Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
J Gene Med ; 26(2): e3679, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38404047

ABSTRACT

BACKGROUND: Junctional adhesion molecule 2 (JAM2) plays a pivotal role in various biological processes, including proliferation, metastasis and angiogenesis, contributing to tumor progression. While previous studies have highlighted the polarizing functions of JAM2 in different cancer types, its specific role in lung adenocarcinoma (LUAD) remains unclear. METHODS: In this study, we harnessed multiple public databases to analyze the expression and prognostic significance of JAM2 in LUAD. Using the Linkedomics database, Matescape database and R package, we explored the associated genes, the potential biological functions and the impact of JAM2 on the tumor microenvironment. Our findings from public databases were further validated using real-time quantitative PCR, western blot and immunohistochemistry. Additionally, in vitro experiments were conducted to assess the influence of JAM2 on LUAD cell proliferation, invasion, migration, apoptosis and epithelial-mesenchymal transition. Furthermore, we established a xenograft model to investigate the in vivo effects of JAM2 on tumorigenesis. RESULTS: Our results revealed a significant downregulation of JAM2 in LUAD, and patients with low JAM2 expression exhibited unfavorable overall survival outcomes. Functional enrichment analysis indicated that JAM2 may be associated with processes such as cell adhesion, extracellular matrix, cell junctions and regulation of proliferation. Notably, increased JAM2 expression correlated with higher tumor microenvironment scores and reduced immune cell abundance. Furthermore, overexpression of JAM2 induced apoptosis, suppressed tumor proliferation and exhibited potential inhibitory effects on tumor invasion and migration through the modulation of epithelial-mesenchymal transition. Additionally, in vivo experiments confirmed that JAM2 overexpression led to a reduction in tumor growth. CONCLUSION: Overall, our study highlights the clinical significance of low JAM2 expression as a predictor of poor prognosis in LUAD patients. Moreover, JAM2 was found to exert inhibitory effects on various aspects of tumor progression. Consequently, JAM2 emerges as a promising prognostic biomarker and a potential therapeutic target for LUAD patients.


Subject(s)
Adenocarcinoma of Lung , Junctional Adhesion Molecule B , Lung Neoplasms , Humans , Adenocarcinoma of Lung/genetics , Biomarkers , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Junctional Adhesion Molecule B/genetics , Lung Neoplasms/genetics , Prognosis , Tumor Microenvironment/genetics
2.
Poult Sci ; 102(7): 102693, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37120869

ABSTRACT

A functional intestinal barrier is essential for a healthy intestine. This barrier includes an apical tight junctional complex between adjacent intestinal epithelial cells. The tight junctions (TJ) are multiprotein junctional complexes that consist of a number of members of the occludin, claudin, zona occludens, and junctional adhesion molecule families. The mRNA expression of junctional adhesin molecule A (JAMA) and junctional adhesion molecule 2 (JAM2) are 2 TJ mRNAs that are often used to assess intestinal barrier integrity. The objective of this study was to use in situ hybridization to identify cells that express JAMA and JAM2 mRNA in the small intestine of chickens. In the jejunum of a 21 d old broiler, JAMA mRNA was highly expressed in the epithelial cells of the villi and crypt. By contrast, JAM2 mRNA was located in the vascular system in the center of the villi and in the lamina propria. These results demonstrate that JAMA and not JAM2 is the appropriate gene to use when assessing TJ between intestinal epithelial cells.


Subject(s)
Junctional Adhesion Molecule A , Junctional Adhesion Molecule B , Animals , Junctional Adhesion Molecule A/genetics , Junctional Adhesion Molecule A/metabolism , Junctional Adhesion Molecule B/metabolism , Chickens/genetics , Epithelial Cells/metabolism , Tight Junctions , RNA, Messenger/genetics , RNA, Messenger/metabolism , Occludin/genetics
3.
Int J Mol Sci ; 23(15)2022 Aug 03.
Article in English | MEDLINE | ID: mdl-35955781

