1,196 results on '"Ginsberg, Mark H."'
Search Results
2. IL-2 can signal via chemokine receptors to promote regulatory T cells’ suppressive function
- Author
-
Sun, Hao, Lee, Ho-Sup, Kim, Sarah Hyun-Ji, de Lima, Mikhael Fernandes, Gingras, Alexandre R, Du, Qinyi, McLaughlin, Wilma, Ablack, Jailail, Lopez-Ramirez, Miguel A, Lagarrigue, Frederic, Fan, Zhichao, Chang, John T, VanDyke, Derek, Spangler, Jamie B, and Ginsberg, Mark H
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Neurodegenerative ,Neurosciences ,Aetiology ,Underpinning research ,2.1 Biological and endogenous factors ,1.1 Normal biological development and functioning ,Inflammatory and immune system ,Mice ,Animals ,T-Lymphocytes ,Regulatory ,Interleukin-2 ,Receptors ,Chemokine ,Interleukin-2 Receptor alpha Subunit ,Receptors ,Interleukin-2 ,Signal Transduction ,Encephalomyelitis ,Autoimmune ,Experimental ,Forkhead Transcription Factors ,CD25 ,CP: Immunology ,IL-2 ,IL-2 receptor ,autoimmunity ,chemokine receptor ,experimental autoimmune encephalomyelitis ,heparan sulfate ,integrins ,regulatory T cells ,signal transduction ,Medical Physiology ,Biological sciences - Abstract
Canonical interleukin-2 (IL-2) signaling via the high-affinity CD25-containing IL-2 receptor-Janus kinase (JAK)1,3-signal transducer and activator of transcription 5 (STAT5) pathway is essential for development and maintenance of CD4+CD25HiFoxp3+ regulatory T cells (Tregs) that support immune homeostasis. Here, we report that IL-2 signaling via an alternative CD25-chemokine receptor pathway promotes the suppressive function of Tregs. Using an antibody against CD25 that biases IL-2 signaling toward this alternative pathway, we establish that this pathway increases the suppressive activity of Tregs and ameliorates murine experimental autoimmune encephalomyelitis (EAE). Furthermore, heparan sulfate, an IL-2-binding element of cell surfaces and extracellular matrix, or an engineered IL-2 immunocytokine can also direct IL-2 signaling toward this alternative pathway. Overall, these data reveal a non-canonical mechanism for IL-2 signaling that promotes suppressive functions of Tregs, further elucidates how IL-2 supports immune homeostasis, and suggests approaches to promote or suppress Treg functions.
- Published
- 2023
3. Mild Hypoxia Accelerates Cerebral Cavernous Malformation Disease Through CX3CR1-CX3CL1 Signaling
- Author
-
Frias-Anaya, Eduardo, Gallego-Gutierrez, Helios, Gongol, Brendan, Weinsheimer, Shantel, Lai, Catherine Chinhchu, Orecchioni, Marco, Sriram, Aditya, Bui, Cassandra M., Nelsen, Bliss, Hale, Preston, Pham, Angela, Shenkar, Robert, DeBiasse, Dorothy, Lightle, Rhonda, Girard, Romuald, Li, Ying, Srinath, Abhinav, Daneman, Richard, Nudleman, Eric, Sun, Hao, Guma, Monica, Dubrac, Alexandre, Mesarwi, Omar A., Ley, Klaus, Kim, Helen, Awad, Issam A., Ginsberg, Mark H., and Lopez-Ramirez, Miguel Alejandro
- Published
- 2024
- Full Text
- View/download PDF
4. Phostensin enables lymphocyte integrin activation and population of peripheral lymphoid organs
- Author
-
Lee, Ho-Sup, Sun, Hao, Lagarrigue, Frédéric, Kim, Sarah Hyun Ji, Fox, Jay W, Sherman, Nicholas E, Gingras, Alexandre R, and Ginsberg, Mark H
- Subjects
Aetiology ,1.1 Normal biological development and functioning ,Underpinning research ,2.1 Biological and endogenous factors ,Adaptor Proteins ,Signal Transducing ,Animals ,Cell Adhesion ,Colitis ,Integrins ,Lymphoid Tissue ,Membrane Proteins ,Mice ,Protein Phosphatase 1 ,T-Lymphocytes ,Talin ,rap1 GTP-Binding Proteins ,Medical and Health Sciences ,Immunology - Abstract
Rap1 GTPase drives assembly of the Mig-10/RIAM/Lamellipodin (MRL protein)-integrin-talin (MIT) complex that enables integrin-dependent lymphocyte functions. Here we used tandem affinity tag-based proteomics to isolate and analyze the MIT complex and reveal that Phostensin (Ptsn), a regulatory subunit of protein phosphatase 1, is a component of the complex. Ptsn mediates dephosphorylation of Rap1, thereby preserving the activity and membrane localization of Rap1 to stabilize the MIT complex. CRISPR/Cas9-induced deletion of PPP1R18, which encodes Ptsn, markedly suppresses integrin activation in Jurkat human T cells. We generated apparently healthy Ppp1r18-/- mice that manifest lymphocytosis and reduced population of peripheral lymphoid tissues ascribable, in part, to defective activation of integrins αLβ2 and α4β7. Ppp1r18-/- T cells exhibit reduced capacity to induce colitis in a murine adoptive transfer model. Thus, Ptsn enables lymphocyte integrin-mediated functions by dephosphorylating Rap1 to stabilize the MIT complex. As a consequence, loss of Ptsn ameliorates T cell-mediated colitis.
- Published
- 2022
5. Circulating Plasma miRNA Homologs in Mice and Humans Reflect Familial Cerebral Cavernous Malformation Disease
- Author
-
Romanos, Sharbel G., Srinath, Abhinav, Li, Ying, Xie, Bingqing, Chen, Chang, Li, Yan, Moore, Thomas, Bi, Dehua, Sone, Je Yeong, Lightle, Rhonda, Hobson, Nick, Zhang, Dongdong, Koskimäki, Janne, Shen, Le, McCurdy, Sara, Lai, Catherine Chinhchu, Stadnik, Agnieszka, Piedad, Kristina, Carrión-Penagos, Julián, Shkoukani, Abdallah, Snellings, Daniel, Shenkar, Robert, Sulakhe, Dinanath, Ji, Yuan, Lopez-Ramirez, Miguel A., Kahn, Mark L., Marchuk, Douglas A., Ginsberg, Mark H., Girard, Romuald, and Awad, Issam A.
- Published
- 2023
- Full Text
- View/download PDF
6. Protease-activated receptors and glycoprotein VI cooperatively drive the platelet component in thromboelastography
- Author
-
Rudran, Tanvi, Antoniak, Silvio, Flick, Matthew J., Ginsberg, Mark H., Wolberg, Alisa S., Bergmeier, Wolfgang, and Lee, Robert H.
