Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 140
Filter
1.
Sci Rep ; 12(1): 30, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34996933

ABSTRACT

Multiple myeloma (MM) is a cancer of bone marrow (BM) plasma cells, which is increasingly treatable but still incurable. In 90% of MM patients, severe osteolysis results from pathological interactions between MM cells and the bone microenvironment. Delineating specific molecules and pathways for their role in cancer supportive interactions in the BM is vital for developing new therapies. Very Late Antigen 4 (VLA4, integrin α4ß1) is a key player in cell-cell adhesion and signaling between MM and BM cells. We evaluated a VLA4 selective near infrared fluorescent probe, LLP2A-Cy5, for in vitro and in vivo optical imaging of VLA4. Furthermore, two VLA4-null murine 5TGM1 MM cell (KO) clones were generated by CRISPR/Cas9 knockout of the Itga4 (α4) subunit, which induced significant alterations in the transcriptome. In contrast to the VLA4+ 5TGM1 parental cells, C57Bl/KaLwRij immunocompetent syngeneic mice inoculated with the VLA4-null clones showed prolonged survival, reduced medullary disease, and increased extramedullary disease burden. The KO tumor foci showed significantly reduced uptake of LLP2A-Cy5, confirming in vivo specificity of this imaging agent. This work provides new insights into the pathogenic role of VLA4 in MM, and evaluates an optical tool to measure its expression in preclinical models.


Subject(s)
Integrin alpha4beta1/metabolism , Multiple Myeloma/metabolism , Animals , Bone Marrow/metabolism , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Integrin alpha4beta1/chemistry , Integrin alpha4beta1/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Multiple Myeloma/chemistry , Multiple Myeloma/genetics
2.
Int J Mol Sci ; 22(18)2021 Sep 13.
Article in English | MEDLINE | ID: mdl-34576061

ABSTRACT

The role of nintedanib, a multiple tyrosine kinase inhibitor, in the treatment of sepsis-induced acute lung injury (ALI) remains unclear. Lipopolysaccharide (LPS), also known as endotoxin, has been used to induce ALI. The goal of this study was to assess the effect of nintedanib in attenuating the histopathological changes of LPS-induced ALI. Nintedanib was administered via oral gavage to male C57BL/6 mice 24 h and 10 min before intratracheal endotoxin instillation. Lung histopathological characteristics, adhesion molecule expression, and the regulatory signaling pathways of neutrophil chemotaxis were analyzed after 24 h. We found that nintedanib significantly reduced histopathological changes and neutrophil recruitment in LPS-induced ALI. The number of neutrophils in bronchoalveolar lavage fluid (BALF) was reduced in nintedanib-treated relative to untreated mice with ALI. Nintedanib mediated the downregulation of the chemotactic response to LPS by reducing the expression of adhesion molecules and the phosphorylated p38:total p38 mitogen-activated protein kinase (MAPK) ratio in the lungs of mice with ALI. Nintedanib also reduced the expression of lymphocyte antigen 6 complex locus G6D (Ly6G) and very late antigen 4 (VLA-4) in BALF neutrophils and mediated the downregulation of chemokine (C-X-C motif) receptor 2 (CXCR2) and upregulation of G protein-coupled receptor kinase 2 (GRK2) activity in peripheral blood neutrophils in mice with LPS-induced ALI. Nintedanib improved the histopathological changes of LPS-induced ALI by reducing neutrophil chemotaxis. These effects were mediated by the inhibition of adhesion molecules via the activation of GRK2 and the inhibition of p38 MAPK and CXCR2.


Subject(s)
Acute Lung Injury/drug therapy , G-Protein-Coupled Receptor Kinase 2/genetics , Indoles/pharmacology , Receptors, Interleukin-8B/genetics , p38 Mitogen-Activated Protein Kinases/genetics , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Animals , Antigens, Ly/genetics , Bronchoalveolar Lavage Fluid , Disease Models, Animal , Endotoxins/toxicity , Integrin alpha4beta1/genetics , Lipopolysaccharides/toxicity , Neutrophils/metabolism , Neutrophils/pathology , Sepsis/chemically induced , Sepsis/complications , Sepsis/drug therapy , Sepsis/pathology
3.
Sci Rep ; 11(1): 18332, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34526555