ABSTRACT

Junctional adhesion molecules (JAMs; comprising JAM-A, -B and -C) act as receptors for viruses, mediate cell permeability, facilitate leukocyte migration during sterile and non-sterile inflammation and are important for the maintenance of epithelial barrier integrity. As such, they are implicated in the development of both communicable and non-communicable chronic diseases. Here, we investigated the expression and regulation of JAM-B in leukocytes under pathogen- and host-derived inflammatory stimuli using immunoassays, qPCR and pharmacological inhibitors of inflammatory signalling pathways. We show that JAM-B is expressed at both the mRNA and protein level in leukocytes. JAM-B protein is localised to the cytoplasm, Golgi apparatus and in the nucleus around ring-shaped structures. We also provide evidence that JAM-B nuclear localisation occurs via the classical importin-α/ß pathway, which is likely mediated through JAM-B protein nuclear localisation signals (NLS) and export signals (NES). In addition, we provide evidence that under both pathogen- and host-derived inflammatory stimuli, JAM-B transcription is regulated via the NF-κB-dependent pathways, whereas at the post-translational level JAM-B is regulated by ubiquitin-proteosome pathways. Anaphase-promoting ubiquitin ligase complex (APC/C) and herpes simplex virus-associated ubiquitin-specific protease (HAUSP/USP) were identified as candidates for JAM-B ubiquitination and de-ubiquitination, respectively. The expression and regulation of JAM-B in leukocytes reported here is a novel observation and contrasts with previous reports. The data reported here suggest that JAM-B expression in leukocytes is under the control of common inflammatory pathways.


Subject(s)
Junctional Adhesion Molecule B , Cell Movement , Humans , Inflammation/metabolism , Junctional Adhesion Molecule B/metabolism , Leukocytes/metabolism , Ubiquitins/metabolism
4.
Dev Biol ; 462(1): 85-100, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32165147

ABSTRACT

Skeletal muscle fusion occurs during development, growth, and regeneration. To investigate how muscle fusion compares among different muscle cell types and developmental stages, we studied muscle cell fusion over time in wild-type, myomaker (mymk), and jam2a mutant zebrafish. Using live imaging, we show that embryonic myoblast elongation and fusion correlate tightly with slow muscle cell migration. In wild-type embryos, only fast muscle fibers are multinucleate, consistent with previous work showing that the cell fusion regulator gene mymk is specifically expressed throughout the embryonic fast muscle domain. However, by 3 weeks post-fertilization, slow muscle fibers also become multinucleate. At this late-larval stage, mymk is not expressed in muscle fibers, but is expressed in small cells near muscle fibers. Although previous work showed that both mymk and jam2a are required for embryonic fast muscle cell fusion, we observe that muscle force and function is almost normal in mymk and jam2a mutant embryos, despite the lack of fast muscle multinucleation. We show that genetic requirements change post-embryonically, with jam2a becoming much less important by late-larval stages and mymk now required for muscle fusion and growth in both fast and slow muscle cell types. Correspondingly, adult mymk mutants perform poorly in sprint and endurance tests compared to wild-type and jam2a mutants. We show that adult mymk mutant muscle contains small mononucleate myofibers with average myonuclear domain size equivalent to that in wild type adults. The mymk mutant fibers have decreased Laminin expression and increased numbers of Pax7-positive cells, suggesting that impaired fiber growth and active regeneration contribute to the muscle phenotype. Our findings identify several aspects of muscle fusion that change with time in slow and fast fibers as zebrafish develop beyond embryonic stages.


Subject(s)
Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Slow-Twitch/metabolism , Muscle, Skeletal/metabolism , Animals , Cell Fusion , Giant Cells/metabolism , Junctional Adhesion Molecule B/genetics , Junctional Adhesion Molecule B/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/physiology , Myoblasts/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Zebrafish/embryology , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
5.
Cell Rep ; 27(10): 2826-2836.e5, 2019 06 04.
Article in English | MEDLINE | ID: mdl-31167130

ABSTRACT

The distinct lineage potential is a key feature of hematopoietic stem cell (HSC) heterogeneity, but a subset of HSCs specialized for a single lymphoid compartment has not been identified. Here we report that HSCs expressing junctional adhesion molecule 2 (Jam2) at a higher level (Jam2high HSCs) have a greater T cell reconstitution capacity. Jam2high HSCs are metabolically dormant but preferentially differentiate toward lymphocytes, especially T cell lineages. Jam2high HSCs uniquely express T cell-related genes, and the interaction with Jam1 facilitates the Notch/Delta signaling pathway. Frequency of Jam2high HSCs changes upon T cell depletion in vivo, potentially suggesting that Jam2 expression may reflect scarcity of T cells and requirement of T cell replenishment. Our findings highlight Jam2 as a potential marker for a subfraction of HSCs with an extensive lymphopoietic capacity, mainly in T lymphopoiesis.


