Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
J Immunol ; 205(10): 2883-2892, 2020 11 15.
Article in English | MEDLINE | ID: mdl-33077644

ABSTRACT

CD98, which is required for the rapid proliferation of both normal and cancer cells, and MET, the hepatocyte growth factor receptor, are potential targets for therapeutic antitumor Abs. In this study, we report that the antiproliferative activity of a prototype anti-CD98 Ab, UM7F8, is due to Ab-induced membrane-associated ring CH (MARCH) E3 ubiquitin ligase-mediated ubiquitination and downregulation of cell surface CD98. MARCH1-mediated ubiquitination of CD98 is required for UM7F8's capacity to reduce CD98 surface expression and its capacity to inhibit the proliferation of murine T cells. Similarly, CD98 ubiquitination is required for UM7F8's capacity to block the colony-forming ability of murine leukemia-initiating cells. To test the potential generality of the paradigm that MARCH E3 ligases can mediate the antiproliferative response to antitumor Abs, we examined the potential effects of MARCH proteins on responses to emibetuzumab, an anti-MET Ab currently in clinical trials for various cancers. We report that MET surface expression is reduced by MARCH1, 4, or 8-mediated ubiquitination and that emibetuzumab-induced MET ubiquitination contributes to its capacity to downregulate MET and inhibit human tumor cell proliferation. Thus, MARCH E3 ligases can act as cofactors for antitumor Abs that target cell surface proteins, suggesting that the MARCH protein repertoire of cells is a determinant of their response to such Abs.


Subject(s)
Antigens, Neoplasm/metabolism , Antineoplastic Agents, Immunological/pharmacology , Fusion Regulatory Protein 1, Heavy Chain/metabolism , Neoplasms/drug therapy , Ubiquitin-Protein Ligases/metabolism , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antineoplastic Agents, Immunological/therapeutic use , Cell Proliferation/drug effects , Fusion Regulatory Protein 1, Heavy Chain/antagonists & inhibitors , Fusion Regulatory Protein 1, Heavy Chain/genetics , Fusion Regulatory Protein 1, Heavy Chain/immunology , Gene Knockout Techniques , HeLa Cells , Humans , Jurkat Cells , Mice , Mice, Knockout , Neoplasms/immunology , Neoplasms/pathology , Proteolysis , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/genetics , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Ubiquitin-Protein Ligases/genetics , Ubiquitination/immunology
2.
J Immunol ; 200(12): 4012-4023, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29703862

ABSTRACT

Maintenance of the regulatory T (Treg) cell pool is essential for peripheral tolerance and prevention of autoimmunity. Integrins, heterodimeric transmembrane proteins consisting of α and ß subunits that mediate cell-to-cell and cell-to-extracellular matrix interactions, play an important role in facilitating Treg cell contact-mediated suppression. In this article, we show that integrin activation plays an essential, previously unappreciated role in maintaining murine Treg cell function. Treg cell-specific loss of talin, a ß integrin-binding protein, or expression of talin(L325R), a mutant that selectively abrogates integrin activation, resulted in lethal systemic autoimmunity. This dysfunction could be attributed, in part, to a global dysregulation of the Treg cell transcriptome. Activation of integrin α4ß1 led to increased suppressive capacity of the Treg cell pool, suggesting that modulating integrin activation on Treg cells may be a useful therapeutic strategy for autoimmune and inflammatory disorders. Taken together, these results reveal a critical role for integrin-mediated signals in controlling peripheral tolerance by virtue of maintaining Treg cell function.


Subject(s)
Integrins/immunology , Peripheral Tolerance/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmunity/immunology , Inflammation/immunology , Mice , Talin/immunology , Transcriptome/immunology
3.
J Immunol ; 198(9): 3410-3415, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28348273

