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 01/11/2016] [Indexed: 12/16/2022]
Abstract
Hematopoietic stem/progenitor cells (HSPCs) reside in specialized bone marrow microenvironmental niches, with vascular elements (endothelial/mesenchymal stromal cells) and CXCR4-CXCL12 interactions playing particularly important roles for HSPC entry, retention, and maintenance. The functional effects of CXCL12 are dependent on its local concentration and rely on complex HSPC-niche interactions. Two Junctional Adhesion Molecule family proteins, Junctional Adhesion Molecule-B (JAM)-B and JAM-C, are reported to mediate HSPC-stromal cell interactions, which in turn regulate CXCL12 production by mesenchymal stromal cells (MSCs). Here, we demonstrate that another JAM family member, JAM-A, is most highly expressed on human hematopoietic stem cells with in vivo repopulating activity (p < .01 for JAM-A(high) compared to JAM-A(Int or Low) cord blood CD34(+) cells). JAM-A blockade, silencing, and overexpression show that JAM-A contributes significantly (p < .05) to the adhesion of human HSPCs to IL-1β activated human bone marrow sinusoidal endothelium. Further studies highlight a novel association of JAM-A with CXCR4, with these molecules moving to the leading edge of the cell upon presentation with CXCL12 (p < .05 compared to no CXCL12). Therefore, we hypothesize that JAM family members differentially regulate CXCR4 function and CXCL12 secretion in the bone marrow niche. Stem Cells 2016;34:1664-1678.
Collapse
Affiliation(s)
- Chao-Hui Chang
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, Stem Cell Research, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.,Radcliffe Department of Medicine, Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| | - Sarah J Hale
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, Stem Cell Research, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.,Radcliffe Department of Medicine, Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| | - Charlotte V Cox
- Bristol Institute for Transfusion Sciences, NHS Blood and Transplant, Bristol, United Kingdom.,Cancer Research School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Allison Blair
- Bristol Institute for Transfusion Sciences, NHS Blood and Transplant, Bristol, United Kingdom.,Cancer Research School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Barbara Kronsteiner
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, Stem Cell Research, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.,Radcliffe Department of Medicine, Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| | - Rita Grabowska
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, Stem Cell Research, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.,Radcliffe Department of Medicine, Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| | - Youyi Zhang
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, Stem Cell Research, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.,Radcliffe Department of Medicine, Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| | - David Cook
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, Stem Cell Research, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.,Radcliffe Department of Medicine, Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| | - Cheen P Khoo
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, Stem Cell Research, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.,Radcliffe Department of Medicine, Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| | - Jack B Schrader
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, Stem Cell Research, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.,Radcliffe Department of Medicine, Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| | - Suranahi Buglass Kabuga
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, Stem Cell Research, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.,Radcliffe Department of Medicine, Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| | - Enca Martin-Rendon
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, Stem Cell Research, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.,Radcliffe Department of Medicine, Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| | - Suzanne M Watt
- Nuffield Division of Clinical Laboratory Medicine, Radcliffe Department of Medicine, Stem Cell Research, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.,Radcliffe Department of Medicine, Stem Cell Research, NHS Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| |
Collapse
|
7
|
Schmidt EP, Kuebler WM, Lee WL, Downey GP. Adhesion Molecules: Master Controllers of the Circulatory System. Compr Physiol 2016; 6:945-73. [PMID: 27065171 DOI: 10.1002/cphy.c150020] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
This manuscript will review our current understanding of cellular adhesion molecules (CAMs) relevant to the circulatory system, their physiological role in control of vascular homeostasis, innate and adaptive immune responses, and their importance in pathophysiological (disease) processes such as acute lung injury, atherosclerosis, and pulmonary hypertension. This is a complex and rapidly changing area of research that is incompletely understood. By design, we will begin with a brief overview of the structure and classification of the major groups of adhesion molecules and their physiological functions including cellular adhesion and signaling. The role of specific CAMs in the process of platelet aggregation and hemostasis and leukocyte adhesion and transendothelial migration will be reviewed as examples of the complex and cooperative interplay between CAMs during physiological and pathophysiological processes. The role of the endothelial glycocalyx and the glycobiology of this complex system related to inflammatory states such as sepsis will be reviewed. We will then focus on the role of adhesion molecules in the pathogenesis of specific disease processes involving the lungs and cardiovascular system. The potential of targeting adhesion molecules in the treatment of immune and inflammatory diseases will be highlighted in the relevant sections throughout the manuscript.
