Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 101
Filter
1.
Biomedicines ; 11(12)2023 Nov 30.
Article in English | MEDLINE | ID: mdl-38137406

ABSTRACT

The melanoma cell adhesion molecule, shed from endothelial and cancer cells, is a soluble growth factor that induces tumor angiogenesis and growth. However, the molecular mechanism accounting for its generation in a tumor context is still unclear. To investigate this mechanism, we performed in vitro experiments with endothelial/cancer cells, gene expression analyses on datasets from human colorectal tumor samples, and applied pharmacological methods in vitro/in vivo with mouse and human colorectal cancer cells. We found that soluble MCAM generation is governed by ADAM17 proteolytic activity and NOX1-regulating ADAM17 expression. The treatment of colorectal tumor-bearing mice with pharmacologic NOX1 inhibitors or tumor growth in NOX1-deficient mice reduced the blood concentration of soluble MCAM and abrogated the anti-tumor effects of anti-soluble MCAM antibodies while ADAM17 pharmacologic inhibitors reduced tumor growth and angiogenesis in vivo. Especially, the expression of MCAM, NOX1, and ADAM17 was more prominent in the angiogenic, colorectal cancer-consensus molecular subtype 4 where high MCAM expression correlated with angiogenic and lymphangiogenic markers. Finally, we demonstrated that soluble MCAM also acts as a lymphangiogenic factor in vitro. These results identify a role for NOX1/ADAM17 in soluble MCAM generation, with potential clinical therapeutic relevance to the aggressive, angiogenic CMS4 colorectal cancer subtype.

2.
Front Immunol ; 14: 1085895, 2023.
Article in English | MEDLINE | ID: mdl-37153600

ABSTRACT

Members of the Regulator of G-protein signaling (Rgs) family regulate the extent and timing of G protein signaling by increasing the GTPase activity of Gα protein subunits. The Rgs family member Rgs1 is one of the most up-regulated genes in tissue-resident memory (TRM) T cells when compared to their circulating T cell counterparts. Functionally, Rgs1 preferentially deactivates Gαq, and Gαi protein subunits and can therefore also attenuate chemokine receptor-mediated immune cell trafficking. The impact of Rgs1 expression on tissue-resident T cell generation, their maintenance, and the immunosurveillance of barrier tissues, however, is only incompletely understood. Here we report that Rgs1 expression is readily induced in naïve OT-I T cells in vivo following intestinal infection with Listeria monocytogenes-OVA. In bone marrow chimeras, Rgs1 -/- and Rgs1 +/+ T cells were generally present in comparable frequencies in distinct T cell subsets of the intestinal mucosa, mesenteric lymph nodes, and spleen. After intestinal infection with Listeria monocytogenes-OVA, however, OT-I Rgs1 +/+ T cells outnumbered the co-transferred OT-I Rgs1- /- T cells in the small intestinal mucosa already early after infection. The underrepresentation of the OT-I Rgs1 -/- T cells persisted to become even more pronounced during the memory phase (d30 post-infection). Remarkably, upon intestinal reinfection, mice with intestinal OT-I Rgs1 +/+ TRM cells were able to prevent the systemic dissemination of the pathogen more efficiently than those with OT-I Rgs1 -/- TRM cells. While the underlying mechanisms are not fully elucidated yet, these data thus identify Rgs1 as a critical regulator for the generation and maintenance of tissue-resident CD8+ T cells as a prerequisite for efficient local immunosurveillance in barrier tissues in case of reinfections with potential pathogens.


Subject(s)
CD8-Positive T-Lymphocytes , GTP-Binding Proteins , Listeria monocytogenes , Animals , Mice , GTP-Binding Proteins/metabolism , Protein Subunits/metabolism , T-Lymphocyte Subsets
3.
Cell Mol Life Sci ; 78(23): 7851-7872, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34719737

ABSTRACT

Although the development of hematopoietic stem cells (HSC) has been studied in great detail, their heterogeneity and relationships to different cell lineages remain incompletely understood. Moreover, the role of Vascular Adhesion Protein-1 in bone marrow hematopoiesis has remained unknown. Here we show that VAP-1, an adhesin and a primary amine oxidase producing hydrogen peroxide, is expressed on a subset of human HSC and bone marrow vasculature forming a hematogenic niche. Bulk and single-cell RNAseq analyses reveal that VAP-1+ HSC represent a transcriptionally unique small subset of differentiated and proliferating HSC, while VAP-1- HSC are the most primitive HSC. VAP-1 generated hydrogen peroxide acts via the p53 signaling pathway to regulate HSC proliferation. HSC expansion and differentiation into colony-forming units are enhanced by inhibition of VAP-1. Contribution of VAP-1 to HSC proliferation was confirmed with mice deficient of VAP-1, mice expressing mutated VAP-1 and using an enzyme inhibitor. In conclusion, VAP-1 expression allows the characterization and prospective isolation of a new subset of human HSC. Since VAP-1 serves as a check point-like inhibitor in HSC differentiation, the use of VAP-1 inhibitors enables the expansion of HSC.