ABSTRACT

Rap1-interacting adaptor molecule (RIAM) is a Rap1 effector that mediates the recruitment of talin to integrins, thereby supporting their activation. In this study, we investigated the role of RIAM in an adoptive transfer model for type I diabetes and report that RIAM expression in T cells is necessary for diabetes development. Loss of RIAM did not prevent lymphocyte recruitment to draining lymph nodes 24 h after transfer, but it was required for Ag-driven proliferation and cytotoxic killing. RIAM is recruited to immune synapses along with talin and LFA-1, and loss of RIAM profoundly suppresses Ag-dependent conjugate formation in primary naive and effector T cells. These data identify the requirement of RIAM for formation of immunological synapses and in resulting T cell functions in autoimmunity. Moreover, because RIAM-null mice are healthy, fertile, and display no bleeding abnormalities, our results identify RIAM and its regulators as potential targets for therapies of T cell-mediated autoimmunity.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Antigen-Presenting Cells/immunology , Diabetes Mellitus, Type 1/immunology , Lymphocyte Function-Associated Antigen-1/metabolism , Membrane Proteins/metabolism , T-Lymphocytes/immunology , Talin/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adoptive Transfer , Animals , Cell Proliferation/genetics , Cells, Cultured , Cytotoxicity, Immunologic/genetics , Humans , Immunological Synapses/immunology , Membrane Proteins/genetics , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/transplantation
4.
Cancer Cell ; 30(5): 792-805, 2016 Nov 14.
Article in English | MEDLINE | ID: mdl-27908736

ABSTRACT

Acute myelogenous leukemia (AML) is an aggressive disease associated with drug resistance and relapse. To improve therapeutic strategies, it is critical to better understand the mechanisms that underlie AML progression. Here we show that the integrin binding glycoprotein CD98 plays a central role in AML. CD98 promotes AML propagation and lethality by driving engagement of leukemia cells with their microenvironment and maintaining leukemic stem cells. Further, delivery of a humanized anti-CD98 antibody blocks growth of patient-derived AML, highlighting the importance of this pathway in human disease. These findings indicate that microenvironmental interactions are key regulators of AML and that disrupting these signals with targeted inhibitors such as CD98 antibodies may be a valuable therapeutic approach for adults and children with this disease.


Subject(s)
Antibodies/administration & dosage , Fusion Regulatory Protein-1/genetics , Leukemia, Myeloid, Acute/drug therapy , Neoplastic Stem Cells/pathology , Animals , Antibodies/pharmacology , Cell Adhesion , Cell Line, Tumor , Cell Proliferation/drug effects , Fusion Regulatory Protein-1/antagonists & inhibitors , Gene Knockout Techniques , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Neoplasm Transplantation
5.
Arterioscler Thromb Vasc Biol ; 36(11): 2163-2166, 2016 11.
Article in English | MEDLINE | ID: mdl-27687603

ABSTRACT

OBJECTIVE: CD98 regulates integrin signaling and is critical for tumor cell proliferation. It is also expressed on endothelial cells (EC), but its role in angiogenesis is unclear. APPROACH AND RESULTS: We used specific genetic targeting and antibody blockade approaches to examine the function of CD98 in EC proliferation, blood vessel growth, and tumor angiogenesis. It is upregulated on angiogenic ECs, and EC-specific deletion of CD98 in mice inhibited tumor growth, retinal angiogenesis, and EC proliferation. Reconstitution with CD98 mutants showed that integrin and CD98 interaction is necessary for EC survival and growth. Moreover, anti-CD98 treatment inhibited vessel formation and reversed EC-assisted tumor growth. CONCLUSIONS: Our findings demonstrate a requirement for CD98 in EC growth and suggest that CD98-specific reagents could have a dual anticancer effect: directly by inhibiting tumor cell proliferation and indirectly by preventing tumor angiogenesis.


Subject(s)
Carcinoma, Lewis Lung/metabolism , Fusion Regulatory Protein 1, Heavy Chain/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Melanoma, Experimental/metabolism , Neovascularization, Pathologic , Neovascularization, Physiologic , Retinal Neovascularization/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Antibodies/pharmacology , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/genetics , Cell Line, Tumor , Cell Proliferation , Female , Fusion Regulatory Protein 1, Heavy Chain/genetics , Fusion Regulatory Protein 1, Heavy Chain/immunology , Genotype , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/transplantation , Humans , Integrins/metabolism , Male , Melanoma, Experimental/blood supply , Melanoma, Experimental/drug therapy , Melanoma, Experimental/genetics , Mice, Knockout , Neovascularization, Physiologic/drug effects , Phenotype , Phosphorylation , Retinal Neovascularization/drug therapy , Retinal Neovascularization/genetics , Retinal Neovascularization/pathology , Signal Transduction , Time Factors , Tumor Burden , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism
6.
Diabetes ; 65(11): 3505-3515, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27495221