Subject(s)
Hematopoietic Stem Cells/metabolism , Junctional Adhesion Molecule B/metabolism , Lymphopoiesis/genetics , T-Lymphocytes/cytology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Lineage , Female , Hematopoietic Stem Cells/cytology , Junctional Adhesion Molecule B/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Receptor, Notch3/genetics , Receptor, Notch3/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Recombinant Proteins , T-Lymphocytes/metabolism
6.
Mar Biotechnol (NY) ; 21(1): 111-123, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30467785

ABSTRACT

Myoblast fusion is a vital step for skeletal muscle development, growth, and regeneration. Loss of Jamb, Jamc, or Myomaker (Mymk) function impaired myoblast fusion in zebrafish embryos. In addition, mymk mutation hampered fish muscle growth. However, the effect of Jamb and Jamc deficiency on fish muscle growth is not clear. Moreover, whether jamb;jamc and jamb;mymk double mutations have stronger effects on myoblast fusion and muscle growth remains to be investigated. Here, we characterized the muscle development and growth in jamb, jamc, and mymk single and double mutants in zebrafish. We found that although myoblast fusion was compromised in jamb and jamc single or jamb;jamc double mutants, these mutant fish showed no defect in muscle cell fusion during muscle growth. The mutant fish were able to grow into adults that were indistinguishable from the wild-type sibling. In contrast, the jamb;mymk double mutants exhibited a stronger muscle phenotype compared to the jamb and jamc single and double mutants. The jamb;mymk double mutant showed reduced growth and partial lethality, similar to a mymk single mutant. Single fiber analysis of adult skeletal myofibers revealed that jamb, jamc, or jamb;jamc mutants contained mainly multinucleated myofibers, whereas jamb;mymk double mutants contained mostly mononucleated fibers. Significant intramuscular adipocyte infiltration was found in skeletal muscles of the jamb;mymk mutant. Collectively, these studies demonstrate that although Jamb, Jamc, and Mymk are all involved in myoblast fusion during early myogenesis, they have distinct roles in myoblast fusion during muscle growth. While Mymk is essential for myoblast fusion during both muscle development and growth, Jamb and Jamc are dispensable for myoblast fusion during muscle growth.


Subject(s)
Junctional Adhesion Molecule B/genetics , Membrane Proteins/genetics , Muscle Development/genetics , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Myoblasts/metabolism , Receptors, Cell Surface/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Animals, Genetically Modified , Cell Communication , Cell Differentiation , Cell Fusion , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , Junctional Adhesion Molecule B/deficiency , Membrane Proteins/deficiency , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/deficiency , Muscle, Skeletal/cytology , Muscle, Skeletal/growth & development , Mutation , Myoblasts/cytology , Receptors, Cell Surface/deficiency , Zebrafish/growth & development , Zebrafish/metabolism , Zebrafish Proteins/deficiency
7.
Brain Behav Immun ; 73: 3-20, 2018 10.
Article in English | MEDLINE | ID: mdl-29920328

ABSTRACT

In multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE) autoaggressive CD4+ T cells cross the blood-brain barrier (BBB) and cause neuroinflammation. Therapeutic targeting of CD4+ T-cell trafficking into the CNS by blocking α4-integrins has proven beneficial for the treatment of MS but comes with associated risks, probably due to blocking CD8+ T cell mediated CNS immune surveillance. Our recent observations show that CD8+ T cells also rely on α4ß1-integrins to cross the BBB. Besides vascular cell adhesion molecule-1 (VCAM-1), we identified junctional adhesion molecule-B (JAM-B) as a novel vascular α4ß1-integrin ligand involved in CD8+ T-cell migration across the BBB. This prompted us to investigate, if JAM-B also mediates CD4+ T-cell migration across the BBB. We first ensured that encephalitogenic T cells can bind to JAM-B in vitro and next compared EAE pathogenesis in JAM-B-/- C57BL/6J mice and their wild-type littermates. Following immunization with MOGaa35-55 peptide, JAM-B-/- mice developed ameliorated EAE compared to their wild-type littermates. At the same time, we isolated higher numbers of CD45+ infiltrating immune cells from the CNS of JAM-B-/- C57BL/6J mice suffering from EAE. Immunofluorescence staining revealed that the majority of CD45+ inflammatory cells accumulated in the leptomeningeal and perivascular spaces of the CNS behind the BBB but do not gain access to the CNS parenchyma. Trapping of CNS inflammatory cells was not due to increased inflammatory cell proliferation. Neither a loss of BBB integrity or BBB polarity potentially affecting local chemokine gradients nor a lack of focal gelatinase activation required for CNS parenchymal immune cell entry across the glia limitans could be detected in JAM-B-/- mice. Lack of a role for JAM-B in the effector phase of EAE was supported by the observation that we did not detect any role for JAM-B in EAE pathogenesis, when EAE was elicited by in vitro activated MOG aa35-55-specific CD4+ effector T cells. On the other hand, we also failed to demonstrate any role of JAM-B in in vivo priming, proliferation or polarization of MOGaa35-55-specific CD4+ T cells in peripheral immune organs. Finally, our study excludes expression of and thus a role for JAM-B on peripheral and CNS infiltrating myeloid cells. Taken together, although endothelial JAM-B is not required for immune cell trafficking across the BBB in EAE, in its absence accumulation of inflammatory cells mainly in CNS leptomeningeal spaces leads to amelioration of EAE.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/metabolism , Junctional Adhesion Molecule B/metabolism , Junctional Adhesion Molecule B/physiology , Animals , Blood-Brain Barrier/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Movement/physiology , Central Nervous System/metabolism , Central Nervous System/physiology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Endothelium, Vascular/metabolism , Female , Integrin alpha4beta1/metabolism , Junctional Adhesion Molecule B/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Sclerosis/metabolism , Multiple Sclerosis/physiopathology , Myelin-Oligodendrocyte Glycoprotein/pharmacology , Myeloid Cells/metabolism , Myeloid Cells/physiology , Tight Junctions/metabolism
8.
Biochim Biophys Acta Mol Cell Res ; 1865(4): 638-649, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29378216