ABSTRACT

Roles of interstitial tissue in morphogenesis of testicular structures remain less well understood. To analyze the roles of CD34+ cells in the reconstruction of interstitial tissue containing Leydig cells (LCs), and testicular structures, we used 3D-reaggregate culture of dissociated testicular cells from prepubertal mouse. After a week of culture, adult Leydig cells (ALCs) were preferentially incorporated within CD34+ cell-aggregates, but fetal LCs (FLCs) were not. Immunofluorescence studies showed that integrins α4, α9 and ß1, and VCAM1, one of the ligands for integrins α4ß1 and α9ß1, are expressed mainly in CD34+ cells and ALCs, but not in FLCs. Addition of function-blocking antibodies against each integrin and VCAM1 to the culture disturbed the reconstruction of testicular structures. Antibodies against α4 and ß1 integrins and VCAM1 robustly inhibited cell-to-cell adhesion between testicular cells and between CD34+ cells. Cell-adhesion assays indicated that CD34+ cells adhere to VCAM1 through the interaction with α4ß1 integrin. Live cell imaging showed that CD34+ cells adhered around ALC-aggregates. CD34+ cells on the dish moved toward the aggregates, extending filopodia, and entered into them, which was disturbed by VCAM1 antibody. These results indicate that VCAM1-α4ß1 integrin interaction plays pivotal roles in formation of testicular interstitial tissues in vitro and also in vivo.


Subject(s)
Integrin alpha4beta1/metabolism , Testis/cytology , Testis/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Animals , Antigens, CD34/metabolism , Biomarkers , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cells, Cultured , Gene Expression , Integrin alpha4beta1/genetics , Leydig Cells/metabolism , Male , Mice , Organogenesis/genetics , Protein Binding , Protein Isoforms , Sexual Maturation , Spheroids, Cellular , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/pharmacology
4.
Cell Rep ; 34(6): 108733, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33567286

ABSTRACT

Using intravital imaging, we report that bone marrow (BM) plasma cells (PCs) are motile. BM PCs exhibit a unique migration pattern, characterized by intermittent periods of high motility and longer stretches of confined migration or arrest. BM PCs accumulate into clusters, which have reduced cell motility. APRIL promotes cluster formation and overall PC motility in the BM. Although CXCL12 and its receptor, CXCR4, promote PC motility in the BM, VLA4 activity promotes arrest. However, blocking either pathway promotes PC egress from the BM. Under steady-state conditions, BM PCs recirculate to other bones and spleen. In older mice, overall PC motility and recirculation increase, and this is correlated with increased CXCR4 expression, which depends on PC age or maturation rather than mouse age. Altogether, these results suggest that changes in PC motility and CXCR4 expression are linked with survival of long-lived PCs in the BM.


Subject(s)
Bone Marrow Cells/metabolism , Cell Movement , Cellular Microenvironment , Plasma Cells/metabolism , Animals , Bone Marrow Cells/cytology , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Integrin alpha4beta1/genetics , Integrin alpha4beta1/metabolism , Mice , Mice, Transgenic , Plasma Cells/cytology , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism
5.
Peptides ; 136: 170470, 2021 02.
Article in English | MEDLINE | ID: mdl-33279572

ABSTRACT

Recently, artificial blood vessels modified by integrin α4ß1 ligand, such as REDV, showed endothelialization improvement and antithrombotic properties have been reported. Early endothelialization was affected by the type of circulating cells captured by the peptide in the initial transplantation state, however, it is still not clarified. In this study, we identified in vitro circulating cells bound with the peptides arginine-glutamic acid-aspartic acid-valine (REDV) or histidine-glycine-glycine-valine-arginine-leucine-tyrosine (HGGVRLY). The effect of free C- or N-terminal of HGGVRLY on the type of peptide-binding cells was also studied. The rat circulating cells were isolated from blood and incubated with 5(6)-carboxyfluorescein (5/6-FAM, F) labeled F-REDV (C-terminal free), F-HGGVRLY (C-terminal free), or HGGVRLY-F (N-terminal free). Furthermore, peptide-binding cells were identified by co-staining with various antibodies labeled with PE, PerCP/Cy5.5, or APC. N-terminal free HGGVRLY-F was found to bind to more circulating cells than C-terminal free F-REDV and F-HGGVRLY. The ratio of integrin α4ß1 positive cell bound with F-REDV, F-HGGVRLY, or HGGVRLY-F reached over 90 %, demonstrating that HGGVRLY is also a ligand of integrin α4ß1. Among identified cell types, we found that F-REDV mainly bounds with EPC and BMSC, while F-HGGVRLY with BMSC. HGGVRLY-F bounds with EPC and BMSC, exhibiting a higher EPC binding ratio than F-REDV and F-HGGVRLY.