Subject(s)
Cell Differentiation , Cell Lineage , Cell Proliferation , Fetal Blood/cytology , Hematopoiesis , Hematopoietic Stem Cells/cytology , Vascular Cell Adhesion Molecule-1/physiology , Animals , Bone Marrow Transplantation , Cell Movement , Female , Hematopoietic Stem Cells/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , RNA-Seq , Stem Cell Niche
4.
Cancers (Basel) ; 13(18)2021 Sep 15.
Article in English | MEDLINE | ID: mdl-34572851

ABSTRACT

The role of the proangiogenic factor olfactomedin-like 3 (OLFML3) in cancer is unclear. To characterize OLFML3 expression in human cancer and its role during tumor development, we undertook tissue expression studies, gene expression analyses of patient tumor samples, in vivo studies in mouse cancer models, and in vitro coculture experiments. OLFML3 was expressed at high levels, mainly in blood vessels, in multiple human cancers. We focused on colorectal cancer (CRC), as elevated expression of OLFML3 mRNA correlated with shorter relapse-free survival, higher tumor grade, and angiogenic microsatellite stable consensus molecular subtype 4 (CMS4). Treatment of multiple in vivo tumor models with OLFML3-blocking antibodies and deletion of the Olfml3 gene from mice decreased lymphangiogenesis, pericyte coverage, and tumor growth. Antibody-mediated blockade of OLFML3 and deletion of host Olfml3 decreased the recruitment of tumor-promoting tumor-associated macrophages and increased infiltration of the tumor microenvironment by NKT cells. Importantly, targeting OLFML3 increased the antitumor efficacy of anti-PD-1 checkpoint inhibitor therapy. Taken together, the results demonstrate that OLFML3 is a promising candidate therapeutic target for CRC.

5.
Molecules ; 26(15)2021 Jul 29.
Article in English | MEDLINE | ID: mdl-34361736

ABSTRACT

We recently developed a molecule (GT-73) that blocked leukocyte transendothelial migration from blood to the peripheral tissues, supposedly by affecting the platelet endothelial cell adhesion molecule (PECAM-1) function. GT-73 was tested in an LPS-induced acute respiratory distress syndrome (ARDS) mouse model. The rationale for this is based on the finding that the mortality of COVID-19 patients is partly caused by ARDS induced by a massive migration of leukocytes to the lungs. In addition, the role of tert-butyl and methyl ester moieties in the biological effect of GT-73 was investigated. A human leukocyte, transendothelial migration assay was applied to validate the blocking effect of GT-73 derivatives. Finally, a mouse model of LPS-induced ARDS was used to evaluate the histological and biochemical effects of GT-73. The obtained results showed that GT-73 has a unique structure that is responsible for its biological activity; two of its chemical moieties (tert-butyl and a methyl ester) are critical for this effect. GT-73 is a prodrug, and its lipophilic tail covalently binds to PECAM-1 via Lys536. GT-73 significantly decreased the number of infiltrating leukocytes in the lungs and reduced the inflammation level. Finally, GT-73 reduced the levels of IL-1ß, IL-6, and MCP-1 in bronchoalveolar lavage fluid (BALF). In summary, we concluded that GT-73, a blocker of white blood cell transendothelial migration, has a favorable profile as a drug candidate for the treatment of ARDS in COVID-19 patients.