- Published
- 2023
- Full Text
- View/download PDF
7. A stem cell reporter based platform to identify and target drug resistant stem cells in myeloid leukemia.
- Author
-
Spinler, Kyle, Bajaj, Jeevisha, Ito, Takahiro, Zimdahl, Bryan, Hamilton, Michael, Ahmadi, Armin, Koechlein, Claire S, Lytle, Nikki, Kwon, Hyog Young, Anower-E-Khuda, Ferdous, Sun, Hao, Blevins, Allen, Weeks, Joi, Kritzik, Marcie, Karlseder, Jan, Ginsberg, Mark H, Park, Pyong Woo, Esko, Jeffrey D, and Reya, Tannishtha
- Subjects
Animals ,Mice ,Transgenic ,Humans ,Blast Crisis ,Disease Models ,Animal ,RNA-Binding Proteins ,Green Fluorescent Proteins ,Integrin beta Chains ,Antineoplastic Agents ,Signal Transduction ,Drug Resistance ,Neoplasm ,Genes ,Reporter ,Syndecan-1 ,Neoplastic Stem Cells ,Leukemia ,Myeloid ,Acute ,Gene Knockout Techniques ,Gene Knock-In Techniques ,High-Throughput Screening Assays ,Chemoradiotherapy ,Imatinib Mesylate ,RNA-Seq ,Stem Cell Research ,Regenerative Medicine ,Biotechnology ,Hematology ,Pediatric Cancer ,Childhood Leukemia ,Stem Cell Research - Nonembryonic - Human ,Pediatric Research Initiative ,Pediatric ,Stem Cell Research - Nonembryonic - Non-Human ,Cancer ,Rare Diseases ,2.1 Biological and endogenous factors ,1.1 Normal biological development and functioning - Abstract
Intratumoral heterogeneity is a common feature of many myeloid leukemias and a significant reason for treatment failure and relapse. Thus, identifying the cells responsible for residual disease and leukemia re-growth is critical to better understanding how they are regulated. Here, we show that a knock-in reporter mouse for the stem cell gene Musashi 2 (Msi2) allows identification of leukemia stem cells in aggressive myeloid malignancies, and provides a strategy for defining their core dependencies. Specifically, we carry out a high throughput screen using Msi2-reporter blast crisis chronic myeloid leukemia (bcCML) and identify several adhesion molecules that are preferentially expressed in therapy resistant bcCML cells and play a key role in bcCML. In particular, we focus on syndecan-1, whose deletion triggers defects in bcCML growth and propagation and markedly improves survival of transplanted mice. Further, live imaging reveals that the spatiotemporal dynamics of leukemia cells are critically dependent on syndecan signaling, as loss of this signal impairs their localization, migration and dissemination to distant sites. Finally, at a molecular level, syndecan loss directly impairs integrin β7 function, suggesting that syndecan exerts its influence, at least in part, by coordinating integrin activity in bcCML. These data present a platform for delineating the biological underpinnings of leukemia stem cell function, and highlight the Sdc1-Itgβ7 signaling axis as a key regulatory control point for bcCML growth and dissemination.
- Published
- 2020
8. MARCH Proteins Mediate Responses to Antitumor Antibodies.
- Author
-
Ablack, Jailal N, Ortiz, Jesus, Bajaj, Jeevisha, Trinh, Kathleen, Lagarrigue, Frederic, Cantor, Joseph M, Reya, Tannishtha, and Ginsberg, Mark H
- Subjects
Cancer ,Aetiology ,2.1 Biological and endogenous factors ,Animals ,Antibodies ,Monoclonal ,Humanized ,Antigens ,Neoplasm ,Antineoplastic Agents ,Immunological ,Cell Proliferation ,Fusion Regulatory Protein 1 ,Heavy Chain ,Gene Knockout Techniques ,HeLa Cells ,Humans ,Jurkat Cells ,Mice ,Mice ,Knockout ,Neoplasms ,Proteolysis ,Proto-Oncogene Proteins c-met ,T-Lymphocytes ,Ubiquitin-Protein Ligases ,Ubiquitination ,Hela Cells ,Immunology - 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.
- Published
- 2020
9. Frontline Science: A flexible kink in the transmembrane domain impairs β2 integrin extension and cell arrest from rolling
- Author
-
Sun, Hao, Fan, Zhichao, Gingras, Alexandre R, Lopez-Ramirez, Miguel A, Ginsberg, Mark H, and Ley, Klaus
- Subjects
Biomedical and Clinical Sciences ,Immunology ,CD18 Antigens ,Cell Cycle Checkpoints ,HL-60 Cells ,Humans ,Intercellular Adhesion Molecule-1 ,Interleukin-8 ,Leukocyte Rolling ,Mutation ,P-Selectin ,Proline ,Protein Conformation ,Protein Domains ,talin ,activation ,affinity ,adhesion ,spreading ,Biochemistry and Cell Biology - Abstract
β2 integrins are the main adhesion molecules in neutrophils and other leukocytes and are rapidly activated by inside-out signaling, which results in conformational changes that are transmitted through the transmembrane domain (TMD). Here, we investigated the biologic effect of introducing a proline mutation in the β2 integrin TMD to create a flexible kink that uncouples the topology of the inner half of the TMD from the outer half and impairs integrin activation. The β2 integrin alpha chains, αL, αM, αX, and αD, all contain an inserted (I) domain with homology to von Willebrand factor A domain. β2 activation was monitored in a homogenous binding assay of 2 reporter monoclonal antibodies: KIM127 reporting extension (E+ ) and mAb24 reporting the high-affinity (H+ ) conformation of the β2 I-like domain. The proline mutation partially diminished chemokine-induced extension, but not the high-affinity conformation. The proline mutation in the TMD of β2 completely inhibited arrest of rolling HL-60 cells in response to the chemokine IL-8. TMD mutant HL-60 cells rolling on P-selectin and ICAM-1 were unable to reduce their rolling velocity in response to IL-8. Quantitative dynamic footprinting live-cell imaging showed that blocking TMD topology transmission impaired the chemokine-induced activation of β2, limiting the appearance of extended high-affinity (E+ H+ ) β2. This also resulted in a defect in early spreading (3 min after arrest), which could be overcome by forced integrin activation using Mn2+ . We conclude that the TMD proline mutation severely impairs β2 integrin extension, cell arrest, and early spreading.
- Published
- 2020
10. β7 Integrin Inhibition Can Increase Intestinal Inflammation by Impairing Homing of CD25hiFoxP3+ Regulatory T Cells
- Author
-
Sun, Hao, Kuk, Wun, Rivera-Nieves, Jesús, Lopez-Ramirez, Miguel Alejandro, Eckmann, Lars, and Ginsberg, Mark H
- Subjects
Biomedical and Clinical Sciences ,Immunology ,Crohn's Disease ,Autoimmune Disease ,Digestive Diseases ,Inflammatory Bowel Disease ,Aetiology ,2.1 Biological and endogenous factors ,Inflammatory and immune system ,Oral and gastrointestinal ,Animals ,Cell Adhesion ,Colitis ,Ulcerative ,Colon ,Dextran Sulfate ,Disease Models ,Animal ,Forkhead Transcription Factors ,Humans ,Integrin beta Chains ,Interleukin-10 ,Interleukin-2 Receptor alpha Subunit ,Intestinal Mucosa ,Mice ,Mice ,Knockout ,T-Lymphocytes ,Regulatory ,Integrin beta 7 Blockade ,Regulatory T Cells ,Gut-Associated Lymphoid Tissue ,Integrin β7 Blockade ,Biochemistry and cell biology ,Clinical sciences - Abstract
Background & aimsIntegrin α4β7 mediates lymphocyte trafficking to the gut and gut-associated lymphoid tissues, a process critical for recruitment of effector lymphocytes from the circulation to the gut mucosa in inflammatory bowel disease (IBD) and murine models of intestinal inflammation. Antibody blockade of β7 integrins generally is efficacious in IBD; however, some patients fail to respond, and a few patients can experience exacerbations. This study examined the effects of loss of β7 integrin function in murine models of IBD.MethodsIn a mouse IBD model caused by lack of interleukin 10, a cytokine important in CD25hiFoxP3+ regulatory T cell (Treg) function, genetic deletion of β7 integrin or antibody blockade of α4β7-mucosal addressin cell adhesion molecule-1 interaction paradoxically exacerbated colitis.ResultsLoss of β7 impaired the capacity of Tregs homing to the gut and therefore suppress intestinal inflammation in an adoptive T-cell transfer model; however, the intrinsic suppressive function of β7-deficient Tregs remained intact, indicating that the β7 deficiency selectively impacts gut homing. Deletion of β7 integrin did not worsen colitis in an acute dextran sodium sulfate model in which Treg number and function were normal.ConclusionsIn Integrin subunit beta (Itgb)7-/-Il10-/- mice, loss of β7-dependent Treg homing to gut-associated lymphoid tissues combined with loss of intrinsic Treg function exacerbated intestinal inflammation. These results suggest that IBD patients with reduced CD25hiFoxP3+ Treg numbers or function or lack of interleukin 10 could be at risk for failure of α4β7 blocking therapy.