Collapse
Affiliation(s)
- Eric P Schmidt
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Wolfgang M Kuebler
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
- Departments of Surgery and Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Warren L Lee
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
- Division of Respirology and the Interdepartmental Division of Critical Care Medicine, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Gregory P Downey
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Departments of Medicine, Pediatrics, and Biomedical Research, National Jewish Health, Denver, Colorado, USA
- Departments of Medicine, and Immunology and Microbiology, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
8
|
Alvarez JI, Kébir H, Cheslow L, Charabati M, Chabarati M, Larochelle C, Prat A. JAML mediates monocyte and CD8 T cell migration across the brain endothelium. Ann Clin Transl Neurol 2015; 2:1032-7. [PMID: 26734656 PMCID: PMC4693623 DOI: 10.1002/acn3.255] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 08/12/2015] [Accepted: 08/28/2015] [Indexed: 01/03/2023] Open
Abstract
Leukocyte transmigration into the central nervous system promotes multiple sclerosis pathogenesis, yet ambiguity remains regarding the mechanisms controlling the migration of distinct immune cell subsets. Using in vitro, ex vivo and postmortem human materials, we identified a significant upregulation of junctional adhesion molecule‐like expression at the blood–brain barrier, monocytes, and CD8 T cells of multiple sclerosis patients. We also detected junctional adhesion molecule‐like+ trans‐migratory cups when monocytes/CD8 T cells adhered to the blood–brain barrier, however, their migratory capacity was significantly compromised when junctional adhesion molecule‐like was blocked. These findings highlight a novel role for junctional adhesion molecule‐like in leukocyte transmigration and its potential as a promising therapeutic target.
Collapse
Affiliation(s)
- Jorge Iván Alvarez
- Neuroimmunology Research Laboratory Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) Montréa lQuébec Canada; Department of Pathobiology School of Veterinary Medicine University of Pennsylvania Philadelphia Pennsylvania
| | - Hania Kébir
- Neuroimmunology Research Laboratory Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) Montréal Québec Canada
| | - Lara Cheslow
- Department of Pathobiology School of Veterinary Medicine University of Pennsylvania Philadelphia Pennsylvania
| | | | - Marc Chabarati
- Neuroimmunology Research Laboratory Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) Montréal Québec Canada
| | - Catherine Larochelle
- Neuroimmunology Research Laboratory Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) Montréal Québec Canada
| | - Alexandre Prat
- Neuroimmunology Research Laboratory Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) Montréal Québec Canada
| |
Collapse
|
9
|
Abstract
PURPOSE OF REVIEW Neutrophil extravasation from the blood into tissues is initiated by tethering and rolling of neutrophils on endothelial cells, followed by neutrophil integrin activation and shear resistant arrest, crawling, diapedesis and breaching the endothelial basement membrane harbouring pericytes. Endothelial intercellular cell adhesion molecule (ICAM)-1 and ICAM-2, in conjunction with ICAM-1 on pericytes, critically contribute to each step. In addition, epithelial ICAM-1 is involved in neutrophil migration to peri-epithelial sites. The most recent findings on the role of ICAM-1 and ICAM-2 for neutrophil migration into tissues will be reviewed here. RECENT FINDINGS Signalling via endothelial ICAM-1 and ICAM-2 contributes to stiffness of the endothelial cells at sites of chronic inflammation and junctional maturation, respectively. Endothelial ICAM-2 contributes to neutrophil crawling and initiation of paracellular diapedesis, which then proceeds independent of ICAM-2. Substantial transcellular neutrophil diapedesis across the blood-brain barrier is strictly dependent on endothelial ICAM-1 and ICAM-2. Endothelial ICAM-1 or ICAM-2 is involved in neutrophil-mediated plasma leakage. ICAM-1 on pericytes assists the final step of neutrophil extravasation. Epithelial ICAM-1 rather indirectly promotes neutrophil migration to peri-epithelial sites. SUMMARY ICAM-1 and ICAM-2 are involved in each step of neutrophil extravasation, and have redundant but also distinct functions. Analysis of the role of endothelial ICAM-1 requires simultaneous consideration of ICAM-2.
Collapse
|
10
|
Sarelius IH, Glading AJ. Control of vascular permeability by adhesion molecules. Tissue Barriers 2015; 3:e985954. [PMID: 25838987 DOI: 10.4161/21688370.2014.985954] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 11/05/2014] [Indexed: 12/13/2022] Open
Abstract
Vascular permeability is a vital function of the circulatory system that is regulated in large part by the limited flux of solutes, water, and cells through the endothelial cell layer. One major pathway through this barrier is via the inter-endothelial junction, which is driven by the regulation of cadherin-based adhesions. The endothelium also forms attachments with surrounding proteins and cells via 2 classes of adhesion molecules, the integrins and IgCAMs. Integrins and IgCAMs propagate activation of multiple downstream signals that potentially impact cadherin adhesion. Here we discuss the known contributions of integrin and IgCAM signaling to the regulation of cadherin adhesion stability, endothelial barrier function, and vascular permeability. Emphasis is placed on known and prospective crosstalk signaling mechanisms between integrins, the IgCAMs- ICAM-1 and PECAM-1, and inter-endothelial cadherin adhesions, as potential strategic signaling nodes for multipartite regulation of cadherin adhesion.