Subject(s)
COVID-19 Drug Treatment , Leukocytes/drug effects , Platelet Endothelial Cell Adhesion Molecule-1/antagonists & inhibitors , Pyrimidines/pharmacology , Respiratory Distress Syndrome/drug therapy , Transendothelial and Transepithelial Migration/drug effects , Animals , COVID-19/pathology , Cell Adhesion/drug effects , Cell Adhesion/immunology , Cell Movement/drug effects , Cytokine Release Syndrome/drug therapy , Cytokines/metabolism , Disease Models, Animal , Female , Humans , Leukocytes/immunology , Lipopolysaccharides/adverse effects , Mice , Mice, Inbred BALB C , Platelet Endothelial Cell Adhesion Molecule-1/immunology , Pyrimidines/chemistry , Respiratory Distress Syndrome/chemically induced , SARS-CoV-2
6.
Eur J Immunol ; 51(1): 231-246, 2021 01.
Article in English | MEDLINE | ID: mdl-32970335

ABSTRACT

CD73 is an important ectoenzyme responsible for the production of extracellular adenosine. It is involved in regulating inflammatory responses and cell migration and is overexpressed in various cancers. The functions of CD73 in blood endothelial cells are understood in detail, but its role on afferent lymphatics remains unknown. Moreover, anti-CD73 antibodies are now used in multiple clinical cancer trials, but their effects on different endothelial cell types have not been studied. This study reveals that a previously unknown role of CD73 on afferent lymphatics is to dampen immune responses. Knocking it out or suppressing it by siRNA leads to the upregulation of inflammation-associated genes on lymphatic endothelial cells and a more pro-inflammatory phenotype of interacting dendritic cells in vitro and in vivo. In striking contrast, anti-CD73 antibodies had only negligible effects on the gene expression of lymphatic- and blood-endothelial cells. Our data thus reveal new functions of lymphatic CD73 and indicate a low likelihood of endothelial cell-related adverse effects by CD73 targeting therapeutic antibodies.


Subject(s)
5'-Nucleotidase/immunology , Endothelial Cells/immunology , Inflammation/prevention & control , 5'-Nucleotidase/antagonists & inhibitors , 5'-Nucleotidase/deficiency , 5'-Nucleotidase/genetics , Animals , Antibodies, Blocking/administration & dosage , Cell Differentiation/immunology , Cells, Cultured , Child , Child, Preschool , Dendritic Cells/immunology , Dendritic Cells/pathology , Endothelial Cells/enzymology , Endothelial Cells/pathology , Female , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/deficiency , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , Gene Knockout Techniques , Gene Silencing , Humans , Inflammation/immunology , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Up-Regulation
7.
FASEB J ; 34(11): 15559-15576, 2020 11.
Article in English | MEDLINE | ID: mdl-32997357

ABSTRACT

Pericytes promote vessel stability and their dysfunction causes pathologies due to blood vessel leakage. Previously, we reported that Olfactomedin-like 3 (Olfml3) is a matricellular protein with proangiogenic properties. Here, we explored the role of Olfml3 in a knockout mouse model engineered to suppress this protein. The mutant mice exhibited vascular defects in pericyte coverage, suggesting that pericytes influence blood vessel formation in an Olfml3-dependent manner. Olfml3-deficient mice exhibited abnormalities in the vasculature causing partial lethality of embryos and neonates. Reduced pericyte coverage was observed at embryonic day 12.5 and persisted throughout development, resulting in perinatal death of 35% of Olfml3-deficient mice. Cultured Olfml3-deficient pericytes exhibited aberrant motility and altered pericyte association to endothelial cells. Furthermore, the proliferative response of Olfml3-/- pericytes upon PDGF-B stimulation was significantly diminished. Subsequent experiments revealed that intact PDGF-B signaling, mediated via Olfml3 binding, is required for pericyte proliferation and activation of downstream kinase pathways. Our findings suggest a model wherein pericyte recruitment to endothelial cells requires Olfml3 to provide early instructive cue and retain PDGF-B along newly formed vessels to achieve optimal angiogenesis.


Subject(s)
Cell Movement , Cell Proliferation , Glycoproteins/physiology , Neovascularization, Pathologic/pathology , Pericytes/pathology , Proto-Oncogene Proteins c-sis/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Animals , Embryonic Development , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic/metabolism , Pericytes/metabolism , Pregnancy , Signal Transduction
8.
Stem Cells Dev ; 29(14): 911-918, 2020 07.
Article in English | MEDLINE | ID: mdl-32423362

ABSTRACT

Many persons with diabetes mellitus have limb ischemia, which is a major clinical problem. A subset of human monocytes that expresses TIE-2 may enhance neovascularization. We performed 179 phlebotomies on 142 patients (or donors), including 61 patients/donors without diabetes or ischemia (controls), 39 diabetic nonischemic patients (controls), and 42 diabetic patients with severe limb ischemia requiring amputation. We compared these groups for the presence of TIE-2-positive proangiogenic monocytes. The proportion of proangiogenic monocytes in the venous blood (on hospital admission) was significantly increased in diabetic patients without ischemia (9.22% ± 1.19%), compared to controls (6.53% ± 0.58%) or ischemic diabetic patients (5.44% ± 0.56%) (P < 0.05). In this pilot evaluation, we succeeded in extracting potential proangiogenic TIE-2 monocytes from the blood of diabetic patients without ischemia, but less in patients with ischemia. The implications for therapeutic neoangiogenesis require further studies.