- Published
- 2020
11. Biomarkers of cavernous angioma with symptomatic hemorrhage
- Author
-
Lyne, Seán B, Girard, Romuald, Koskimäki, Janne, Zeineddine, Hussein A, Zhang, Dongdong, Cao, Ying, Li, Yan, Stadnik, Agnieszka, Moore, Thomas, Lightle, Rhonda, Shi, Changbin, Shenkar, Robert, Carrión-Penagos, Julián, Polster, Sean P, Romanos, Sharbel, Akers, Amy, Lopez-Ramirez, Miguel, Whitehead, Kevin J, Kahn, Mark L, Ginsberg, Mark H, Marchuk, Douglas A, and Awad, Issam A
- Subjects
Prevention ,Clinical Research ,Clinical Trials and Supportive Activities ,4.1 Discovery and preclinical testing of markers and technologies ,Detection ,screening and diagnosis ,4.2 Evaluation of markers and technologies ,Good Health and Well Being ,Adult ,Biomarkers ,Cohort Studies ,Female ,Hemangioma ,Cavernous ,Hemorrhage ,Humans ,Inflammation Mediators ,Longitudinal Studies ,Machine Learning ,Male ,Transcriptome ,Diagnostics ,Inflammation ,Stroke ,Vascular Biology - Abstract
BACKGROUNDCerebral cavernous angiomas (CAs) with a symptomatic hemorrhage (CASH) have a high risk of recurrent hemorrhage and serious morbidity.METHODSEighteen plasma molecules with mechanistic roles in CA pathobiology were investigated in 114 patients and 12 healthy subjects. The diagnostic biomarker of a CASH in the prior year was derived as that minimizing the Akaike information criterion and validated using machine learning, and was compared with the prognostic CASH biomarker predicting bleeding in the subsequent year. Biomarkers were longitudinally followed in a subset of cases. The biomarkers were queried in the lesional neurovascular unit (NVU) transcriptome and in plasma miRNAs from CASH and non-CASH patients.RESULTSThe diagnostic CASH biomarker included a weighted combination of soluble CD14 (sCD14), VEGF, C-reactive protein (CRP), and IL-10 distinguishing CASH patients with 76% sensitivity and 80% specificity (P = 0.0003). The prognostic CASH biomarker (sCD14, VEGF, IL-1β, and sROBO-4) was confirmed to predict a bleed in the subsequent year with 83% sensitivity and 93% specificity (P = 0.001). Genes associated with diagnostic and prognostic CASH biomarkers were differentially expressed in CASH lesional NVUs. Thirteen plasma miRNAs were differentially expressed between CASH and non-CASH patients.CONCLUSIONShared and unique biomarkers of recent symptomatic hemorrhage and of future bleeding in CA are mechanistically linked to lesional transcriptome and miRNA. The biomarkers may be applied for risk stratification in clinical trials and developed as a tool in clinical practice.FUNDINGNIH, William and Judith Davis Fund in Neurovascular Surgery Research, Be Brave for Life Foundation, Safadi Translational Fellowship, Pritzker School of Medicine, and Sigrid Jusélius Foundation.
- Published
- 2019
12. Rap1 binding and a lipid-dependent helix in talin F1 domain promote integrin activation in tandem
- Author
-
Gingras, Alexandre R, Lagarrigue, Frederic, Cuevas, Monica N, Valadez, Andrew J, Zorovich, Marcus, McLaughlin, Wilma, Lopez-Ramirez, Miguel Alejandro, Seban, Nicolas, Ley, Klaus, Kiosses, William B, and Ginsberg, Mark H
- Subjects
1.1 Normal biological development and functioning ,Underpinning research ,Animals ,Binding Sites ,CHO Cells ,Cricetinae ,Cricetulus ,Humans ,Integrins ,Lipids ,Mutation ,Protein Binding ,Protein Conformation ,Protein Domains ,Shelterin Complex ,Talin ,Telomere-Binding Proteins ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology - Abstract
Rap1 GTPases bind effectors, such as RIAM, to enable talin1 to induce integrin activation. In addition, Rap1 binds directly to the talin1 F0 domain (F0); however, this interaction makes a limited contribution to integrin activation in CHO cells or platelets. Here, we show that talin1 F1 domain (F1) contains a previously undetected Rap1-binding site of similar affinity to that in F0. A structure-guided point mutant (R118E) in F1, which blocks Rap1 binding, abolishes the capacity of Rap1 to potentiate talin1-induced integrin activation. The capacity of F1 to mediate Rap1-dependent integrin activation depends on a unique loop in F1 that has a propensity to form a helix upon binding to membrane lipids. Basic membrane-facing residues of this helix are critical, as charge-reversal mutations led to dramatic suppression of talin1-dependent activation. Thus, a novel Rap1-binding site and a transient lipid-dependent helix in F1 work in tandem to enable a direct Rap1-talin1 interaction to cause integrin activation.
- Published
- 2019
13. High-Affinity Bent β2-Integrin Molecules in Arresting Neutrophils Face Each Other through Binding to ICAMs In cis
- Author
-
Fan, Zhichao, Kiosses, William Bill, Sun, Hao, Orecchioni, Marco, Ghosheh, Yanal, Zajonc, Dirk M, Arnaout, M Amin, Gutierrez, Edgar, Groisman, Alex, Ginsberg, Mark H, and Ley, Klaus
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,5.1 Pharmaceuticals ,Development of treatments and therapeutic interventions ,CD18 Antigens ,Cell Adhesion Molecules ,Humans ,Neutrophils ,Protein Binding ,STORM ,TIRF ,human ,integrin ,integrin activation ,molecular modeling ,neutrophil ,superresolution ,Medical Physiology ,Biological sciences - Abstract
Leukocyte adhesion requires β2-integrin activation. Resting integrins exist in a bent-closed conformation-i.e., not extended (E-) and not high affinity (H-)-unable to bind ligand. Fully activated E+H+ integrin binds intercellular adhesion molecules (ICAMs) expressed on the opposing cell in trans. E-H- transitions to E+H+ through E+H- or through E-H+, which binds to ICAMs on the same cell in cis. Spatial patterning of activated integrins is thought to be required for effective arrest, but no high-resolution cell surface localization maps of activated integrins exist. Here, we developed Super-STORM by combining super-resolution microscopy with molecular modeling to precisely localize activated integrin molecules and identify the molecular patterns of activated integrins on primary human neutrophils. At the time of neutrophil arrest, E-H+ integrins face each other to form oriented (non-random) nanoclusters. To address the mechanism causing this pattern, we blocked integrin binding to ICAMs in cis, which significantly relieved the face-to-face orientation.
- Published
- 2019
14. Integrin Activation Controls Regulatory T Cell–Mediated Peripheral Tolerance
- Author
-
Klann, Jane E, Kim, Stephanie H, Remedios, Kelly A, He, Zhaoren, Metz, Patrick J, Lopez, Justine, Tysl, Tiffani, Olvera, Jocelyn G, Ablack, Jailal N, Cantor, Joseph M, Boland, Brigid S, Yeo, Gene, Zheng, Ye, Lu, Li-Fan, Bui, Jack D, Ginsberg, Mark H, Petrich, Brian G, and Chang, John T
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Prevention ,Autoimmune Disease ,Aetiology ,1.1 Normal biological development and functioning ,Underpinning research ,2.1 Biological and endogenous factors ,Inflammatory and immune system ,Animals ,Autoimmunity ,Inflammation ,Integrins ,Mice ,Peripheral Tolerance ,T-Lymphocytes ,Regulatory ,Talin ,Transcriptome ,Immunology ,Biochemistry and cell biology - 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 β 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 β 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 α4β1 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.
- Published
- 2018
15. Transmission of integrin β7 transmembrane domain topology enables gut lymphoid tissue development
- Author
-
Sun, Hao, Lagarrigue, Frederic, Gingras, Alexandre R, Fan, Zhichao, Ley, Klaus, and Ginsberg, Mark H
- Subjects
2.1 Biological and endogenous factors ,Aetiology ,1.1 Normal biological development and functioning ,Underpinning research ,Animals ,Cell Adhesion ,Female ,Gastrointestinal Tract ,HEK293 Cells ,Humans ,Integrin beta Chains ,Integrins ,Jurkat Cells ,Leukocyte Rolling ,Lymphocyte Activation ,Lymphocytes ,Lymphoid Tissue ,Male ,Mice ,Inbred C57BL ,Mice ,Transgenic ,Mutation ,Protein Binding ,Protein Interaction Domains and Motifs ,Signal Transduction ,Structure-Activity Relationship ,Talin ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology - Abstract
Integrin activation regulates adhesion, extracellular matrix assembly, and cell migration, thereby playing an indispensable role in development and in many pathological processes. A proline mutation in the central integrin β3 transmembrane domain (TMD) creates a flexible kink that uncouples the topology of the inner half of the TMD from the outer half. In this study, using leukocyte integrin α4β7, which enables development of gut-associated lymphoid tissue (GALT), we examined the biological effect of such a proline mutation and report that it impairs agonist-induced talin-mediated activation of integrin α4β7, thereby inhibiting rolling lymphocyte arrest, a key step in transmigration. Furthermore, the α4β7(L721P) mutation blocks lymphocyte homing to and development of the GALT. These studies show that impairing the ability of an integrin β TMD to transmit talin-induced TMD topology inhibits agonist-induced physiological integrin activation and biological function in development.