Collapse
Key Words
- ICAM-1
- ICAM-1, intercellular adhesion molecule 1
- IgCAM, immunoglobulin superfamily cell adhesion molecule
- JAM, junctional adhesion molecule
- LPS, lipopolysaccharide
- PECAM-1
- PECAM-1, platelet endothelial cell adhesion molecule 1
- PKC, protein kinase C
- RDG, arginine-aspartic acid- glutamine
- S1P, sphingosine 1 phosphate
- SHP-2, Src homology region 2 domain-containing phosphatase
- TGF-β, transforming growth factor-β
- TNF-α, tumor necrosis factor α
- VCAM-1, vascular cell adhesion molecule 1
- VE-PTP, Receptor-type tyrosine-protein phosphatase β
- VE-cadherin
- VEGF, vascular endothelial growth factor
- adhesion
- eNOS, endothelial nitric oxide synthase
- endothelial barrier function
- fMLP, f-Met-Leu-Phe
- iNOS, inducible nitric oxide synthase
- integrins
- permeability
- transendothelial migration
Collapse
Affiliation(s)
- Ingrid H Sarelius
- University of Rochester; Department of Pharmacology and Physiology ; Rochester, NY USA
| | - Angela J Glading
- University of Rochester; Department of Pharmacology and Physiology ; Rochester, NY USA
| |
Collapse
|
11
|
Arcangeli ML, Bardin F, Frontera V, Bidaut G, Obrados E, Adams RH, Chabannon C, Aurrand-Lions M. Function of Jam-B/Jam-C interaction in homing and mobilization of human and mouse hematopoietic stem and progenitor cells. Stem Cells 2015; 32:1043-54. [PMID: 24357068 DOI: 10.1002/stem.1624] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 09/21/2013] [Accepted: 10/12/2013] [Indexed: 01/05/2023]
Abstract
The junctional adhesion molecules Jam-b and Jam-c interact together at interendothelial junctions and have been involved in the regulation of immune response, inflammation, and leukocyte migration. More recently, Jam-c has been found to be expressed by hematopoietic stem and progenitor cells (HSPC) in mouse. Conversely, we have reported that Jam-b is present on bone marrow stromal cells and that Jam-b-deficient mice have defects in the regulation of hematopoietic stem cell pool. In this study, we have addressed whether interaction between Jam-b and Jam-c participates to HSPC mobilization or hematopoietic reconstitution after irradiation. We show that a blocking monoclonal antibody directed against Jam-c inhibits hematopoietic reconstitution, progenitor homing to the bone marrow, and induces HSPC mobilization in a Jam-b dependent manner. In the latter setting, antibody treatment over a period of 3 days does not alter hematopoietic differentiation nor induce leukocytosis. Results are translated to human hematopoietic system in which a functional adhesive interaction between JAM-B and JAM-C is found between human HSPC and mesenchymal stem cells. Such an interaction does not occur between HSPC and human endothelial cells or osteoblasts. It is further shown that anti-JAM-C blocking antibody interferes with CD34(+) hematopoietic progenitor homing in mouse bone marrow suggesting that monoclonal antibodies inhibiting JAM-B/JAM-C interaction may represent valuable therapeutic tools to improve stem cell mobilization protocols.
Collapse
Affiliation(s)
- Marie-Laure Arcangeli
- Centre de Recherche en Cancérologie de Marseille, Inserm, UMR1068, Marseille, France; Institut Paoli-Calmettes, Marseille, France; Aix-Marseille Université, Marseille, France; CNRS, UMR7258, Marseille, France
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Sukriti S, Tauseef M, Yazbeck P, Mehta D. Mechanisms regulating endothelial permeability. Pulm Circ 2015; 4:535-51. [PMID: 25610592 DOI: 10.1086/677356] [Citation(s) in RCA: 196] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 03/03/2014] [Indexed: 12/26/2022] Open
Abstract
The endothelial monolayer partitioning underlying tissue from blood components in the vessel wall maintains tissue fluid balance and host defense through dynamically opening intercellular junctions. Edemagenic agonists disrupt endothelial barrier function by signaling the opening of the intercellular junctions leading to the formation of protein-rich edema in the interstitial tissue, a hallmark of tissue inflammation that, if left untreated, causes fatal diseases, such as acute respiratory distress syndrome. In this review, we discuss how intercellular junctions are maintained under normal conditions and after stimulation of endothelium with edemagenic agonists. We have focused on reviewing the new concepts dealing with the alteration of adherens junctions after inflammatory stimulus.
Collapse
Affiliation(s)
- Sukriti Sukriti
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, Illinois, USA
| | - Mohammad Tauseef
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, Illinois, USA
| | - Pascal Yazbeck
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, Illinois, USA
| | - Dolly Mehta
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, Illinois, USA
| |
Collapse
|
13
|
Barrera M, Bahamondes V, Sepúlveda D, Quest A, Castro I, Cortés J, Aguilera S, Urzúa U, Molina C, Pérez P, Ewert P, Alliende C, Hermoso M, González S, Leyton C, González M. Sjögren's syndrome and the epithelial target: A comprehensive review. J Autoimmun 2013; 42:7-18. [DOI: 10.1016/j.jaut.2013.02.001] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 02/11/2013] [Indexed: 12/12/2022]
|