Subject(s)
Diabetic Foot/blood , Ischemia/blood , Monocytes/pathology , Neovascularization, Physiologic , Adult , Aged , Aged, 80 and over , Case-Control Studies , Cell Adhesion Molecules/metabolism , Chronic Disease , Diabetic Foot/pathology , Female , Glycosylation , Humans , Ischemia/pathology , Male , Membrane Glycoproteins/metabolism , Middle Aged , Receptor, TIE-2/metabolism , Receptors, Chemokine/metabolism , Tissue Donors , Transendothelial and Transepithelial Migration
9.
Thromb Res ; 183: 56-62, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31669824

ABSTRACT

BACKGROUND: Hemostasis is a tightly regulated physiological process to rapidly induce hemostatic plugs at sites of vascular injury. Inappropriate activation of this process may lead to thrombosis, i.e. pathological blood clot formation in uninjured vessels or on atherosclerotic lesions. ATP release through Pannexin1 (Panx1) membrane channels contributes to collagen-induced platelet aggregation in vitro. OBJECTIVE: To investigate the effects of genetic and pharmacological inhibition of Panx1 on hemostasis and thrombosis in vivo. RESULTS: Bleeding time after tail clipping was increased by 2.5-fold in Panx1-/- mice compared to wild-type controls, suggesting that Panx1 deficiency impairs primary hemostasis. Wire myography on mesenteric arteries revealed diminished vasoconstriction in response to phenylephrine or U446619 in Panx1-/- mice. Mice with platelet-specific deletion of Panx1 (Panx1PDel) displayed 2-fold longer tail bleeding times than Panx1fl/fl controls. Moreover, venous thromboembolism (VTE) after injection of collagen/epinephrine in the jugular vein was reduced in Panx1-/- and Panx1PDel mice. Panx1PDel mice also showed reduced FeCl3-induced thrombosis in mesenteric arteries. BrilliantBlue-FCF, a Panx1 channel inhibitor, decreased collagen-induced platelet aggregation in vitro, increased tail bleeding time and reduced VTE in wild-type mice. Furthermore, we developed a specific Panx1 blocking antibody targeting a Panx1 extracellular loop, which reduced ATP release from platelets in vitro. Treating wild-type mice with this antibody increased tail bleeding time and decreased VTE compared to control antibody. CONCLUSIONS: Panx1 channel deletion or inhibition diminishes clot formation during hemostasis and thrombosis in vivo. Blocking Panx1 channels may be an attractive strategy for modulating platelet aggregation in thrombotic disease.


Subject(s)
Connexins/antagonists & inhibitors , Hemostasis/physiology , Nerve Tissue Proteins/antagonists & inhibitors , Thrombosis/therapy , Animals , Humans , Male , Mice
10.
Bioorg Chem ; 92: 103250, 2019 11.
Article in English | MEDLINE | ID: mdl-31580982

ABSTRACT

Leukocyte transendothelial migration is one of the most important step in launching an inflammatory immune response and chronic inflammation can lead to devastating diseases. Leukocyte migration inhibitors are considered as promising and potentially effective therapeutic agents to treat inflammatory and auto-immune disorders. In this study, based on previous trioxotetrahydropyrimidin based integrin inhibitors that suboptimally blocked leukocyte adhesion, twelve molecules with a modified scaffold were designed, synthesized, and tested in vitro for their capacity to block the transendothelial migration of immune cells. One of the molecules, namely, methyl 4-((2-(tert-butyl)-6-((2,4,6-trioxotetrahydropyrimidin-5(2H)-ylidene) methyl) phenoxy) methyl) benzoate, (compound 12), completely blocked leukocyte transendothelial migration, without any toxic effects on immune or endothelial cells (IC50 = 2.4 µM). In vivo, compound 12 exhibited significant therapeutic effects in inflammatory bowel disease (IBD)/Crohn's disease, multiple sclerosis, fatty liver disease, and rheumatoid arthritis models. A detailed acute and chronic toxicity profile of the lead compound in vivo did not reveal any toxic effects. Such a type of molecule might therefore provide a unique starting point for designing a novel class of leukocyte transmigration blocking agents with broad therapeutic applications in inflammatory and auto-immune pathologies.