ABSTRACT

Diabetes is associated with a deficit of circulating endothelial progenitor cells (EPCs), which has been attributed to their defective mobilization from the bone marrow. The basis for this mobilization defect is not completely understood, and we sought to determine if hyperglycemic conditions enhanced EPC adhesion. We found that culturing EPCs in high glucose media increased adhesion to bone marrow stromal cells. This enhanced adhesion was associated with decreased expression of protein kinase A regulatory subunit 1ß (PRKAR1ß), activation of protein kinase A (PKA), and phosphorylation of α4-integrin on serine 988. This potentiated adhesion was reversed by treatment with a PKA inhibitor, overexpression of PRKAR1ß, or expression of a phosphorylation-defective α4-integrin variant (α4[S988A]). Using a model of type 1 diabetes, we showed that α4(S988A)-expressing mice have more circulating EPCs than their wild-type counterparts. Moreover, diabetic α4(S988A) mice demonstrate enhanced revascularization after hind limb ischemia. Thus, we have identified a novel signaling mechanism activating PKA in diabetes (downregulation of an inhibitory regulatory subunit) that leads to deficits of circulating EPCs and impaired vascular repair, which could be reversed by α4-integrin mutation.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Endothelial Progenitor Cells/metabolism , Integrin alpha4beta1/metabolism , Animals , Cell Adhesion/genetics , Cell Adhesion/physiology , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Diabetes Mellitus, Experimental/genetics , Hindlimb/pathology , Integrin alpha4beta1/genetics , Ischemia/metabolism , Male , Mice , Mutation/genetics , Phosphorylation , Signal Transduction/genetics , Signal Transduction/physiology
7.
J Cell Sci ; 128(23): 4273-8, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26493331

ABSTRACT

CD98 heavy chain (SLC3A2) facilitates lymphocyte clonal expansion that enables adaptive immunity; however, increased expression of CD98 is also a feature of both lymphomas and leukemias and represents a potential therapeutic target in these diseases. CD98 is transcriptionally regulated and ectopic expression of the membrane-associated RING-CH (MARCH) E3 ubiquitin ligases MARCH1 or MARCH8 leads to ubiquitylation and lysosomal degradation of CD98. Here, we examined the potential role of ubiquitylation in regulating CD98 expression and cell proliferation. We report that blocking ubiquitylation by use of a catalytically inactive MARCH or by creating a ubiquitylation-resistant CD98 mutant, prevents MARCH-induced CD98 downregulation in HeLa cells. March1-null T cells display increased CD98 expression. Similarly, T cells expressing ubiquitylation-resistant CD98 manifest increased proliferation in vitro and clonal expansion in vivo. Thus, ubiquitylation and the resulting downregulation of CD98 can limit cell proliferation and clonal expansion.


Subject(s)
Cell Proliferation/physiology , Fusion Regulatory Protein 1, Heavy Chain/metabolism , Lysosomes/metabolism , Proteolysis , Ubiquitination/physiology , Animals , Fusion Regulatory Protein 1, Heavy Chain/genetics , HeLa Cells , Humans , Jurkat Cells , Lysosomes/genetics , Mice , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
8.
Int J Cancer ; 137(3): 710-20, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-25556716

ABSTRACT

CD98 is expressed on several tissue types and specifically upregulated on fast-cycling cells undergoing clonal expansion. Various solid (e.g., nonsmall cell lung carcinoma) as well as hematological malignancies (e.g., acute myeloid leukemia) overexpress CD98. We have identified a CD98-specific mouse monoclonal antibody that exhibits potent preclinical antitumor activity against established lymphoma tumor xenografts. Additionally, the humanized antibody designated IGN523 demonstrated robust tumor growth inhibition in leukemic cell-line derived xenograft models and was as efficacious as standard of care carboplatin in patient-derived nonsmall lung cancer xenografts. In vitro studies revealed that IGN523 elicited strong ADCC activity, induced lysosomal membrane permeabilization and inhibited essential amino acid transport function, ultimately resulting in caspase-3 and -7-mediated apoptosis of tumor cells. IGN523 is currently being evaluated in a Phase I clinical trial for acute myeloid leukemia (NCT02040506). Furthermore, preclinical data support the therapeutic potential of IGN523 in solid tumors.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Fusion Regulatory Protein-1/antagonists & inhibitors , Amino Acids/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Antibody-Dependent Cell Cytotoxicity/immunology , Antineoplastic Agents/administration & dosage , Biological Transport , Caspases/metabolism , Cell Line, Tumor , Cell Membrane Permeability , Complement System Proteins/immunology , Cytotoxicity, Immunologic , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Humans , Lysosomes/metabolism , Mice , Models, Biological , Protein Binding , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
9.
Cancer Immunol Res ; 3(6): 661-7, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25600437