Subject(s)
Antibodies/chemistry , Integrin alpha4beta1/genetics , Oligopeptides/chemistry , Peptides/genetics , Animals , Antibodies/genetics , Cell-Derived Microparticles/drug effects , Fluoresceins/chemistry , Humans , Integrin alpha4beta1/chemistry , Ligands , Oligopeptides/genetics , Peptides/chemistry , Peptides/pharmacology , Protein Binding/drug effects , Rats
6.
J Pathol ; 252(1): 29-40, 2020 09.
Article in English | MEDLINE | ID: mdl-32501543

ABSTRACT

The interaction of multiple myeloma (MM) cells with the bone marrow (BM) microenvironment promotes MM cell retention, survival, and resistance to different anti-MM agents, including proteasome inhibitors (PIs) such as bortezomib (BTZ). The α4ß1 integrin is a main adhesion receptor mediating MM cell-stroma interactions and MM cell survival, and its expression and function are downregulated by BTZ, leading to inhibition of cell adhesion-mediated drug resistance (CAM-DR) and MM cell apoptosis. Whether decreased α4ß1 expression and activity are maintained or recovered upon development of resistance to BTZ represents an important question, as a potential rescue of α4ß1 function could boost MM cell survival and disease progression. Using BTZ-resistant MM cells, we found that they not only rescue their α4ß1 expression, but its levels were higher than in parental cells. Increased α4ß1 expression in resistant cells correlated with enhanced α4ß1-mediated cell lodging in the BM, and with disease progression. BTZ-resistant MM cells displayed enhanced NF-κB pathway activation relative to parental counterparts, which contributed to upregulated α4 expression and to α4ß1-dependent MM cell adhesion. These data emphasize the upregulation of α4ß1 expression and function as a key event during resistance to BTZ in MM, which might indirectly contribute to stabilize this resistance, as stronger MM cell attachment to BM stroma will regain CAM-DR and MM cell growth and survival. Finally, we found a strong correlation between high ITGB1 (integrin ß1) expression in MM and poor progression-free survival (PFS) and overall survival (OS) during treatment of MM patients with BTZ and IMIDs, and combination of high ITGB1 levels and presence of the high-risk genetic factor amp1q causes low PFS and OS. These results unravel a novel prognostic value for ITGB1 in myeloma. © 2020 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Antineoplastic Agents/administration & dosage , Bortezomib/administration & dosage , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Integrin alpha4beta1/metabolism , Multiple Myeloma/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Integrin alpha4beta1/genetics , Mice , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Tumor Microenvironment
7.
PLoS One ; 15(3): e0228878, 2020.
Article in English | MEDLINE | ID: mdl-32134938

ABSTRACT

We studied a cohort of 367 healthy related donors who volunteered to donate their hematopoietic stem cells for allogeneic transplantation. All donors were homogeneously cared for at a single institution, and received rhG-CSF as a mobilization treatment prior to undergoing apheresis. Peripheral blood CD34+ cell counts were used as the main surrogate marker for rhG-CSF induced mobilization. We searched whether inter-individual variations in known genetic polymorphisms located in genes whose products are functionally important for mobilization, could affect the extent of CD34+ mobilization, either individually or in combination. We found little or no influence of individual SNPs or haplotypes for the SDF1, CXCR4, VCAM and VLA4 genes, whether using CD34+ cell counts as a continuous or a categorical variable. Simple clinical characteristics describing donors such as body mass index, age and possibly sex are more potent predictors of stem cell mobilization. The size of our cohort remains relatively small for genetic analyses, however compares favorably with cohorts analyzed in previously published reports suggesting associations of genetic traits to response to rhG-CSF; notwithstanding this limitation, our data do not support the use of genetic analyses when the choice exists of several potential donors for a given patient.


Subject(s)
Chemokine CXCL12/genetics , Granulocyte Colony-Stimulating Factor/administration & dosage , Integrin alpha4beta1/genetics , Polymorphism, Single Nucleotide , Receptors, CXCR4/genetics , Vascular Cell Adhesion Molecule-1/genetics , Adult , Aged , Female , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Humans , Living Donors , Male , Middle Aged , Transplantation, Homologous , Young Adult
8.
Int J Mol Sci ; 21(6)2020 Mar 23.
Article in English | MEDLINE | ID: mdl-32210016

ABSTRACT

Lineage commitment and differentiation of hematopoietic cells takes place in well-defined microenvironmental surroundings. Communication with other cell types is a vital prerequisite for the normal functions of the immune system, while disturbances in this communication support the development and progression of neoplastic disease. Integrins such as the integrin very late antigen-4 (VLA-4; CD49d/CD29) control the localization of healthy as well as malignant B cells within the tissue, and thus determine the patterns of organ infiltration. Malignant B cells retain some key characteristics of their normal counterparts, with B cell receptor (BCR) signaling and integrin-mediated adhesion being essential mediators of tumor cell homing, survival and proliferation. It is thus not surprising that targeting the BCR pathway using small molecule inhibitors has proved highly effective in the treatment of B cell malignancies. Attenuation of BCR-dependent lymphoma-microenvironment interactions was, in this regard, described as a main mechanism critically contributing to the efficacy of these agents. Here, we review the contribution of VLA-4 to normal B cell differentiation on the one hand, and to the pathophysiology of B cell malignancies on the other hand. We describe its impact as a prognostic marker, its interplay with BCR signaling and its predictive role for novel BCR-targeting therapies, in chronic lymphocytic leukemia and beyond.