Subject(s)
B-Lymphocytes/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Monocytes/drug effects , Pyrimidines/chemical synthesis , T-Lymphocytes/drug effects , Transcellular Cell Migration/drug effects , Transendothelial and Transepithelial Migration/drug effects , B-Lymphocytes/immunology , Cell Adhesion/drug effects , Cell Adhesion Molecules/immunology , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation , Molecular Structure , Monocytes/immunology , Mucoproteins/immunology , Pyrimidines/chemistry , Pyrimidines/pharmacology , T-Lymphocytes/immunology , Vascular Cell Adhesion Molecule-1/immunology
11.
JCI Insight ; 4(21)2019 11 01.
Article in English | MEDLINE | ID: mdl-31581149

ABSTRACT

The RBC storage lesion is a multiparametric response that occurs during storage at 4°C, but its impact on transfused patients remains unclear. In studies of the RBC storage lesion, the temperature transition from cold storage to normal body temperature that occurs during transfusion has received limited attention. We hypothesized that multiple deleterious events might occur in this period of increasing temperature. We show dramatic alterations in several properties of therapeutic blood units stored at 4°C after warming them to normal body temperature (37°C), as well as febrile temperature (40°C). In particular, the intracellular content and redox state of NADP(H) were directly affected by post-storage incubation at 37°C, as well as by pro-oxidant storage conditions. Modulation of the NADPH-producing pentose phosphate pathway, but not the prevention of hemoglobin autoxidation by conversion of oxyhemoglobin to carboxyhemoglobin, provided protection against storage-induced alterations in RBCs, demonstrating the central role of NADPH in mitigating increased susceptibility of stored RBCs to oxidative stress. We propose that assessing RBC oxidative status after restoration of body temperature constitutes a sensitive method for detecting storage-related alterations that has the potential to improve the quality of stored RBCs for transfusion.


Subject(s)
Erythrocytes/metabolism , Hot Temperature , NADP/metabolism , Oxidative Stress , Adult , Female , Humans , Inosine/administration & dosage , Male , Middle Aged , Pyruvic Acid/administration & dosage
12.
Life Sci Alliance ; 2(4)2019 08.
Article in English | MEDLINE | ID: mdl-31249132

ABSTRACT

NADPH oxidases catalyze the production of reactive oxygen species and are involved in physio/pathological processes. NOX1 is highly expressed in colon cancer and promotes tumor growth. To investigate the efficacy of NOX1 inhibition as an anticancer strategy, tumors were grown in immunocompetent, immunodeficient, or NOX1-deficient mice and treated with the novel NOX1-selective inhibitor GKT771. GKT771 reduced tumor growth, lymph/angiogenesis, recruited proinflammatory macrophages, and natural killer T lymphocytes to the tumor microenvironment. GKT771 treatment was ineffective in immunodeficient mice bearing tumors regardless of their NOX-expressing status. Genetic ablation of host NOX1 also suppressed tumor growth. Combined treatment with the checkpoint inhibitor anti-PD1 antibody had a greater inhibitory effect on colon carcinoma growth than each compound alone. In conclusion, GKT771 suppressed tumor growth by inhibiting angiogenesis and enhancing the recruitment of immune cells. The antitumor activity of GKT771 requires an intact immune system and enhances anti-PD1 antibody activity. Based on these results, we propose blocking of NOX1 by GKT771 as a potential novel therapeutic strategy to treat colorectal cancer, particularly in combination with checkpoint inhibition.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colonic Neoplasms/drug therapy , NADPH Oxidase 1/antagonists & inhibitors , NADPH Oxidases/antagonists & inhibitors , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Tumor Microenvironment/drug effects , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Immunotherapy , Interferon-gamma/metabolism , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Natural Killer T-Cells/drug effects , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Programmed Cell Death 1 Receptor/immunology , Reactive Oxygen Species/metabolism , Reactive Oxygen Species/pharmacology , Tumor Microenvironment/immunology
13.
J Vis Exp ; (141)2018 11 26.
Article in English | MEDLINE | ID: mdl-30531721