- Published
- 2018
16. Thrombospondin1 (TSP1) replacement prevents cerebral cavernous malformations
- Author
-
Lopez-Ramirez, Miguel Alejandro, Fonseca, Gregory, Zeineddine, Hussein A, Girard, Romuald, Moore, Thomas, Pham, Angela, Cao, Ying, Shenkar, Robert, de Kreuk, Bart-Jan, Lagarrigue, Frederic, Lawler, Jack, Glass, Christopher K, Awad, Issam A, and Ginsberg, Mark H
- Subjects
Rare Diseases ,Brain Disorders ,Neurosciences ,Good Health and Well Being ,Animals ,Cells ,Cultured ,Endothelial Cells ,Gene Expression Profiling ,Genetic Therapy ,HEK293 Cells ,Hemangioma ,Cavernous ,Central Nervous System ,Humans ,KRIT1 Protein ,Kruppel-Like Factor 4 ,Kruppel-Like Transcription Factors ,Mice ,Inbred C57BL ,Mice ,Knockout ,Mice ,Transgenic ,RNA Interference ,Thrombospondin 1 ,Medical and Health Sciences ,Immunology - Abstract
KRIT1 mutations are the most common cause of cerebral cavernous malformation (CCM). Acute Krit1 gene inactivation in mouse brain microvascular endothelial cells (BMECs) changes expression of multiple genes involved in vascular development. These changes include suppression of Thbs1, which encodes thrombospondin1 (TSP1) and has been ascribed to KLF2- and KLF4-mediated repression of Thbs1 In vitro reconstitution of TSP1 with either full-length TSP1 or 3TSR, an anti-angiogenic TSP1 fragment, suppresses heightened vascular endothelial growth factor signaling and preserves BMEC tight junctions. Furthermore, administration of 3TSR prevents the development of lesions in a mouse model of CCM1 (Krit1ECKO ) as judged by histology and quantitative micro-computed tomography. Conversely, reduced TSP1 expression contributes to the pathogenesis of CCM, because inactivation of one or two copies of Thbs1 exacerbated CCM formation. Thus, loss of Krit1 function disables an angiogenic checkpoint to enable CCM formation. These results suggest that 3TSR, or other angiogenesis inhibitors, can be repurposed for TSP1 replacement therapy for CCMs.
- Published
- 2017
17. Tissue Factor Prothrombotic Activity Is Regulated by Integrin-arf6 Trafficking
- Author
-
Rothmeier, Andrea S, Marchese, Patrizia, Langer, Florian, Kamikubo, Yuichi, Schaffner, Florence, Cantor, Joseph, Ginsberg, Mark H, Ruggeri, Zaverio M, and Ruf, Wolfram
- Subjects
Biomedical and Clinical Sciences ,Cardiovascular Medicine and Haematology ,Clinical Sciences ,Hematology ,Biotechnology ,2.1 Biological and endogenous factors ,Aetiology ,ADP-Ribosylation Factor 6 ,ADP-Ribosylation Factors ,Adenosine Triphosphate ,Animals ,Blood Coagulation ,Cell Line ,Tumor ,Cell-Derived Microparticles ,Factor VIIa ,Gene Knock-In Techniques ,Genotype ,Humans ,Integrin alpha4 ,Integrin alpha4beta1 ,Macrophages ,Mice ,Inbred C57BL ,Mice ,Transgenic ,Phenotype ,Phospholipids ,Protein Transport ,Receptors ,Purinergic P2X7 ,Signal Transduction ,Thromboplastin ,Thrombosis ,Transfection ,dynamins ,extracellular vesicles ,fibrin ,macrophages ,proteome ,Cardiorespiratory Medicine and Haematology ,Cardiovascular System & Hematology ,Cardiovascular medicine and haematology ,Clinical sciences - Abstract
ObjectiveCoagulation initiation by tissue factor (TF) is regulated by cellular inhibitors, cell surface availability of procoagulant phosphatidylserine, and thiol-disulfide exchange. How these mechanisms contribute to keeping TF in a noncoagulant state and to generating prothrombotic TF remain incompletely understood.Approach and resultsHere, we study the activation of TF in primary macrophages by a combination of pharmacological, genetic, and biochemical approaches. We demonstrate that primed macrophages effectively control TF cell surface activity by receptor internalization. After cell injury, ATP signals through the purinergic receptor P2rx7 induce release of TF+ microvesicles. TF cell surface availability for release onto microvesicles is regulated by the GTPase arf6 associated with integrin α4β1. Furthermore, microvesicles proteome analysis identifies activation of Gαi2 as a participating factor in the release of microvesicles with prothrombotic activity in flowing blood. ATP not only prevents TF and phosphatidylserine internalization but also induces TF conversion to a conformation with high affinity for its ligand, coagulation factor VII. Although inhibition of dynamin-dependent internalization also exposes outer membrane procoagulant phosphatidylserine, the resulting TF+ microvesicles distinctly lack protein disulfide isomerase and high affinity TF and fail to produce fibrin strands typical for microvesicles generated by thrombo-inflammatory P2rx7 activation.ConclusionsThese data show that procoagulant phospholipid exposure is not sufficient and that TF affinity maturation is required to generate prothrombotic microvesicles from a variety of cell types. These findings are significant for understanding TF-initiated thrombosis and should be considered in designing functional microvesicles-based diagnostic approaches.
- Published
- 2017
18. Talin Plays a Critical Role in the Maintenance of the Regulatory T Cell Pool
- Author
-
Klann, Jane E, Remedios, Kelly A, Kim, Stephanie H, Metz, Patrick J, Lopez, Justine, Mack, Lauren A, Zheng, Ye, Ginsberg, Mark H, Petrich, Brian G, and Chang, John T
- Subjects
Underpinning research ,2.1 Biological and endogenous factors ,1.1 Normal biological development and functioning ,Aetiology ,Inflammatory and immune system ,Animals ,Apoptosis ,Dendritic Cells ,Genes ,bcl-2 ,Homeostasis ,Interleukin-2 ,Interleukin-2 Receptor alpha Subunit ,Lymphocyte Activation ,Lymphocyte Function-Associated Antigen-1 ,Mice ,Myeloid Cell Leukemia Sequence 1 Protein ,Signal Transduction ,T-Lymphocytes ,Regulatory ,Talin ,Immunology - Abstract
Talin, a cytoskeletal protein essential in mediating integrin activation, has been previously shown to be involved in the regulation of T cell proliferation and function. In this study, we describe a role for talin in maintaining the homeostasis and survival of the regulatory T (Treg) cell pool. T cell-specific deletion of talin in Tln1fl/flCd4Cre mice resulted in spontaneous lymphocyte activation, primarily due to numerical and functional deficiencies of Treg cells in the periphery. Peripheral talin-deficient Treg cells were unable to maintain high expression of IL-2Rα, resulting in impaired IL-2 signaling and ultimately leading to increased apoptosis through downregulation of prosurvival proteins Bcl-2 and Mcl-1. The requirement for talin in maintaining high IL-2Rα expression by Treg cells was due, in part, to integrin LFA-1-mediated interactions between Treg cells and dendritic cells. Collectively, our data suggest a critical role for talin in Treg cell-mediated maintenance of immune homeostasis.