Subject(s)
Integrin alpha4beta1/genetics , Integrin alpha4beta1/metabolism , Leukemia, B-Cell/etiology , Leukemia, B-Cell/metabolism , Lymphoma, B-Cell/etiology , Lymphoma, B-Cell/metabolism , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Bone Marrow/metabolism , Cell Differentiation/genetics , Cellular Microenvironment/genetics , Disease Management , Disease Susceptibility , Gene Expression Regulation, Neoplastic , Hematopoiesis/genetics , Humans , Integrins/genetics , Integrins/metabolism , Leukemia, B-Cell/drug therapy , Leukemia, B-Cell/pathology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/pathology , Molecular Targeted Therapy , Signal Transduction
10.
Cancer Lett ; 473: 62-73, 2020 03 31.
Article in English | MEDLINE | ID: mdl-31904479

ABSTRACT

Several studies have indicated that cancer-associated fibroblasts (CAFs) could promote cancer progression in many malignancies. However, the mechanism by which CAFs promote the growth and metastasis of lung cancer remains poorly defined. In the present study, CAFs and normal fibroblasts (NFs) were isolated from human lung cancer and adjacent tissue. The data showed that the conditional medium (CM) of CAFs could increase the proliferation, migration and invasion of lung cancer cells. Vascular cell adhesion molecule-1 (VCAM-1) showed a higher expression in CAF-CM than NF-CM, and blocking VCAM-1 in CAF-CM attenuated the proliferation and invasion of cancer cells. Further, the results showed that VCAM-1 secreted from CAFs activated AKT and MAPK signaling via receptor α4ß1 integrin (very-late antigen (VLA)-4) in lung cancer cells. Moreover, CAFs promoted VCAM-1 expression and tumor growth in vivo. Additionally, bioinformatics analysis indicated a positive correlation on the CAF marker protein alpha-smooth muscle actin (α-SMA) and VCAM-1 expression, which was associated with a poor prognosis in lung cancer patients. These findings demonstrate that the VCAM-1 secreted from CAFs enhances growth and invasion by activating the AKT and MAPK signaling of lung cancer cells.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Lung Neoplasms/pathology , MAP Kinase Signaling System , Proto-Oncogene Proteins c-akt/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , A549 Cells , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Humans , Integrin alpha4beta1/genetics , Integrin alpha4beta1/metabolism , Kaplan-Meier Estimate , Lung/cytology , Lung/pathology , Lung/surgery , Lung Neoplasms/mortality , Lung Neoplasms/surgery , Male , Middle Aged , Neoplasm Invasiveness/pathology , Pneumonectomy , Primary Cell Culture , Prognosis , RNA, Small Interfering/metabolism , Survival Rate , Tumor Microenvironment , Xenograft Model Antitumor Assays
11.
Exp Hematol ; 81: 16-31.e4, 2020 01.
Article in English | MEDLINE | ID: mdl-31887343

ABSTRACT

We previously studied the role of ß1 integrin and some of its different α partners relevant to erythropoiesis. Although clear and consistent answers regarding the role of α4ß1 (VLA-4) were evident, the role of its companion integrin α5ß1 (VLA-5) was clouded by inconsistent outcomes in all prior publications. Furthermore, the functional consequences of integrin deficiencies only in microenvironmental (ME) cells supporting erythroid cell expansion and maturation post stress have never been explored. In the study described here, we created several additional mouse models in the aim of addressing unanswered questions regarding functional consequences of single or combined integrin deficiencies in erythroid cells or only in ME supporting cells. Our novel and expansive data solidified the intrinsic requirement of both α4 and α5 integrins in erythroid cells for their proliferative expansion and maturation in response to stress; α5 integrin alone, deleted either early in all hematopoietic cells or only in erythroid cell, has only a redundant role in proliferative expansion and is dispensable for erythroid maturation. By contrast, α4 integrin, on its own, exerts a dominant effect on timely and optimal erythroid maturation. Deficiency of both α4 and α5 integrins in ME cells, including macrophages, does not negatively influence stress response by normal erythroid cells, in great contrast to the effect of ME cells deficient in all ß1 integrins. Collectively the present data offer deeper insight into the coordination of different ß1 integrin functional activities in erythroid cells or in ME cells for optimal erythroid stress response.