ABSTRACT

Most cancer deaths result from metastasis, which is the dissemination of cells from a primary tumor to distant organs. Metastasis involves changes to molecules that are essential for tumor cell adhesion to the extracellular matrix and to endothelial cells. Junctional Adhesion Molecule C (JAM-C) localizes at intercellular junctions as homodimers or more affine heterodimers with JAM-B. We previously showed that the homodimerization site (E66) in JAM-C is also involved in JAM-B binding. Here we show that neoexpression of JAM-C in a JAM-C-negative carcinoma cell line induced loss of adhesive property and pro-metastatic capacities. We also identify two critical structural sites (E66 and K68) for JAM-C/JAM-B interaction by directed mutagenesis of JAM-C and studied their implication on tumor cell behavior. JAM-C mutants did not bind to JAM-B or localize correctly to junctions. Moreover, mutated JAM-C proteins increased adhesion and reduced proliferation and migration of lung carcinoma cell lines. Carcinoma cells expressing mutant JAM-C grew slower than with JAM-C WT and were not able to establish metastatic lung nodules in mice. Overall these data demonstrate that the dimerization sites E66-K68 of JAM-C affected cell adhesion, polarization and migration and are essential for tumor cell metastasis.


Subject(s)
Cell Movement , Junctional Adhesion Molecule C/metabolism , Protein Multimerization , Amino Acid Sequence , Animals , Cell Adhesion , Cell Line, Tumor , Cell Polarity , Cell Proliferation , Epithelial Cells/pathology , Junctional Adhesion Molecule B/metabolism , Junctional Adhesion Molecule C/chemistry , Junctional Adhesion Molecule C/genetics , Lung/pathology , Mice, Inbred NOD , Mice, SCID , Mutant Proteins/metabolism , Mutation/genetics , Neoplasm Metastasis , Phenotype , Protein Binding
9.
Sci Rep ; 6: 32428, 2016 08 31.
Article in English | MEDLINE | ID: mdl-27577048

ABSTRACT

Mutations of isocitrate dehydrogenase 1 (IDH1) and IDH2 in acute myeloid leukemia (AML) cells produce the oncometabolite R-2-hydroxyglutarate (R-2HG) to induce epigenetic alteration and block hematopoietic differentiation. However, the effect of R-2HG released by IDH-mutated AML cells on the bone marrow microenvironment is unclear. Here, we report that R-2HG induces IκB kinase-independent activation of NF-κB in bone marrow stromal cells. R-2HG acts via a reactive oxygen species/extracellular signal-regulated kinase (ERK)-dependent pathway to phosphorylate NF-κB on the Thr254 residue. This phosphorylation enhances the interaction of NF-κB and the peptidyl-prolyl cis-trans isomerase PIN1 and increases the protein stability and transcriptional activity of NF-κB. As a consequence, R-2HG enhances NF-κB-dependent expression of cytokines including IL-6, IL-8 and complement 5a to stimulate proliferation of AML cells. In addition, R-2HG also upregulates vascular endothelial adhesion molecule 1 and CXCR4 in stromal cells to enhance the contact between AML and stromal cells and attenuates chemotherapy-induced apoptosis. More importantly, we validated the R-2HG-activated gene signature in the primary bone marrow stromal cells isolated from IDH-mutated AML patients. Collectively, our results suggest that AML cell-derived R-2HG may be helpful for the establishment of a supportive bone marrow stromal niche to promote AML progression via paracrine stimulation.