- Published
- 2017
19. Epigallocatechin gallate has pleiotropic effects on transmembrane signaling by altering the embedding of transmembrane domains
- Author
-
Ye, Feng, Yang, Chansik, Kim, Jiyoon, MacNevin, Christopher J, Hahn, Klaus M, Park, Dongeun, Ginsberg, Mark H, and Kim, Chungho
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Complementary and Integrative Health ,Nutrition ,Amino Acid Substitution ,Animals ,Antioxidants ,CHO Cells ,Catechin ,Cricetulus ,Dietary Supplements ,Dimerization ,ErbB Receptors ,Humans ,Integrin alpha2 ,Integrin beta3 ,Ligands ,Lipid Bilayers ,Models ,Molecular ,Mutation ,Peptide Fragments ,Platelet Glycoprotein GPIIb-IIIa Complex ,Protein Interaction Domains and Motifs ,Recombinant Fusion Proteins ,Signal Transduction ,Talin ,epidermal growth factor receptor ,integrin ,polyphenol ,talin ,transmembrane domain ,Chemical Sciences ,Medical and Health Sciences ,Biochemistry & Molecular Biology ,Biological sciences ,Biomedical and clinical sciences ,Chemical sciences - Abstract
Epigallocatechin gallate (EGCG) is the principal bioactive ingredient in green tea and has been reported to have many health benefits. EGCG influences multiple signal transduction pathways related to human diseases, including redox, inflammation, cell cycle, and cell adhesion pathways. However, the molecular mechanisms of these varying effects are unclear, limiting further development and utilization of EGCG as a pharmaceutical compound. Here, we examined the effect of EGCG on two representative transmembrane signaling receptors, integrinαIIbβ3 and epidermal growth factor receptor (EGFR). We report that EGCG inhibits talin-induced integrin αIIbβ3 activation, but it activates αIIbβ3 in the absence of talin both in a purified system and in cells. This apparent paradox was explained by the fact that the activation state of αIIbβ3 is tightly regulated by the topology of β3 transmembrane domain (TMD); increases or decreases in TMD embedding can activate integrins. Talin increases the embedding of integrin β3 TMD, resulting in integrin activation, whereas we observed here that EGCG decreases the embedding, thus opposing talin-induced integrin activation. In the absence of talin, EGCG decreases the TMD embedding, which can also disrupt the integrin α-β TMD interaction, leading to integrin activation. EGCG exhibited similar paradoxical behavior in EGFR signaling. EGCG alters the topology of EGFR TMD and activates the receptor in the absence of EGF, but inhibits EGF-induced EGFR activation. Thus, this widely ingested polyphenol exhibits pleiotropic effects on transmembrane signaling by modifying the topology of TMDs.
- Published
- 2017
20. Cutting Edge: Loss of T Cell RIAM Precludes Conjugate Formation with APC and Prevents Immune-Mediated Diabetes.
- Author
-
Lagarrigue, Frederic, Gertler, Frank B, Ginsberg, Mark H, and Cantor, Joseph M
- Subjects
Diabetes ,Prevention ,2.1 Biological and endogenous factors ,Aetiology ,Adaptor Proteins ,Signal Transducing ,Adoptive Transfer ,Animals ,Antigen-Presenting Cells ,Cell Proliferation ,Cells ,Cultured ,Cytotoxicity ,Immunologic ,Diabetes Mellitus ,Type 1 ,Humans ,Immunological Synapses ,Lymphocyte Function-Associated Antigen-1 ,Membrane Proteins ,Mice ,Mice ,129 Strain ,Mice ,Inbred C57BL ,Mice ,Knockout ,T-Lymphocytes ,Talin ,Immunology - 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.
- Published
- 2017
21. CD98 regulates vascular smooth muscle cell proliferation in atherosclerosis
- Author
-
Baumer, Yvonne, McCurdy, Sara, Alcala, Martin, Mehta, Nehal, Lee, Bog-Hieu, Ginsberg, Mark H, and Boisvert, William A
- Subjects
Medical Physiology ,Biomedical and Clinical Sciences ,Heart Disease ,Heart Disease - Coronary Heart Disease ,Atherosclerosis ,Cardiovascular ,Aging ,Animals ,Apoptosis ,Cell Movement ,Cell Proliferation ,Cells ,Cultured ,Disease Models ,Animal ,Elastin ,Fusion Regulatory Protein 1 ,Heavy Chain ,Genetic Predisposition to Disease ,Male ,Mice ,Inbred C57BL ,Mice ,Knockout ,Muscle ,Smooth ,Vascular ,Myocytes ,Smooth Muscle ,Necrosis ,Neointima ,Phenotype ,Plaque ,Atherosclerotic ,Receptors ,LDL ,Rupture ,Spontaneous ,Time Factors ,Vascular smooth muscle cell ,CD98 ,Cell proliferation ,Cardiorespiratory Medicine and Haematology ,Clinical Sciences ,Cardiovascular System & Hematology ,Cardiovascular medicine and haematology ,Clinical sciences - Abstract
Background and aimsVascular smooth muscle cells (VSMC) migrate and proliferate to form a stabilizing fibrous cap that encapsulates atherosclerotic plaques. CD98 is a transmembrane protein made of two subunits, CD98 heavy chain (CD98hc) and one of six light chains, and is known to be involved in cell proliferation and survival. Because the influence of CD98hc on atherosclerosis development is unknown, our aim was to determine if CD98hc expressed on VSMC plays a role in shaping the morphology of atherosclerotic plaques by regulating VSMC function.MethodsIn addition to determining the role of CD98hc in VSMC proliferation and apoptosis, we utilized mice with SMC-specific deletion of CD98hc (CD98hcfl/flSM22αCre+) to determine the effects of CD98hc deficiency on VSMC function in atherosclerotic plaque.ResultsAfter culturing for 5 days in vitro, CD98hc-/- VSMC displayed dramatically reduced cell counts, reduced proliferation, as well as reduced migration compared to control VSMC. Analysis of aortic VSCM after 8 weeks of HFD showed a reduction in CD98hc-/- VSMC proliferation as well as increased apoptosis compared to controls. A long-term atherosclerosis study using SMC-CD98hc-/-/ldlr-/- mice was performed. Although total plaque area was unchanged, CD98hc-/- mice showed reduced presence of VSMC within the plaque (2.1 ± 0.4% vs. 4.3 ± 0.4% SM22α-positive area per plaque area, p
- Published
- 2017
22. Structural Basis of Dimeric Rasip1 RA Domain Recognition of the Ras Subfamily of GTP-Binding Proteins
- Author
-
Gingras, Alexandre R, Puzon-McLaughlin, Wilma, Bobkov, Andrey A, and Ginsberg, Mark H
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,1.1 Normal biological development and functioning ,Underpinning research ,Binding Sites ,Dimerization ,Humans ,Intracellular Signaling Peptides and Proteins ,Models ,Molecular ,Protein Binding ,Protein Domains ,Protein Structure ,Secondary ,rap1 GTP-Binding Proteins ,ras Proteins ,GTP-binding proteins ,Radil ,Rap ,Ras ,Ras-association (RA) domain ,Ras-interacting protein 1 ,crystal structure ,dimer ,heart of glass ,rain ,Chemical Sciences ,Information and Computing Sciences ,Biophysics ,Biological sciences ,Chemical sciences - Abstract
Ras-interacting protein 1 (Rasip1) is an endothelial-specific Rap1 and Ras effector, important for vascular development and angiogenesis. Here, we report the crystal structure of the Rasip1 RA domain (RRA) alone, revealing the basis of dimerization, and in complex with Rap1 at 2.8 Å resolution. In contrast to most RA domains, RRA formed a dimer that can bind two Rap1 (KD = 0.9 μM) or Ras (KD = 2.2 μM) molecules. We solved the Rap1-RRA complex and found that Rasip1 binds Rap1 in the Switch I region, and Rap1 binding induces few conformation changes to Rasip1 stabilizing a β strand and an unstructured loop. Our data explain how Rasip1 can act as a Rap1 and Ras effector and show that Rasip1 defines a subgroup of dimeric RA domains that could mediate cooperative binding to membrane-associated Ras superfamily members.