ABSTRACT

Inefficient T-cell homing to tissues limits adoptive T-cell immunotherapy of solid tumors. αLß2 and α4ß1 integrins mediate trafficking of T cells into tissues via engagement of ICAM-1 and VCAM-1, respectively. Inhibiting protein kinase A (PKA)-mediated phosphorylation of α4 integrin in cells results in an increase in αLß2-mediated migration on mixed ICAM-1-VCAM-1 substrates in vitro, a phenomenon termed "integrin trans-regulation." Here, we created an α4(S988A)-bearing mouse, which precludes PKA-mediated α4 phosphorylation, to examine the effect of integrin trans-regulation in vivo. The α4(S988A) mouse exhibited a dramatic and selective increase in migration of lymphocytes, but not myeloid cells, to sites of inflammation. Importantly, we found that the α4(S988A) mice exhibited a marked increase in T-cell entry into and reduced growth of B16 melanomas, consistent with antitumor roles of infiltrating T cells and progrowth functions of tumor-associated macrophages. Thus, increased α4 trans-regulation of αLß2 integrin function biases leukocyte emigration toward lymphocytes relative to myeloid cells and enhances tumor immunity.


Subject(s)
Integrins/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Animals , Chemotaxis, Leukocyte/immunology , Disease Models, Animal , Humans , Integrin alpha4/genetics , Integrin alpha4/metabolism , Integrins/genetics , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/metabolism , Mice , Mice, Transgenic , Mutation , Neoplasms/genetics , Neoplasms/pathology , Protein Binding , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tumor Burden
10.
Blood ; 125(12): 1995-2004, 2015 Mar 19.
Article in English | MEDLINE | ID: mdl-25587038

ABSTRACT

The bidirectional signaling and hemostatic functions of platelet αIIbß3 are regulated by kindlin-3 through interactions with the ß3 cytoplasmic tail. Little is known about kindlin regulation of the related "vitronectin receptor," αVß3. These relationships were investigated in endothelial cells, which express αVß3 and kindlin-2 endogenously. "ß3ΔRGT" knock-in mice lack the 3 C-terminal ß3 tail residues, whereas in "ß3/ß1(EGK)" mice, RGT is replaced by the corresponding residues of ß1. The wild-type ß3 tail pulled down kindlin-2 and c-Src in vitro, whereas ß3ΔRGT bound neither protein and ß3/ß1(EGK) bound kindlin-2, but not c-Src. ß3ΔRGT endothelial cells, but not ß3/ß1(EGK) endothelial cells, exhibited migration and spreading defects on vitronectin and reduced sprouting in 3-dimensional fibrin. Short hairpin RNA silencing of kindlin-2, but not c-Src, blocked sprouting by ß3 wild-type endothelial cells. Moreover, defective sprouting by ß3ΔRGT endothelial cells could be rescued by conditional, forced interaction of αVß3ΔRGT with kindlin-2. Stimulation of ß3ΔRGT endothelial cells led to normal extracellular ligand binding to αVß3, pin-pointing their defect to one of outside-in αVß3 signaling. ß3ΔRGT mice, but not ß3/ß1(EGK) mice, exhibited defects in both developmental and tumor angiogenesis, responses that require endothelial cell function. Thus, the ß3/kindlin-2 interaction promotes outside-in αVß3 signaling selectively, with biological consequences in vivo.


Subject(s)
Cytoskeletal Proteins/metabolism , Integrin alphaVbeta3/metabolism , Integrin beta3/metabolism , Muscle Proteins/metabolism , Animals , Blood Platelets/metabolism , Bone Marrow Transplantation , Cell Movement , Cytoplasm/metabolism , Endothelial Cells , Humans , Melanoma, Experimental , Membrane Proteins/metabolism , Mice , Mutation , Neoplasm Proteins/metabolism , Neoplasm Transplantation , Neovascularization, Pathologic , Protein Binding , Protein Structure, Tertiary , RNA Interference , Signal Transduction
11.
J Neuroimmunol ; 274(1-2): 230-3, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25002078

ABSTRACT

Current B cell-directed therapies for multiple sclerosis impact multiple B cell functions. CD98hc enables B cell clonal expansion and antibody production. I probed the relative importance of autoantibody secretion vs. other B cell functions in MS and targeted CD98hc as a possible therapeutic strategy. I report that the loss of CD98hc function in B cells largely prevents autoantibody production while preserving antigen-presenting and T cell-directing capacities. Mice lacking CD98hc in B cells are protected from EAE; importantly this is overcome with autoantibody-containing plasma. Thus CD98hc blockade is a possible avenue to treat MS by inhibiting clonal expansion and autoantibody.