Subject(s)
Gene Expression Regulation, Leukemic , Glutarates/metabolism , Leukemia, Myeloid, Acute/metabolism , Mesenchymal Stem Cells/drug effects , Myeloid Cells/drug effects , NF-kappa B/genetics , Paracrine Communication/drug effects , Complement C5a/genetics , Complement C5a/metabolism , Glutarates/pharmacology , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Junctional Adhesion Molecule B/genetics , Junctional Adhesion Molecule B/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Mutation , Myeloid Cells/metabolism , Myeloid Cells/pathology , NF-kappa B/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/genetics , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Paracrine Communication/genetics , Phosphorylation , Primary Cell Culture , Protein Stability , Reactive Oxygen Species/metabolism , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Signal Transduction
10.
Neuron ; 91(4): 824-836, 2016 Aug 17.
Article in English | MEDLINE | ID: mdl-27499083

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.


Subject(s)
Junctional Adhesion Molecule B/physiology , Myelin Sheath/metabolism , Oligodendroglia/metabolism , Animals , Coculture Techniques , Junctional Adhesion Molecule B/biosynthesis , Junctional Adhesion Molecule B/genetics , Mice , Mice, Knockout , Myelin Sheath/ultrastructure , Oligodendroglia/ultrastructure , Primary Cell Culture , Rats , Spinal Cord/physiology , Spinal Cord/ultrastructure
11.
Cell Adh Migr ; 10(4): 419-33, 2016 07 03.
Article in English | MEDLINE | ID: mdl-27111582

ABSTRACT

Classical junctional adhesion molecules JAM-A, JAM-B and JAM-C influence vascular permeability, cell polarity as well as leukocyte recruitment and immigration into inflamed tissue. As the vasculature becomes remodelled in chronically injured, fibrotic livers we aimed to determine distribution and role of junctional adhesion molecules during this pathological process. Therefore, livers of naïve or carbon tetrachloride-treated mice were analyzed by immunohistochemistry to localize all 3 classical junctional adhesion molecules. Hepatic stellate cells and endothelial cells were isolated and subjected to immunocytochemistry and flow cytometry to determine localization and functionality of JAM-B and JAM-C. Cells were further used to perform contractility and migration assays and to study endothelial tubulogenesis and pericytic coverage by hepatic stellate cells. We found that in healthy tissue, JAM-A was ubiquitously expressed whereas JAM-B and JAM-C were restricted to the vasculature. During fibrosis, JAM-B and JAM-C levels increased in endothelial cells and JAM-C was de novo generated in myofibroblastic hepatic stellate cells. Soluble JAM-C blocked contractility but increased motility in hepatic stellate cells. Furthermore, soluble JAM-C reduced endothelial tubulogenesis and endothelial cell/stellate cell interaction. Thus, during liver fibrogenesis, JAM-B and JAM-C expression increase on the vascular endothelium. More importantly, JAM-C appears on myofibroblastic hepatic stellate cells linking them as pericytes to JAM-B positive endothelial cells. This JAM-B/JAM-C mediated interaction between endothelial cells and stellate cells stabilizes vessel walls and may control the sinusoidal diameter. Increased hepatic stellate cell contraction mediated by JAM-C/JAM-C interaction may cause intrahepatic vasoconstriction, which is a major complication in liver cirrhosis.


Subject(s)
Cell Communication , Endothelial Cells/pathology , Hepatic Stellate Cells/pathology , Junctional Adhesion Molecule B/metabolism , Junctional Adhesion Molecule C/metabolism , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Animals , Carbon Tetrachloride , Cell Communication/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Movement/drug effects , Cells, Cultured , Collagen/pharmacology , Drug Combinations , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Intercellular Junctions/metabolism , Laminin/pharmacology , Liver/metabolism , Liver/pathology , Mice , Mice, Inbred C57BL , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Myofibroblasts/pathology , Protein Binding/drug effects , Proteoglycans/pharmacology , Recombinant Fusion Proteins/metabolism , Up-Regulation/drug effects
12.
Oncol Rep ; 36(1): 3-9, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27121546