ABSTRACT

The recruitment of monocytes from the blood to targeted peripheral tissues is critical to the inflammatory process during tissue injury, tumor development and autoimmune diseases. This is facilitated through a process of capture from free flow onto the luminal surface of activated endothelial cells, followed by their adhesion and transendothelial migration (transmigration) into the underlying affected tissue. However, the mechanisms that support the preferential and context-dependent recruitment of monocyte subpopulations are still not fully understood. Therefore, we have developed a method that allows the recruitment of different monocyte subpopulations to be simultaneously visualized and measured under flow. This method, based on time-lapse confocal imaging, allows for the unambiguous distinction between adherent and transmigrated monocytes. Here, we describe how this method can be used to simultaneously study the recruitment cascade of pro-angiogenic and non-angiogenic monocytes in vitro. Furthermore, this method can be extended to study the different steps of recruitment of up to three monocyte populations.


Subject(s)
Cell Movement/physiology , Monocytes/physiology , Time-Lapse Imaging/methods , Transendothelial and Transepithelial Migration/physiology , Cell Adhesion/physiology , Cells, Cultured , Cohort Studies , Endothelium, Vascular/chemistry , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Humans , Monocytes/chemistry
14.
J Autoimmun ; 91: 83-96, 2018 07.
Article in English | MEDLINE | ID: mdl-29753567

ABSTRACT

Fibrosis remains a serious health concern in patients with chronic liver disease. We recently reported that chemically induced chronic murine liver injury triggers increased expression of junctional adhesion molecules (JAMs) JAM-B and JAM-C by endothelial cells and de novo synthesis of JAM-C by hepatic stellate cells (HSCs). Here, we demonstrate that biopsies of patients suffering from primary biliary cholangitis (PBC), primary sclerosing cholangitis (PSC) or autoimmune hepatitis (AIH) display elevated levels of JAM-C on portal fibroblasts (PFs), HSCs, endothelial cells and cholangiocytes, whereas smooth muscle cells expressed JAM-C constitutively. Therefore, localization and function of JAM-B and JAM-C were investigated in three mouse models of autoimmune-driven liver inflammation. A PBC-like disease was induced by immunization with 2-octynoic acid-BSA conjugate, which resulted in the upregulation of both JAMs in fibrotic portal triads. Analysis of a murine model of PSC revealed a role of JAM-C in PF cell-cell adhesion and contractility. In mice suffering from AIH, endothelial cells increased JAM-B level and HSCs and capsular fibroblasts became JAM-C-positive. Most importantly, AIH-mediated liver fibrosis was reduced in JAM-B-/- mice or when JAM-C was blocked by soluble recombinant JAM-C. Interestingly, loss of JAM-B/JAM-C function had no effect on leukocyte infiltration, suggesting that the well-documented function of JAMs in leukocyte recruitment to inflamed tissue was not effective in the tested chronic models. This might be different in patients and may even be complicated by the fact that human leukocytes express JAM-C. Our findings delineate JAM-C as a mediator of myofibroblast-operated contraction of the liver capsule, intrahepatic vasoconstriction and bile duct stricture. Due to its potential to interact heterophilically with endothelial JAM-B, JAM-C supports also HSC/PF mural cell function. Together, these properties allow JAM-B and JAM-C to actively participate in vascular remodeling associated with liver/biliary fibrosis and suggest them as valuable targets for anti-fibrosis therapies.


Subject(s)
Cell Adhesion Molecules/metabolism , Cholangitis, Sclerosing/metabolism , Endothelial Cells/metabolism , Hepatitis, Autoimmune/metabolism , Immunoglobulins/metabolism , Inflammation/metabolism , Liver Cirrhosis, Biliary/metabolism , Liver/pathology , Myocytes, Smooth Muscle/metabolism , Myofibroblasts/metabolism , Animals , Cell Adhesion , Cell Adhesion Molecules/genetics , Cells, Cultured , Disease Models, Animal , Fatty Acids, Monounsaturated/immunology , Female , Fibrosis , Humans , Immunoglobulins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Vascular Remodeling , Vasoconstriction
15.
J Vis Exp ; (133)2018 03 30.
Article in English | MEDLINE | ID: mdl-29658927