Subject(s)
Erythroid Cells/metabolism , Erythropoiesis , Integrin alpha5/metabolism , Stem Cell Niche , Stress, Physiological , Animals , Erythroid Cells/cytology , Integrin alpha4beta1/genetics , Integrin alpha4beta1/metabolism , Integrin alpha5/genetics , Integrin alpha5beta1/genetics , Integrin alpha5beta1/metabolism , Integrin beta1/genetics , Integrin beta1/metabolism , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Knockout
12.
J Cell Sci ; 132(20)2019 10 28.
Article in English | MEDLINE | ID: mdl-31562188

ABSTRACT

When targeted by the tumor-promoting enzyme heparanase, cleaved and shed syndecan-1 (Sdc1) then couples VEGFR2 (also known as KDR) to VLA-4, activating VEGFR2 and the directed migration of myeloma cells. But how VEGFR2 activates VLA-4-mediated motility has remained unknown. We now report that VEGFR2 causes PKA-mediated phosphorylation of VLA-4 on S988, an event known to stimulate tumor metastasis while suppressing cytotoxic immune cells. A key partner in this mechanism is the chemokine receptor CXCR4, a well-known mediator of cell motility in response to gradients of the chemokine SDF-1 (also known as CXCL12). The entire machinery necessary to phosphorylate VLA-4, consisting of CXCR4, AC7 (also known as ADCY7) and PKA, is constitutively associated with VEGFR2 and is localized to the integrin by Sdc1. VEGFR2 carries out the novel phosphorylation of Y135 within the DRY microswitch of CXCR4, sequentially activating Gαißγ, AC7 and PKA, which phosphorylates S988 on the integrin. This mechanism is blocked by a syndecan-mimetic peptide (SSTNVEGFR2), which, by preventing VEGFR2 linkage to VLA-4, arrests tumor cell migration that depends on VLA-4 phosphorylation and stimulates the LFA-1-mediated migration of cytotoxic leukocytes.


Subject(s)
Cell Movement/immunology , Integrin alpha4beta1/immunology , Neoplasm Proteins/immunology , Neoplasms/immunology , Receptors, CXCR4/immunology , Syndecan-1/immunology , Vascular Endothelial Growth Factor Receptor-2/immunology , Cell Line, Tumor , Cell Movement/genetics , Humans , Immunologic Surveillance , Integrin alpha4beta1/genetics , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , Phosphorylation/genetics , Phosphorylation/immunology , Receptors, CXCR4/genetics , Syndecan-1/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics
13.
World J Emerg Surg ; 14: 24, 2019.
Article in English | MEDLINE | ID: mdl-31164913

ABSTRACT

Background: Trauma leads to a complex inflammatory cascade that induces both immune activation and a refractory immune state in parallel. Although both components are deemed necessary for recovery, the balance is tight and easily lost. Losing the balance can lead to life-threatening infectious complications as well as long-term immunosuppression with recurrent infections. Neutrophils are known to play a key role in these processes. Therefore, this review focuses on neutrophil characteristics and function after trauma and how these features can be used to identify trauma patients at risk for infectious complications. Results: Distinct neutrophil subtypes exist that play their own role in the recovery and/or development of infectious complications after trauma. Furthermore, the refractory immune state is related to the risk of infectious complications. These findings change the initial concepts of the immune response after trauma and give rise to new biomarkers for monitoring and predicting inflammatory complications in severely injured patients. Conclusion: For early recognition of patients at risk, the immune system should be monitored. Several neutrophil biomarkers show promising results and analysis of these markers has become accessible to such extent that they can be used for point-of-care decision making after trauma.


Subject(s)
Communicable Diseases/classification , Genetic Heterogeneity , Neutrophils/classification , Communicable Diseases/physiopathology , Humans , Integrin alpha4beta1/genetics , Integrin alpha4beta1/metabolism , Wounds and Injuries/complications
14.
J Clin Invest ; 129(7): 2745-2759, 2019 05 14.
Article in English | MEDLINE | ID: mdl-31085833

ABSTRACT

Mobilized peripheral blood has become the primary source of hematopoietic stem and progenitor cells (HSPCs) for stem cell transplantation, with a five-day course of granulocyte colony stimulating factor (G-CSF) as the most common regimen used for HSPC mobilization. The CXCR4 inhibitor, plerixafor, is a more rapid mobilizer, yet not potent enough when used as a single agent, thus emphasizing the need for faster acting agents with more predictable mobilization responses and fewer side effects. We sought to improve hematopoietic stem cell transplantation by developing a new mobilization strategy in mice through combined targeting of the chemokine receptor CXCR2 and the very late antigen 4 (VLA4) integrin. Rapid and synergistic mobilization of HSPCs along with an enhanced recruitment of true HSCs was achieved when a CXCR2 agonist was co-administered in conjunction with a VLA4 inhibitor. Mechanistic studies revealed involvement of CXCR2 expressed on BM stroma in addition to stimulation of the receptor on granulocytes in the regulation of HSPC localization and egress. Given the rapid kinetics and potency of HSPC mobilization provided by the VLA4 inhibitor and CXCR2 agonist combination in mice compared to currently approved HSPC mobilization methods, it represents an exciting potential strategy for clinical development in the future.