ABSTRACT

The junctional adhesion molecule B (JAM-B) is a multifunctional transmembrane protein, which belongs to the immunoglobulin superfamily (IgSF). JAM-B is localized to cell-cell contacts and enriched at cell junctions in epithelial and endothelial cells, as well as on the surface of erythrocytes, leukocytes, and platelets. Recent research in this field has shown that JAM-B plays an important role in numerous cellular processes, such as tight junction assembly, spermatogenesis, regulation of paracellular permeability, leukocytic transmigration, angiogenesis, tumor metastasis and cell proliferation. This study provides a new research direction for the diagnosis and treatment of relevant diseases. In this review, we briefly focus on what is currently known about the structure, function, and mechanism of JAM-B, with particular emphasis on cancer.


Subject(s)
Cell Adhesion Molecules/metabolism , Intercellular Junctions/metabolism , Junctional Adhesion Molecule B/metabolism , Neoplasms/pathology , Tight Junctions/metabolism , Endothelial Cells/metabolism , Epithelial Cells/metabolism , Humans , Protein Structure, Tertiary , Signal Transduction
13.
Sci Rep ; 6: 21996, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26912257

ABSTRACT

Patients with risks of ischemic injury, e.g. during circulatory arrest in cardiac surgery, or after resuscitation are subjected to therapeutic hypothermia. For aortic surgery, the body is traditionally cooled down to 18 °C and then rewarmed to body temperature. The role of hypothermia and the subsequent rewarming process on leukocyte-endothelial interactions and expression of junctional-adhesion-molecules is not clarified yet. Thus, we investigated in an in-vitro model the influence of temperature modulation during activation and transendothelial migration of leukocytes through human endothelial cells. Additionally, we investigated the expression of JAMs in the rewarming phase. Exposure to low temperatures alone during transmigration scarcely affects leukocyte extravasation, whereas hypothermia during treatment and transendothelial migration improves leukocyte-endothelial interactions. Rewarming causes a significant up-regulation of transmigration with falling temperatures. JAM-A is significantly modulated during rewarming. Our data suggest that transendothelial migration of leukocytes is not only modulated by cell-activation itself. Activation temperatures and the rewarming process are essential. Continued hypothermia significantly inhibits transendothelial migration, whereas the rewarming process enhances transmigration strongly. The expression of JAMs, especially JAM-A, is strongly modulated during the rewarming process. Endothelial protection prior to warm reperfusion and mild hypothermic conditions reducing the difference between hypothermia and rewarming temperatures should be considered.


Subject(s)
Cell Communication , Endothelial Cells/physiology , Hypothermia , Junctional Adhesion Molecule A/metabolism , Junctional Adhesion Molecule B/metabolism , Leukocytes/physiology , Rewarming , Cell Membrane/metabolism , Gene Expression , Humans , Interleukin-10/metabolism , Interleukin-6/metabolism , Junctional Adhesion Molecule A/genetics , Junctional Adhesion Molecule B/genetics , Transendothelial and Transepithelial Migration
14.
Cell Adh Migr ; 10(1-2): 2-17, 2016 03 03.
Article in English | MEDLINE | ID: mdl-26914234

ABSTRACT

The junctional adhesion molecule (JAM)-B, a member of the immunoglobulin superfamily, is involved in stabilization of interendothelial cell-cell contacts, formation of vascular tubes, homeostasis of stem cell niches and promotion of leukocyte adhesion and transmigration. In the human placenta, JAM-B protein is abundant and mRNA transcripts are enriched in first-trimester extravillous trophoblast in comparison to the villous trophoblast. We here aimed to elucidate the yet unexplored spatio-temporal expression of JAM-B in the mouse placenta. We investigated and semi-quantified JAM-B protein expression by immunohistochemistry in early post-implantation si tes and in mid- to late gestation placentae of various murine mating combinations. Surprisingly, the endothelium of the placental labyrinth was devoid of JAM-B expression. JAM-B was mainly present in spongiotrophoblast cells of the junctional zone, as well as in the fetal vessels of the chorionic plate, the umbilical cord and in maternal myometrial smooth muscle. We observed a strain-specific placental increase of JAM-B protein expression from mid- to late gestation in Balb/c-mated C57BL/6 females, which was absent in DBA/2J-mated Balb/c females. Due to the essential role of progesterone during gestation, we further assessed a possible modulation of JAM-B in mid-gestational placentae deficient in the progesterone receptor (Pgr(-/-)) and observed an increased expression of JAM-B in Pgr(-/-) placentae, compared to Pgr(+/+) tissue samples. We propose that JAM-B is an as yet underappreciated trophoblast lineage-specific protein, which is modulated via the progesterone receptor and shows unique strain-specific kinetics. Future work is needed to elucidate its possible contribution to placental processes necessary to ensuring its integrity, ultimately facilitating placental development and fetal growth.