- Published
- 2016
23. Kindlin-3 recruitment to the plasma membrane precedes high-affinity β2-integrin and neutrophil arrest from rolling
- Author
-
Wen, Lai, Marki, Alex, Roy, Payel, McArdle, Sara, Sun, Hao, Fan, Zhichao, Gingras, Alexandre R., Ginsberg, Mark H., and Ley, Klaus
- Published
- 2021
- Full Text
- View/download PDF
24. Enhanced Integrin α4β1–Mediated Adhesion Contributes to a Mobilization Defect of Endothelial Progenitor Cells in Diabetes
- Author
-
Abplanalp, Wesley T, Conklin, Daniel J, Cantor, Joseph M, Ginsberg, Mark H, Wysoczynski, Marcin, Bhatnagar, Aruni, and O’Toole, Timothy E
- Subjects
Biomedical and Clinical Sciences ,Diabetes ,Stem Cell Research - Nonembryonic - Human ,Stem Cell Research ,Stem Cell Research - Nonembryonic - Non-Human ,Regenerative Medicine ,Aetiology ,2.1 Biological and endogenous factors ,Metabolic and endocrine ,Animals ,Cell Adhesion ,Cells ,Cultured ,Cyclic AMP-Dependent Protein Kinases ,Diabetes Mellitus ,Experimental ,Endothelial Progenitor Cells ,Hindlimb ,Integrin alpha4beta1 ,Ischemia ,Male ,Mice ,Mutation ,Phosphorylation ,Signal Transduction ,Medical and Health Sciences ,Endocrinology & Metabolism ,Biomedical and clinical sciences - 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 1β (PRKAR1β), 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 PRKAR1β, 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.
- Published
- 2016
25. CD98-Mediated Adhesive Signaling Enables the Establishment and Propagation of Acute Myelogenous Leukemia
- Author
-
Bajaj, Jeevisha, Konuma, Takaaki, Lytle, Nikki K, Kwon, Hyog Young, Ablack, Jailal N, Cantor, Joseph M, Rizzieri, David, Chuah, Charles, Oehler, Vivian G, Broome, Elizabeth H, Ball, Edward D, van der Horst, Edward H, Ginsberg, Mark H, and Reya, Tannishtha
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,Hematology ,Pediatric ,Childhood Leukemia ,Pediatric Cancer ,Pediatric Research Initiative ,Rare Diseases ,Cancer ,5.1 Pharmaceuticals ,Development of treatments and therapeutic interventions ,Aetiology ,2.1 Biological and endogenous factors ,Animals ,Antibodies ,Cell Adhesion ,Cell Line ,Tumor ,Cell Proliferation ,Fusion Regulatory Protein-1 ,Gene Knockout Techniques ,Humans ,Leukemia ,Myeloid ,Acute ,Mice ,Neoplasm Transplantation ,Neoplastic Stem Cells ,CD98 ,SLC3A2 ,acute myelogenous leukemia ,adhesion ,cancer ,cancer stem cell ,imaging ,integrin ,leukemia ,microenvironment ,Neurosciences ,Oncology and Carcinogenesis ,Oncology & Carcinogenesis ,Biochemistry and cell biology ,Oncology and carcinogenesis - 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.
- Published
- 2016
26. A Conserved Ectodomain-Transmembrane Domain Linker Motif Tunes the Allosteric Regulation of Cell Surface Receptors*
- Author
-
Schmidt, Thomas, Ye, Feng, Situ, Alan J, An, Woojin, Ginsberg, Mark H, and Ulmer, Tobias S
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Mental Health ,Allosteric Regulation ,Animals ,CHO Cells ,Cricetinae ,Cricetulus ,Cytokine Receptor Common beta Subunit ,Humans ,Integrin beta Chains ,Protein Domains ,Protein Structure ,Secondary ,allosteric regulation ,cytokine ,integrin ,isothermal titration calorimetry ,nuclear magnetic resonance ,protein dynamics ,receptor signaling ,transmembrane cell surface receptors ,Chemical Sciences ,Medical and Health Sciences ,Biochemistry & Molecular Biology ,Biological sciences ,Biomedical and clinical sciences ,Chemical sciences - Abstract
In many families of cell surface receptors, a single transmembrane (TM) α-helix separates ecto- and cytosolic domains. A defined coupling of ecto- and TM domains must be essential to allosteric receptor regulation but remains little understood. Here, we characterize the linker structure, dynamics, and resulting ecto-TM domain coupling of integrin αIIb in model constructs and relate it to other integrin α subunits by mutagenesis. Cellular integrin activation assays subsequently validate the findings in intact receptors. Our results indicate a flexible yet carefully tuned ecto-TM coupling that modulates the signaling threshold of integrin receptors. Interestingly, a proline at the N-terminal TM helix border, termed NBP, is critical to linker flexibility in integrins. NBP is further predicted in 21% of human single-pass TM proteins and validated in cytokine receptors by the TM domain structure of the cytokine receptor common subunit β and its P441A-substituted variant. Thus, NBP is a conserved uncoupling motif of the ecto-TM domain transition and the degree of ecto-TM domain coupling represents an important parameter in the allosteric regulation of diverse cell surface receptors.
- Published
- 2016
27. Amino Acid Transport Associated to Cluster of Differentiation 98 Heavy Chain (CD98hc) Is at the Cross-road of Oxidative Stress and Amino Acid Availability*
- Author
-
de la Ballina, Laura R, Cano-Crespo, Sara, González-Muñoz, Elena, Bial, Susanna, Estrach, Soline, Cailleteau, Laurence, Tissot, Floriane, Daniel, Hannelore, Zorzano, Antonio, Ginsberg, Mark H, Palacín, Manuel, and Féral, Chloé C
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,1.1 Normal biological development and functioning ,Underpinning research ,Amino Acid Transport System y+ ,Amino Acid Transport System y+L ,Amino Acids ,Animals ,Biological Transport ,Active ,Cell Differentiation ,Cell Line ,Cell Proliferation ,Cell Survival ,Fusion Regulatory Protein 1 ,Heavy Chain ,Fusion Regulatory Protein 1 ,Light Chains ,Gene Deletion ,Mice ,Mouse Embryonic Stem Cells ,Oxidative Stress ,Reactive Oxygen Species ,SLC3A2/CD98hc/4F2hc ,SLC7 family ,amino acid transport ,cell proliferation ,oxidative stress ,peptide transport ,stress response ,Chemical Sciences ,Medical and Health Sciences ,Biochemistry & Molecular Biology ,Biological sciences ,Biomedical and clinical sciences ,Chemical sciences - Abstract
CD98hc functions as an amino acid (AA) transporter (together with another subunit) and integrin signaling enhancer. It is overexpressed in highly proliferative cells in both physiological and pathological conditions. CD98hc deletion induces strong impairment of cell proliferation in vivo and in vitro Here, we investigate CD98hc-associated AA transport in cell survival and proliferation. By using chimeric versions of CD98hc, the two functions of the protein can be uncoupled. Although recovering the CD98hc AA transport capacity restores the in vivo and in vitro proliferation of CD98hc-null cells, reconstitution of the integrin signaling function of CD98hc is unable to restore in vitro proliferation of those cells. CD98hc-associated transporters (i.e. xCT, LAT1, and y(+)LAT2 in wild-type cells) are crucial to control reactive oxygen species and intracellular AA levels, thus sustaining cell survival and proliferation. Moreover, in CD98hc-null cells the deficiency of CD98hc/xCT cannot be compensated, leading to cell death by ferroptosis. Supplementation of culture media with β-mercaptoethanol rescues CD98hc-deficient cell survival. Under such conditions null cells show oxidative stress and intracellular AA imbalance and, consequently, limited proliferation. CD98hc-null cells also present reduced intracellular levels of branched-chain and aromatic amino acids (BCAAs and ARO AAs, respectively) and induced expression of peptide transporter 1 (PEPT1). Interestingly, external supply of dipeptides containing BCAAs and ARO AAs rescues cell proliferation and compensates for impaired uptake of CD98hc/LAT1 and CD98hc/y(+)LAT2. Our data establish CD98hc as a master protective gene at the cross-road of redox control and AA availability, making it a relevant therapeutic target in cancer.