Subject(s)
Bone Marrow/metabolism , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Integrin alpha4beta1 , Receptors, Interleukin-8B , Allografts , Animals , Granulocytes/metabolism , Integrin alpha4beta1/antagonists & inhibitors , Integrin alpha4beta1/genetics , Integrin alpha4beta1/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Receptors, Interleukin-8B/antagonists & inhibitors , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/metabolism
15.
Asian Pac J Cancer Prev ; 19(9): 2493-2498, 2018 Sep 26.
Article in English | MEDLINE | ID: mdl-30256042

ABSTRACT

Objective: To investigate the regulatory role of fibronectin (FN) in the formation of multicellular aggregate (MCA) in ovarian cancer SKOV3 and OVCAR-3 cells and integrin expression. Methods: The dynamic formation of MCA in SKOV3 and OVCAR-3 was determined using the liquid overlay technique in the presence or absence of FN, anti-FN, RGD peptide, control RGE. The expression of α3ß1, α4ß1 and α5ß1 integrin in monolayer cells, MCA and FN-treated MCA were determined by flow cytometry and quantitative RT-PCR. Results: OVCAR-3 and SKOV3 MCA were formed on the 4th and 8th day and peaked on the 6th and 9th day, respectively. Treatment with different concentrations of FN, LN, type IV collagen and control RGE peptide promoted MCA growth, which was mitigated by anti-FN and RGD peptide. In comparison with monolayer cells, up-regulated α3ß1, α4ß1 and α5ß1 expression were detected in MCA while treatment with FN in both cells. Conclusions: OVCAR-3 and SKOV3 cells had varying dynamic formation of MCA in our experimental system. FN enhanced MCA formation in both cells, which was associated with increased expression of 3ß1, α4ß1 and α5ß1 in the MCA. Therefore, FN and these integrins may be new therapeutic targets for intervention of ovarian cancer metastasis.


Subject(s)
Fibronectins/pharmacology , Gene Expression Regulation/drug effects , Integrin alpha3beta1/metabolism , Integrin alpha4beta1/metabolism , Integrin alpha5beta1/metabolism , Ovarian Neoplasms/pathology , Protein Aggregates/drug effects , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Female , Humans , Integrin alpha3beta1/genetics , Integrin alpha4beta1/genetics , Integrin alpha5beta1/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Tumor Cells, Cultured
16.
J Neuroinflammation ; 15(1): 236, 2018 Aug 22.
Article in English | MEDLINE | ID: mdl-30134924

ABSTRACT

BACKGROUND: Very late antigen 4 (VLA-4; integrin α4ß1) is critical for transmigration of T helper (TH) 1 cells into the central nervous system (CNS) under inflammatory conditions such as multiple sclerosis (MS). We have previously shown that VLA-4 and melanoma cell adhesion molecule (MCAM) are important for trans-endothelial migration of human TH17 cells in vitro and here investigate their contribution to pathogenic CNS inflammation. METHODS: Antibody blockade of VLA-4 and MCAM is assessed in murine models of CNS inflammation in conjunction with conditional ablation of α4-integrin expression in T cells. Effects of VLA-4 and MCAM blockade on lymphocyte migration are further investigated in the human system via in vitro T cell transmigration assays. RESULTS: Compared to the broad effects of VLA-4 blockade on encephalitogenic T cell migration over endothelial barriers, MCAM blockade impeded encephalitogenic T cell migration in murine models of MS that especially depend on CNS migration across the choroid plexus (CP). In transgenic mice lacking T cell α4-integrin expression (CD4::Itga4-/-), MCAM blockade delayed disease onset. Migration of MCAM-expressing T cells through the CP into the CNS was restricted, where laminin 411 (composed of α4, ß1, γ1 chains), the proposed major ligand of MCAM, is detected in the endothelial basement membranes of murine CP tissue. This finding was translated to the human system; blockade of MCAM with a therapeutic antibody reduced in vitro transmigration of MCAM-expressing T cells across a human fibroblast-derived extracellular matrix layer and a brain-derived endothelial monolayer, both expressing laminin α4. Laminin α4 was further detected in situ in CP endothelial-basement membranes in MS patients' brain tissue. CONCLUSIONS: Our findings suggest that MCAM-laminin 411 interactions facilitate trans-endothelial migration of MCAM-expressing T cells into the CNS, which seems to be highly relevant to migration via the CP and to potential future clinical applications in neuroinflammatory disorders.