Subject(s)
Autoantibodies/immunology , B-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Fusion Regulatory Protein-1/immunology , Multiple Sclerosis/immunology , Animals , Autoantibodies/blood , B-Lymphocytes/cytology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Humans , Mice , Mice, Mutant Strains , Multiple Sclerosis/drug therapy , Myelin-Oligodendrocyte Glycoprotein/immunology , Myelin-Oligodendrocyte Glycoprotein/pharmacology , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology
12.
J Cell Sci ; 125(Pt 6): 1373-82, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22499670

ABSTRACT

Adaptive immunity, a vertebrate specialization, adds memory and exquisite specificity to the basic innate immune responses present in invertebrates while conserving metabolic resources. In adaptive immunity, antigenic challenge requires extremely rapid proliferation of rare antigen-specific lymphocytes to produce large, clonally expanded effector populations that neutralize pathogens. Rapid proliferation and resulting clonal expansion are dependent on CD98, a protein whose well-conserved orthologs appear restricted to vertebrates. Thus, CD98 supports lymphocyte clonal expansion to enable protective adaptive immunity, an advantage that could account for the presence of CD98 in vertebrates. CD98 supports lymphocyte clonal expansion by amplifying integrin signals that enable proliferation and prevent apoptosis. These integrin-dependent signals can also provoke cancer development and invasion, anchorage-independence and the rapid proliferation of tumor cells. CD98 is highly expressed in many cancers and contributes to formation of tumors in experimental models. Strikingly, vertebrates, which possess highly conserved CD98 proteins, CD98-binding integrins and adaptive immunity, also display propensity towards invasive and metastatic tumors. In this Commentary, we review the roles of CD98 in lymphocyte biology and cancer. We suggest that the CD98 amplification of integrin signaling in adaptive immunity provides survival benefits to vertebrates, which, in turn, bear the price of increased susceptibility to cancer.


Subject(s)
Adaptive Immunity/physiology , Fusion Regulatory Protein-1/physiology , Neoplasms/immunology , Neoplasms/metabolism , Animals , Cell Proliferation , Clone Cells , Humans , Integrins/physiology
13.
Immunol Rev ; 223: 236-51, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18613840

ABSTRACT

SUMMARY: Integrins are adhesion receptors important for hematopoiesis, leukocyte trafficking, and formation of immunological synapses; hence, they may provide targets for therapeutic intervention in leukocyte-driven pathologies. Blocking integrin-ligand binding is one strategy for inhibiting integrins; however, a complete loss of integrin function can lead to mechanism-based toxicities. Because integrin alpha and beta subunits interact with a variety of other proteins to receive and transmit cellular signals, targeting these integrin-associated proteins may utilize alternative sites for intervention that lead to therapies with fewer side effects. This review summarizes integrin-associated proteins in leukocytes and focuses on four of these proteins with perceived therapeutic potential. Specific mutations in the alpha4 integrin cytoplasmic tail block or enforce binding to paxillin and thus modulate integrin signaling required for efficient cell migration. Similarly, the association of RAPL(NORE1B) with beta2 integrins may participate in adhesive and migratory events in leukocytes. The beta integrin cytoplasmic tail-binding protein talin is critical for increasing the affinity of integrins (activation), and blockade of talin binding can prevent leukocyte arrest on the endothelium. Finally, the membrane protein CD98 mediates beta1 and beta3 integrin signaling and may be involved in leukocyte functions. Identification of biologically important interactions of integrins and signaling proteins can thus pave the way to new strategies for manipulating leukocyte functions.


Subject(s)
Cell Adhesion/immunology , Integrins/immunology , Monomeric GTP-Binding Proteins/immunology , Paxillin/immunology , Adaptor Proteins, Signal Transducing , Animals , Apoptosis Regulatory Proteins , Cell Adhesion/genetics , Cell Movement/immunology , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Fusion Regulatory Protein-1/immunology , Fusion Regulatory Protein-1/metabolism , Humans , Integrins/chemistry , Integrins/genetics , Integrins/metabolism , Leukocytes/cytology , Leukocytes/enzymology , Leukocytes/metabolism , Monomeric GTP-Binding Proteins/metabolism , Paxillin/metabolism , Protein Binding , Talin/immunology , Talin/metabolism , Thrombosis/immunology
SELECTION OF CITATIONS
SEARCH DETAIL