Subject(s)
Junctional Adhesion Molecule B/metabolism , Placenta/metabolism , Animals , Chorion/metabolism , Embryo Implantation , Embryonic Development , Female , Humans , Male , Mice , Pregnancy , Receptors, Progesterone/metabolism , Species Specificity
15.
Dev Dyn ; 244(12): 1574-80, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26370768

ABSTRACT

BACKGROUND: Members of the junctional adhesion molecule (JAM) family function as cell adhesion molecules and cell surface receptors. The zebrafish genome contains six different jam genes, and jam-b and jam-c were shown to be essential for myoblast fusion during skeletal muscle development. However, little is known about jam-b2 expression and function. RESULTS: We isolated the cDNA of zebrafish jam-b2. jam-b2 is expressed specifically in extraocular muscles (EOMs), jaw muscles, and pectoral fins in zebrafish larvae, but not in trunk muscles. The identified jam-b2 expression pattern is supported by the analysis of a zebrafish Gal4-enhancer trap line, in which the coding sequence of the transcriptional activator KalTA4 together with a Gal4-dependent UAS-mCherry expression cassette was inserted into the jam-b2 locus. Intercrosses with an UAS:EGFP strain proves the possibility for targeting transgene expression to EOMs, jaw muscles and fins. Finally, we characterized the concerted contraction pattern of EOMs in larvae performing an optokinetic response. CONCLUSIONS: The expression pattern of jam-b2 suggests that it may contribute different properties to EOMs, jaw muscles, and pectoral fins. The jam-b2:KalTA4-UAS-mCherry transgenic strain serves a dual role as both a reporter for these muscles and as a valuable genetic tool for targeting transgene expression to EOMs.


Subject(s)
Animal Fins/metabolism , Junctional Adhesion Molecule B/metabolism , Muscle, Skeletal/metabolism , Zebrafish Proteins/metabolism , Animal Fins/embryology , Animals , Animals, Genetically Modified , Gene Expression Regulation, Developmental , Junctional Adhesion Molecule B/genetics , Muscle, Skeletal/embryology , Zebrafish/embryology , Zebrafish Proteins/genetics
16.
Biochim Biophys Acta ; 1849(6): 601-11, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25817991

ABSTRACT

Junctional adhesion molecule-B (JAM-B) is found between Sertoli cells at the blood-testis barrier (BTB) as well as between Sertoli and germ cells at the apical ectoplasmic specializations (ES) in the testis. The expression of JAM-B is tightly regulated to modulate the passage of spermatocytes across the BTB as well as the release of mature spermatozoa from the seminiferous epithelium. Transforming growth factor beta (TGF-ß) family is implicated in the regulation of testicular cell junction dynamics during spermatogenesis. This study aims to investigate the effects of TGF-ß3 on the expression of JAM-B as well as the underlying mechanisms on how TGF-ß3 regulates JAM-B expression to facilitate the disassembly of the BTB and apical ES. Our results revealed that TGF-ß3 suppresses JAM-B at post-transcriptional and post-translational levels. Inhibitor, siRNA knockdown and co-immunoprecipitation have shown that TGF-ß3 induces JAM-B protein degradation via ubiquitin-proteasome pathway. Immunofluorescence staining further confirmed that blockage of ubiquitin-proteasome pathway could abrogate TGF-ß3-induced loss of JAM-B at the cell-cell interface. siRNA knockdown and immunofluorescence staining also demonstrated that activation of Smad signaling is required for TGF-ß3-induced JAM-B protein degradation. In addition, TGF-ß3 reduces JAM-B mRNA levels, at least in part, via post-transcriptional regulation. mRNA stability assay has confirmed that TGF-ß3 promotes the degradation of JAM-B transcript and TGF-ß3-mediated mRNA destabilization requires the activation of ERK1/2 and p54 JNK signal cascades. Taken together, TGF-ß3 significantly downregulates JAM-B expression via post-transcriptional and post-translational modulation and results in the disruption of BTB and apical ES.