Subject(s)
CD146 Antigen/metabolism , Choroid Plexus/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , T-Lymphocytes/drug effects , Animals , Antibodies/therapeutic use , CD146 Antigen/immunology , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Central Nervous System/pathology , Choroid Plexus/diagnostic imaging , Choroid Plexus/metabolism , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Endothelial Cells/drug effects , Freund's Adjuvant/toxicity , Humans , Integrin alpha4beta1/genetics , Integrin alpha4beta1/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin-Oligodendrocyte Glycoprotein/toxicity , Peptide Fragments/toxicity , Protein Kinases/genetics , Protein Kinases/metabolism
17.
J Clin Invest ; 128(10): 4359-4371, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30024857

ABSTRACT

JAK2-V617F-positive chronic myeloproliferative neoplasia (CMN) commonly displays dysfunction of integrins and adhesion molecules expressed on platelets, erythrocytes, and leukocytes. However, the mechanism by which the 2 major leukocyte integrin chains, ß1 and ß2, may contribute to CMN pathophysiology remained unclear. ß1 (α4ß1; VLA-4) and ß2 (αLß2; LFA-1) integrins are essential regulators for attachment of leukocytes to endothelial cells. We here showed enhanced adhesion of granulocytes from mice with JAK2-V617F knockin (JAK2+/VF mice) to vascular cell adhesion molecule 1- (VCAM1-) and intercellular adhesion molecule 1-coated (ICAM1-coated) surfaces. Soluble VCAM1 and ICAM1 ligand binding assays revealed increased affinity of ß1 and ß2 integrins for their respective ligands. For ß1 integrins, this correlated with a structural change from the low- to the high-affinity conformation induced by JAK2-V617F. JAK2-V617F triggered constitutive activation of the integrin inside-out signaling molecule Rap1, resulting in translocation toward the cell membrane. Employing a venous thrombosis model, we demonstrated that neutralizing anti-VLA-4 and anti-ß2 integrin antibodies suppress pathologic thrombosis as observed in JAK2+/VF mice. In addition, aberrant homing of JAK2+/VF leukocytes to the spleen was inhibited by neutralizing anti-ß2 antibodies and by pharmacologic inhibition of Rap1. Thus, our findings identified cross-talk between JAK2-V617F and integrin activation promoting pathologic thrombosis and abnormal trafficking of leukocytes to the spleen.


Subject(s)
CD18 Antigens/metabolism , Integrin beta1/metabolism , Janus Kinase 2/metabolism , Mutation, Missense , Venous Thrombosis/metabolism , Amino Acid Substitution , Animals , CD18 Antigens/genetics , Cell Adhesion , Integrin alpha4beta1/genetics , Integrin alpha4beta1/metabolism , Integrin beta1/genetics , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Janus Kinase 2/genetics , Leukocytes/metabolism , Leukocytes/pathology , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/metabolism , Mice , Mice, Mutant Strains , Spleen/metabolism , Spleen/pathology , Venous Thrombosis/genetics , Venous Thrombosis/pathology , rap1 GTP-Binding Proteins/genetics , rap1 GTP-Binding Proteins/metabolism
18.
Sci Rep ; 8(1): 10358, 2018 07 09.
Article in English | MEDLINE | ID: mdl-29985430

ABSTRACT

Fatty liver (FL) disease is the most common type of chronic liver disease. We hypothesized that liver's response to the process where large droplets of triglyceride fat accumulate in liver cells is reflected also in gene pathway expression in blood. Peripheral blood genome wide gene expression analysis and ultrasonic imaging of liver were performed for 1,650 participants (316 individuals with FL and 1,334 controls) of the Young Finns Study. Gene set enrichment analysis (GSEA) was performed for the expression data. Fourteen gene sets were upregulated (false discovery rate, FDR < 0.05) in subjects with FL. These pathways related to extracellular matrix (ECM) turnover, immune response regulation, prothrombotic state and neural tissues. After adjustment for known risk factors and biomarkers of FL, we found i) integrin A4B1 signaling, ii) leukocyte transendothelial migration, iii) CD40/CD40L and iv) netrin-1 signaling pathways to be upregulated in individuals with FL (nominal p < 0.05). From these all but not ii) remained significantly upregulated when analyzing only subjects without history of heavy alcohol use. In conclusion, FL was associated with blood gene sets of ECM turnover, inflammatory response, immune system activation and prothrombotic state. These may form a systemic link between FL and the development of cardiovascular diseases.