ABSTRACT

B lymphocytes are key players in immune cell circulation and they mainly home to and reside in lymphoid organs. While normal B cells only proliferate when stimulated by T lymphocytes, oncogenic B cells survive and expand autonomously in undefined organ niches. Mantle cell lymphoma (MCL) is one such B cell disorder, where the median survival rate of patients is 4 - 5 years. This calls for the need of effective mechanisms by which the homing and engraftment of these cells are blocked in order to increase the survival and longevity of patients. Therefore, the effort to develop a xenograft mouse model to study the efficacy of MCL therapeutics by blocking the homing mechanism in vivo is of utmost importance. Development of animal recipients for human cell xenotransplantation to test early stage drugs have long been pursued, as relevant preclinical mouse models are crucial to screen new therapeutic agents. This animal model is developed to avoid human graft rejection and to establish a model for human diseases, and it may be an extremely useful tool to study disease progression of different lymphoma types and to perform preclinical testing of candidate drugs for hematologic malignancies, like MCL. We established a xenograft mouse model that will serve as an excellent resource to study and develop novel therapeutic approaches for MCL.


Subject(s)
B-Lymphocytes/metabolism , Heterografts/transplantation , Lymphoma, Mantle-Cell/surgery , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Disease Models, Animal , Humans , Mice , Mice, SCID , Transplantation, Heterologous , Xenograft Model Antitumor Assays
16.
Nat Commun ; 9(1): 355, 2018 01 24.
Article in English | MEDLINE | ID: mdl-29367702

ABSTRACT

Recruitment of circulating monocytes is critical for tumour angiogenesis. However, how human monocyte subpopulations extravasate to tumours is unclear. Here we show mechanisms of extravasation of human CD14dimCD16+ patrolling and CD14+CD16+ intermediate proangiogenic monocytes (HPMo), using human tumour xenograft models and live imaging of transmigration. IFNγ promotes an increase of the chemokine CX3CL1 on vessel lumen, imposing continuous crawling to HPMo and making these monocytes insensitive to chemokines required for their extravasation. Expression of the angiogenic factor VEGF and the inflammatory cytokine TNF by tumour cells enables HPMo extravasation by inducing GATA3-mediated repression of CX3CL1 expression. Recruited HPMo boosts angiogenesis by secreting MMP9 leading to release of matrix-bound VEGF-A, which amplifies the entry of more HPMo into tumours. Uncovering the extravasation cascade of HPMo sets the stage for future tumour therapies.


Subject(s)
Adenocarcinoma/immunology , Breast Neoplasms/immunology , Cell Movement/immunology , Colorectal Neoplasms/immunology , Inflammation/immunology , Monocytes/immunology , Neovascularization, Pathologic/immunology , Animals , Cell Line, Tumor , Chemokine CX3CL1/immunology , GATA3 Transcription Factor/immunology , Humans , Interferon-gamma/immunology , Matrix Metalloproteinase 9/immunology , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Tumor Necrosis Factor-alpha/immunology , Vascular Endothelial Growth Factor A/immunology
17.
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
18.
Eur J Med Chem ; 133: 24-35, 2017 Jun 16.
Article in English | MEDLINE | ID: mdl-28371678

ABSTRACT

Taking into account the structure activity relationship information given by our previous studies, we designed and synthesized a small library of pyrazolylureas and imidazopyrazolecarboxamides fluorinated on urea moiety and differently decorated on pyrazole nucleus. All compounds were preliminary screened by Western blotting technique to evaluate their activity on MAPK and PI3K pathways by monitoring ERK1/2, p38MAPK and Akt phosphorylation, and also screened with a wound healing assay to assess their capacity in inhibiting endothelial cell migration, using human umbilical vein endothelial cells stimulated with VEGF. Pyrazoles and imidazopyrazoles did not show the same activity profile. SAR consideration showed that specific substituents and their position in pyrazole nucleus, as well as the type of substituent on the phenylurea moiety play a pivotal role in determining increase or decrease of kinases phosphorylation. On the other hand the loss of flexibility in imidazopyrazole derivatives is responsible for activity potentiation. Screening of the compound library for inhibition of endothelial cell migration, a function required for angiogenesis, showed significant activity for compound 3. This compound might interfere with cell migration by modulating the activity of different upstream target kinases. Therefore, compound 3 represents a potential inhibitor of angiogenesis. Furthermore, it may be used as a tool to identify unknown mediators of endothelial migration and thereby unveiling new therapeutic targets for controlling pathological angiogenesis in diseases such as cancers.