Subject(s)
Blood-Testis Barrier , Intercellular Junctions/genetics , Junctional Adhesion Molecule B/genetics , Spermatogenesis/genetics , Transforming Growth Factor beta3/biosynthesis , Endocytosis/genetics , Gene Expression Regulation, Developmental , Humans , Intercellular Junctions/metabolism , Junctional Adhesion Molecule B/biosynthesis , Male , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Proteolysis , RNA Stability/genetics , Sertoli Cells/metabolism , Spermatocytes/growth & development , Spermatocytes/metabolism , Testis/growth & development , Testis/metabolism , Transforming Growth Factor beta3/genetics
17.
Nature ; 512(7514): 319-23, 2014 Aug 21.
Article in English | MEDLINE | ID: mdl-25119047

ABSTRACT

Notch signalling plays a key role in the generation of haematopoietic stem cells (HSCs) during vertebrate development and requires intimate contact between signal-emitting and signal-receiving cells, although little is known regarding when, where and how these intercellular events occur. We previously reported that the somitic Notch ligands, Dlc and Dld, are essential for HSC specification. It has remained unclear, however, how these somitic requirements are connected to the later emergence of HSCs from the dorsal aorta. Here we show in zebrafish that Notch signalling establishes HSC fate as their shared vascular precursors migrate across the ventral face of the somite and that junctional adhesion molecules (JAMs) mediate this required Notch signal transduction. HSC precursors express jam1a (also known as f11r) and migrate axially across the ventral somite, where Jam2a and the Notch ligands Dlc and Dld are expressed. Despite no alteration in the expression of Notch ligand or receptor genes, loss of function of jam1a led to loss of Notch signalling and loss of HSCs. Enforced activation of Notch in shared vascular precursors rescued HSCs in jam1a or jam2a deficient embryos. Together, these results indicate that Jam1a-Jam2a interactions facilitate the transduction of requisite Notch signals from the somite to the precursors of HSCs, and that these events occur well before formation of the dorsal aorta.


Subject(s)
Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Junctional Adhesion Molecule A/metabolism , Junctional Adhesion Molecule B/metabolism , Receptors, Cell Surface/metabolism , Receptors, Notch/metabolism , Signal Transduction , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Aorta/cytology , Aorta/growth & development , Aorta/metabolism , Cell Differentiation , Cell Movement , Junctional Adhesion Molecule A/genetics , Junctional Adhesion Molecule B/genetics , Phenotype , Receptors, Cell Surface/genetics , Somites/cytology , Somites/embryology , Somites/metabolism , Zebrafish/embryology , Zebrafish Proteins/genetics
20.
PLoS Biol ; 9(12): e1001216, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22180726

ABSTRACT

Cellular fusion is required in the development of several tissues, including skeletal muscle. In vertebrates, this process is poorly understood and lacks an in vivo-validated cell surface heterophilic receptor pair that is necessary for fusion. Identification of essential cell surface interactions between fusing cells is an important step in elucidating the molecular mechanism of cellular fusion. We show here that the zebrafish orthologues of JAM-B and JAM-C receptors are essential for fusion of myocyte precursors to form syncytial muscle fibres. Both jamb and jamc are dynamically co-expressed in developing muscles and encode receptors that physically interact. Heritable mutations in either gene prevent myocyte fusion in vivo, resulting in an overabundance of mononuclear, but otherwise overtly normal, functional fast-twitch muscle fibres. Transplantation experiments show that the Jamb and Jamc receptors must interact between neighbouring cells (in trans) for fusion to occur. We also show that jamc is ectopically expressed in prdm1a mutant slow muscle precursors, which inappropriately fuse with other myocytes, suggesting that control of myocyte fusion through regulation of jamc expression has important implications for the growth and patterning of muscles. Our discovery of a receptor-ligand pair critical for fusion in vivo has important implications for understanding the molecular mechanisms responsible for myocyte fusion and its regulation in vertebrate myogenesis.


Subject(s)
Cell Fusion , Muscle Development/genetics , Muscle Fibers, Fast-Twitch/physiology , Muscle, Skeletal/embryology , Receptors, Cell Surface/physiology , Zebrafish Proteins/physiology , Animals , Cell Transplantation , Junctional Adhesion Molecule B , Muscle Fibers, Fast-Twitch/cytology , Muscle Fibers, Slow-Twitch/cytology , Muscle Fibers, Slow-Twitch/metabolism , Myoblasts/metabolism , Receptors, Cell Surface/genetics , Zebrafish , Zebrafish Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...