Subject(s)
Fatty Liver/blood , Immune System/metabolism , Signal Transduction/genetics , Adult , Biomarkers/blood , Biomarkers/metabolism , CD40 Antigens/genetics , CD40 Antigens/metabolism , CD40 Ligand/genetics , CD40 Ligand/metabolism , Case-Control Studies , Extracellular Matrix/metabolism , Fatty Liver/genetics , Fatty Liver/pathology , Female , Finland , Gene Expression Regulation , Humans , Integrin alpha4beta1/genetics , Integrin alpha4beta1/metabolism , Leukocytes/cytology , Leukocytes/physiology , Liver/diagnostic imaging , Liver/metabolism , Male , Middle Aged , Netrin-1/genetics , Netrin-1/metabolism , Risk Factors
19.
Mediators Inflamm ; 2018: 7623823, 2018.
Article in English | MEDLINE | ID: mdl-29769839

ABSTRACT

Alzheimer's disease (AD) is the most prevalent cause of dementia in elderly people worldwide. Many studies support the hypothesis that the inflammation of the CNS contributes to the neurodegeneration and disease progression. The integrin molecule α4ß1, also known as very late antigen 4 (VLA-4), belongs to adhesion molecules that activate the inflammatory process through the migration of immune cells into the CNS. Therefore, the objective of our study was to analyze the association between two polymorphisms located in the ITGA4 gene encoding the α4 subunit of VLA-4 and the risk of AD. 104 late-onset AD patients and 206 control subjects from Slovakia were genotyped for ITGA4 gene SNP polymorphism rs113276800 (-269C/A) and rs1143676 (+3061A/G). The same study cohorts were also genotyped for the APOE-ε4, which is a known genetic factor associated with increased risk of AD developing. ITGA4 polymorphism analysis revealed significantly higher frequency of the +3061AG carriers in AD group compared to the controls (P ≤ 0.05). Following the APOE-ε4 stratification of study groups, the association remained significant only in APOE-ε4 noncarriers. Our study suggests a novel association of ITGA4 +3061A/G polymorphism with AD and its possible contribution to the disease pathology.


Subject(s)
Alzheimer Disease/genetics , Integrin alpha4beta1/genetics , Aged , Aged, 80 and over , Alzheimer Disease/epidemiology , Female , Gene Frequency , Genetic Predisposition to Disease/genetics , Genotype , Humans , Male , Polymorphism, Genetic/genetics , Polymorphism, Single Nucleotide/genetics
20.
Mol Biotechnol ; 60(7): 455-467, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29730712

ABSTRACT

Umbilical cord blood (UCB)-derived hematopoietic stem cells (HSCs) are considered because of their self-renewing, differentiating, proliferating, and readily available properties. Moreover, HSCs' homing to the hematopoietic microenvironment is an important step in their transplantation process. But low content of progenitor cells in one unit of UCB and defect in the bone marrow (BM) homing limit their applications. Hence, we decided to correct this deficiency with ex vivo incubation of CD133+ cells using fucosyltransferase VI and GDP-fucose. Then C-X-C chemokines receptor-4 (CXCR4), very late activation antigen-4 (VLA4), very late activation antigen-5 (VLA5), lymphocyte function-associated antigen-1 (LFA-1), and E-cadherin (E-cad) genes expressions were investigated with the goal of homing evaluation. The purity of MACS isolated CD133+ cells and confirmation of fucosylation were done by flow cytometry, and the viability of cells seeded on protein-coated poly L-lactic acid (PLLA) scaffold was proven via MTT assay. Scanning electron microscopy (SEM), CFU assays, and expression assays of CXCR4, VLA4, VLA5, LFA-1 and E-cad by real-time PCR were performed, too. Flow cytometry data showed that isolated cells were suitable for fucosyltransferase VI (FT-VI) incubation and expansion on nanoscaffolds. MTT, CFU assays, and SEM micrographs demonstrated fibronectin (FN)-collagen-selectin (FCS)-coated scaffold serve as best environment for viability, clonogenicity, and cell attachment. High levels of homing genes expression were also observed in cells seeded on FCS-coated scaffolds. Also, CXCR4 flow cytometry analysis confirmed real-time data. FCS-PLLA scaffolds provided optimal conditions for viability of FT-VI-treated CD133+ cells, and clonogenicity with the goal of improving homing following UCB-HSCs transplantation.


Subject(s)
AC133 Antigen/analysis , Fetal Blood/cytology , Fucosyltransferases/pharmacology , Gene Expression Regulation/drug effects , Guanosine Diphosphate Fucose/pharmacology , Hematopoietic Stem Cells/drug effects , AC133 Antigen/drug effects , Cadherins/genetics , Cadherins/metabolism , Cell Movement , Cell Survival/drug effects , Cells, Cultured , Cellular Microenvironment , Hematopoietic Stem Cells/metabolism , Humans , Integrin alpha4beta1/genetics , Integrin alpha4beta1/metabolism , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/metabolism , Nanofibers , Tissue Scaffolds
SELECTION OF CITATIONS
SEARCH DETAIL
...