Subject(s)
Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Urea/analogs & derivatives , Urea/pharmacology , Cell Movement/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Imidazoles/chemistry , Imidazoles/pharmacology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Phosphorylation/drug effects , Vascular Endothelial Growth Factor A/metabolism
19.
Eur J Immunol ; 47(6): 1002-1008, 2017 06.
Article in English | MEDLINE | ID: mdl-28299776

ABSTRACT

Leukocyte recruitment is an important process in combating pathogens. The largest class of circulating leukocytes are neutrophils, which rapidly invade inflamed tissue, followed by inflammatory Ly6C+ monocytes. Ly6Clow monocytes patrol the endothelial wall routinely in the steady state. We recently reported early luminal recruitment of Ly6Clow monocytes, which preceded and orchestrated neutrophil arrival and extravasation in response to TLR7/8-mediated vascular inflammation. Here we dissected the kinetics of recruitment of monocytes and neutrophils and examined the dynamics of Ly6Clow monocytes in response to several other Toll-like receptor (TLR) agonists, using intravital confocal microscopy. We observed two types of kinetics in mesenteric veins. TLR2, TLR5 and TLR9 agonists caused early monocyte and neutrophil influx whereas TLR3 and TLR4 agonists rapidly recruited neutrophils and caused Ly6Clow monocytes to arrive at low levels later on. All TLR agonists, except TLR9, led Ly6Clow monocytes to meticulously patrol the vascular wall. Finally, these monocytes released pro-inflammatory cytokines and chemokines implicated in neutrophil recruitment in response to TLR2, TLR4, and TLR9 stimulation but not to TLR3 and TLR5 agonists. These results refine our understanding of the early events in the leukocyte recruitment cascade, including the patrolling behavior of Ly6Clow monocytes, in TLR-mediated acute vascular inflammation.


Subject(s)
Endothelium/immunology , Inflammation/immunology , Monocytes/immunology , Neutrophil Infiltration , Toll-Like Receptors/immunology , Animals , Antigens, Ly/immunology , Antigens, Ly/metabolism , Chemokines/biosynthesis , Chemokines/immunology , Cytokines/biosynthesis , Cytokines/immunology , Kinetics , Mice , Monocytes/metabolism , Toll-Like Receptor 2/agonists , Toll-Like Receptor 2/immunology , Toll-Like Receptor 9/agonists , Toll-Like Receptor 9/immunology , Toll-Like Receptors/agonists , Toll-Like Receptors/metabolism
20.
J Immunol ; 198(7): 2834-2843, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28193827

ABSTRACT

Two major monocyte subsets, CD14+CD16- (classical) and CD14+/dimCD16+ (nonclassical/intermediate), have been described. Each has different functions ascribed in its interactions with vascular endothelial cells (EC), including migration and promoting inflammation. Although monocyte subpopulations have been studied in isolated systems, their influence on EC and on the course of inflammation has been ignored. In this study, using unstimulated or cytokine-activated EC, we observed significant differences in the recruitment, migration, and reverse migration of human monocyte subsets. Associated with this, and based on their patterns of cytokine secretion, there was a difference in their capacity to activate EC and support the secondary recruitment of flowing neutrophils. High levels of TNF were detected in cocultures with nonclassical/intermediate monocytes, the blockade of which significantly reduced neutrophil recruitment. In contrast, classical monocytes secreted high levels of IL-6, the blockade of which resulted in increased neutrophil recruitment. When cocultures contained both monocyte subsets, or when conditioned supernatant from classical monocytes cocultures (IL-6hi) was added to nonclassical/intermediate monocyte cocultures (TNFhi), the activating effects of TNF were dramatically reduced, implying that when present, the anti-inflammatory activities of IL-6 were dominant over the proinflammatory activities of TNF. These changes in neutrophil recruitment could be explained by regulation of E-selectin on the cocultured EC. This study suggests that recruited human monocyte subsets trigger a regulatory pathway of cytokine-mediated signaling at the EC interface, and we propose that this is a mechanism for limiting the phlogistic activity of newly recruited monocytes.


Subject(s)
Chemotaxis, Leukocyte/immunology , Endothelial Cells/immunology , Inflammation/immunology , Monocytes/immunology , Signal Transduction/immunology , Cell Separation , Flow Cytometry , Humans , Interleukin-6/immunology , Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/immunology
SELECTION OF CITATIONS
SEARCH DETAIL