- Published
- 2016
28. Heart of glass anchors Rasip1 at endothelial cell-cell junctions to support vascular integrity.
- Author
-
de Kreuk, Bart-Jan, Gingras, Alexandre R, Knight, James Dr, Liu, Jian J, Gingras, Anne-Claude, and Ginsberg, Mark H
- Subjects
Cell Line ,Intercellular Junctions ,Endothelial Cells ,Humans ,Intracellular Signaling Peptides and Proteins ,Membrane Proteins ,Protein Binding ,Heart of Glass ,Rap1 ,Rasip1 ,Rho Kinase ,cell biology ,developmental biology ,endothelial cells ,human ,stem cells ,Biochemistry and Cell Biology - Abstract
Heart of Glass (HEG1), a transmembrane receptor, and Rasip1, an endothelial-specific Rap1-binding protein, are both essential for cardiovascular development. Here we performed a proteomic screen for novel HEG1 interactors and report that HEG1 binds directly to Rasip1. Rasip1 localizes to forming endothelial cell (EC) cell-cell junctions and silencing HEG1 prevents this localization. Conversely, mitochondria-targeted HEG1 relocalizes Rasip1 to mitochondria in cells. The Rasip1-binding site in HEG1 contains a 9 residue sequence, deletion of which abrogates HEG1's ability to recruit Rasip1. HEG1 binds to a central region of Rasip1 and deletion of this domain eliminates Rasip1's ability to bind HEG1, to translocate to EC junctions, to inhibit ROCK activity, and to maintain EC junctional integrity. These studies establish that the binding of HEG1 to Rasip1 mediates Rap1-dependent recruitment of Rasip1 to and stabilization of EC cell-cell junctions.
- Published
- 2016
29. The Structure of a Full-length Membrane-embedded Integrin Bound to a Physiological Ligand*
- Author
-
Dai, Aguang, Ye, Feng, Taylor, Dianne W, Hu, Guiqing, Ginsberg, Mark H, and Taylor, Kenneth A
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Blood Platelets ,Fibrin ,Humans ,In Vitro Techniques ,Ligands ,Manganese ,Membrane Lipids ,Microscopy ,Electron ,Transmission ,Models ,Molecular ,Nanostructures ,Platelet Glycoprotein GPIIb-IIIa Complex ,Protein Binding ,Protein Conformation ,cell adhesion ,electron microscopy ,fibrin ,nanodisc ,platelet ,signal transduction ,αIIbβ3 integrin ,Chemical Sciences ,Medical and Health Sciences ,Biochemistry & Molecular Biology ,Biological sciences ,Biomedical and clinical sciences ,Chemical sciences - Abstract
Increased ligand binding to integrin ("activation") underpins many biological processes, such as leukocyte trafficking, cell migration, host-pathogen interaction, and hemostasis. Integrins exist in several conformations, ranging from compact and bent to extended and open. However, the exact conformation of membrane-embedded, full-length integrin bound to its physiological macromolecular ligand is still unclear. Integrin αIIbβ3, the most abundant integrin in platelets, has been a prototype for integrin activation studies. Using negative stain electron microscopy and nanodisc-embedding to provide a membrane-like environment, we visualized the conformation of full-length αIIbβ3 in both a Mn(2+)-activated, ligand-free state and a Mn(2+)-activated, fibrin-bound state. Activated but ligand-free integrins exist mainly in the compact conformation, whereas fibrin-bound αIIbβ3 predominantly exists in a fully extended, headpiece open conformation. Our results show that membrane-embedded, full-length integrin adopts an extended and open conformation when bound to its physiological macromolecular ligand.
- Published
- 2015
30. RLIP76 regulates Arf6-dependent cell spreading and migration by linking ARNO with activated R-Ras at recycling endosomes
- Author
-
Wurtzel, Jeremy GT, Lee, Seunghyung, Singhal, Sharad S, Awasthi, Sanjay, Ginsberg, Mark H, and Goldfinger, Lawrence E
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,ADP-Ribosylation Factor 6 ,ADP-Ribosylation Factors ,Animals ,Base Sequence ,Cell Movement ,Endosomes ,Female ,Fibroblasts ,GTPase-Activating Proteins ,Mice ,Inbred C57BL ,Mice ,Knockout ,Molecular Sequence Data ,Multiprotein Complexes ,Signal Transduction ,ras Proteins ,Ras ,Migration ,Spreading ,Recycling endosome ,Small GTPase ,Medicinal and Biomolecular Chemistry ,Medical Biochemistry and Metabolomics ,Biochemistry & Molecular Biology ,Biochemistry and cell biology ,Medicinal and biomolecular chemistry - Abstract
R-Ras small GTPase enhances cell spreading and motility via RalBP1/RLIP76, an R-Ras effector that links GTP-R-Ras to activation of Arf6 and Rac1 GTPases. Here, we report that RLIP76 performs these functions by binding cytohesin-2/ARNO, an Arf GTPase guanine exchange factor, and connecting it to R-Ras at recycling endosomes. RLIP76 formed a complex with R-Ras and ARNO by binding ARNO via its N-terminus (residues 1-180) and R-Ras via residues 180-192. This complex was present in Rab11-positive recycling endosomes and the presence of ARNO in recycling endosomes required RLIP76, and was not supported by RLIP76(Δ1-180) or RLIP76(Δ180-192). Spreading and migration required RLIP76(1-180), and RLIP76(Δ1-180) blocked ARNO recruitment to recycling endosomes, and spreading. Arf6 activation with an ArfGAP inhibitor overcame the spreading defects in RLIP76-depleted cells or cells expressing RLIP76(Δ1-180). Similarly, RLIP76(Δ1-180) or RLIP76(Δ180-192) suppressed Arf6 activation. Together these results demonstrate that RLIP76 acts as a scaffold at recycling endosomes by binding activated R-Ras, recruiting ARNO to activate Arf6, thereby contributing to cell spreading and migration.
- Published
- 2015
31. A RIAM/lamellipodin-talin-integrin complex forms the tip of sticky fingers that guide cell migration.
- Author
-
Lagarrigue, Frederic, Vikas Anekal, Praju, Lee, Ho-Sup, Bachir, Alexia I, Ablack, Jailal N, Horwitz, Alan F, and Ginsberg, Mark H
- Subjects
Cells ,Humans ,Adaptor Proteins ,Signal Transducing ,Carrier Proteins ,Talin ,Membrane Proteins ,Integrins ,Cell Movement ,Protein Binding ,Adaptor Proteins ,Signal Transducing - Abstract
The leading edge of migrating cells contains rapidly translocating activated integrins associated with growing actin filaments that form 'sticky fingers' to sense extracellular matrix and guide cell migration. Here we utilized indirect bimolecular fluorescence complementation to visualize a molecular complex containing a Mig-10/RIAM/lamellipodin (MRL) protein (Rap1-GTP-interacting adaptor molecule (RIAM) or lamellipodin), talin and activated integrins in living cells. This complex localizes at the tips of growing actin filaments in lamellipodial and filopodial protrusions, thus corresponding to the tips of the 'sticky fingers.' Formation of the complex requires talin to form a bridge between the MRL protein and the integrins. Moreover, disruption of the MRL protein-integrin-talin (MIT) complex markedly impairs cell protrusion. These data reveal the molecular basis of the formation of 'sticky fingers' at the leading edge of migrating cells and show that an MIT complex drives these protrusions.
- Published
- 2015
32. An Isoform-Specific Myristylation Switch Targets Type II PKA Holoenzymes to Membranes.
- Author
-
Zhang, Ping, Ye, Feng, Bastidas, Adam C, Kornev, Alexandr P, Wu, Jian, Ginsberg, Mark H, and Taylor, Susan S
- Subjects
Humans ,Holoenzymes ,Myristic Acid ,Protein Isoforms ,Crystallography ,X-Ray ,Catalytic Domain ,Protein Structure ,Secondary ,Protein Binding ,Models ,Molecular ,Cyclic AMP-Dependent Protein Kinase RIalpha Subunit ,Cyclic AMP-Dependent Protein Kinase RIIbeta Subunit ,Molecular Docking Simulation ,Aetiology ,1.1 Normal biological development and functioning ,Underpinning research ,2.1 Biological and endogenous factors ,Chemical Sciences ,Biological Sciences ,Information and Computing Sciences ,Biophysics - Abstract
Cyclic AMP-dependent protein kinase (PKA) is regulated in part by N-terminal myristylation of its catalytic (C) subunit. Structural information about the role of myristylation in membrane targeting of PKA has been limited. In mammalian cells there are four functionally non-redundant PKA regulatory subunits (RIα, RIβ, RIIα, and RIIβ). PKA is assembled as an inactive R2C2 holoenzyme in cells. To explore the role of N-myristylation in membrane targeting of PKA holoenzymes, we solved crystal structures of RIα:myrC and RIIβ2:myrC2, and showed that the N-terminal myristylation site in the myrC serves as a flexible "switch" that can potentially be mobilized for membrane anchoring of RII, but not RI, holoenzymes. Furthermore, we synthesized nanodiscs and showed by electron microscopy that membrane targeting through the myristic acid is specific for the RII holoenzyme. This membrane-anchoring myristylation switch is independent of A Kinase Anchoring Proteins (AKAPs) that target PKA to membranes by other mechanisms.
- Published
- 2015
33. Antitumor activity of an anti‐CD98 antibody
- Author
-
Hayes, Gregory M, Chinn, Lawrence, Cantor, Joseph M, Cairns, Belinda, Levashova, Zoia, Tran, Hoang, Velilla, Timothy, Duey, Dana, Lippincott, John, Zachwieja, Joseph, Ginsberg, Mark H, and van der Horst, Edward H
- Subjects
Orphan Drug ,Lung ,Cancer ,Biotechnology ,Rare Diseases ,Hematology ,Lung Cancer ,Amino Acids ,Animals ,Antibodies ,Monoclonal ,Antibodies ,Monoclonal ,Humanized ,Antibody-Dependent Cell Cytotoxicity ,Antineoplastic Agents ,Biological Transport ,Caspases ,Cell Line ,Tumor ,Cell Membrane Permeability ,Complement System Proteins ,Cytotoxicity ,Immunologic ,Disease Models ,Animal ,Drug Evaluation ,Preclinical ,Female ,Fusion Regulatory Protein-1 ,Humans ,Lysosomes ,Mice ,Models ,Biological ,Protein Binding ,Tumor Burden ,Xenograft Model Antitumor Assays ,phenotypic screening ,anti-CD98 monoclonal antibody ,multiple mechanism of action ,acute myeloid leukemia ,non-small cell lung cancer ,Oncology and Carcinogenesis ,Oncology & Carcinogenesis - 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.
- Published
- 2015
34. Blocking neutrophil integrin activation prevents ischemia–reperfusion injury
- Author
-
Yago, Tadayuki, Petrich, Brian G, Zhang, Nan, Liu, Zhenghui, Shao, Bojing, Ginsberg, Mark H, and McEver, Rodger P
- Subjects
Kidney Disease ,Aetiology ,2.1 Biological and endogenous factors ,Animals ,Blotting ,Western ,CD18 Antigens ,Cell Movement ,DNA Primers ,Immunoprecipitation ,Kidney Diseases ,Leukocyte Rolling ,Mice ,Mice ,Transgenic ,Mutation ,Missense ,Neutrophils ,Reperfusion Injury ,Talin ,Medical and Health Sciences ,Immunology - Abstract
Neutrophil recruitment, mediated by β2 integrins, combats pyogenic infections but also plays a key role in ischemia-reperfusion injury and other inflammatory disorders. Talin induces allosteric rearrangements in integrins that increase affinity for ligands (activation). Talin also links integrins to actin and other proteins that enable formation of adhesions. Structural studies have identified a talin1 mutant (L325R) that perturbs activation without impairing talin's capacity to link integrins to actin and other proteins. Here, we found that mice engineered to express only talin1(L325R) in myeloid cells were protected from renal ischemia-reperfusion injury. Dissection of neutrophil function in vitro and in vivo revealed that talin1(L325R) neutrophils had markedly impaired chemokine-induced, β2 integrin-mediated arrest, spreading, and migration. Surprisingly, talin1(L325R) neutrophils exhibited normal selectin-induced, β2 integrin-mediated slow rolling, in sharp contrast to the defective slow rolling of neutrophils lacking talin1 or expressing a talin1 mutant (W359A) that blocks talin interaction with integrins. These studies reveal the importance of talin-mediated activation of integrins for renal ischemia-reperfusion injury. They further show that neutrophil arrest requires talin recruitment to and activation of integrins. However, although neutrophil slow rolling requires talin recruitment to integrins, talin-mediated integrin activation is dispensable.
- Published
- 2015
35. Fine-tuning Tumor Immunity with Integrin Trans-regulation
- Author
-
Cantor, Joseph M, Rose, David M, Slepak, Marina, and Ginsberg, Mark H
- Subjects
Biomedical and Clinical Sciences ,Immunology ,Immunization ,Cancer ,Vaccine Related ,1.1 Normal biological development and functioning ,Aetiology ,Underpinning research ,2.1 Biological and endogenous factors ,Inflammatory and immune system ,Animals ,Chemotaxis ,Leukocyte ,Disease Models ,Animal ,Humans ,Integrin alpha4 ,Integrins ,Lymphocyte Function-Associated Antigen-1 ,Mice ,Mice ,Transgenic ,Mutation ,Neoplasms ,Protein Binding ,T-Lymphocyte Subsets ,Tumor Burden ,Oncology and Carcinogenesis ,Pharmacology and Pharmaceutical Sciences ,Oncology and carcinogenesis - Abstract
Inefficient T-cell homing to tissues limits adoptive T-cell immunotherapy of solid tumors. αLβ2 and α4β1 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 αLβ2-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 αLβ2 integrin function biases leukocyte emigration toward lymphocytes relative to myeloid cells and enhances tumor immunity.
- Published
- 2015
36. Caspase-1–mediated pathway promotes generation of thromboinflammatory microparticles
- Author
-
Rothmeier, Andrea S, Marchese, Patrizia, Petrich, Brian G, Furlan-Freguia, Christian, Ginsberg, Mark H, Ruggeri, Zaverio M, and Ruf, Wolfram
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,Actins ,Adenosine Triphosphate ,Animals ,Caspase 1 ,Cell-Derived Microparticles ,Extracellular Fluid ,Immunity ,Innate ,Inflammasomes ,Inflammation ,Macrophages ,Membrane Microdomains ,Mice ,Mice ,Inbred C57BL ,Pseudopodia ,Receptors ,Purinergic P2X7 ,Signal Transduction ,Thioredoxin-Disulfide Reductase ,Thioredoxins ,Thrombosis ,Medical and Health Sciences ,Immunology ,Biological sciences ,Biomedical and clinical sciences ,Health sciences - Abstract
Extracellular ATP is a signal of tissue damage and induces macrophage responses that amplify inflammation and coagulation. Here we demonstrate that ATP signaling through macrophage P2X7 receptors uncouples the thioredoxin (TRX)/TRX reductase (TRXR) system and activates the inflammasome through endosome-generated ROS. TRXR and inflammasome activity promoted filopodia formation, cellular release of reduced TRX, and generation of extracellular thiol pathway-dependent, procoagulant microparticles (MPs). Additionally, inflammasome-induced activation of an intracellular caspase-1/calpain cysteine protease cascade degraded filamin, thereby severing bonds between the cytoskeleton and tissue factor (TF), the cell surface receptor responsible for coagulation activation. This cascade enabled TF trafficking from rafts to filopodia and ultimately onto phosphatidylserine-positive, highly procoagulant MPs. Furthermore, caspase-1 specifically facilitated cell surface actin exposure, which was required for the final release of highly procoagulant MPs from filopodia. Together, the results of this study delineate a thromboinflammatory pathway and suggest that components of this pathway have potential as pharmacological targets to simultaneously attenuate inflammation and innate immune cell-induced thrombosis.
- Published
- 2015
37. Annular Anionic Lipids Stabilize the Integrin αIIbβ3 Transmembrane Complex*
- Author
-
Schmidt, Thomas, Suk, Jae-Eun, Ye, Feng, Situ, Alan J, Mazumder, Parichita, Ginsberg, Mark H, and Ulmer, Tobias S
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Amino Acid Sequence ,Humans ,Molecular Dynamics Simulation ,Molecular Sequence Data ,Nuclear Magnetic Resonance ,Biomolecular ,Phosphatidylcholines ,Phosphatidylserines ,Platelet Glycoprotein GPIIb-IIIa Complex ,Protein Stability ,Protein Structure ,Secondary ,Biophysics ,Integrin ,Membrane Lipid ,Membrane Protein ,Molecular Dynamics ,Nuclear Magnetic Resonance ,Protein-Lipid Interaction ,Chemical Sciences ,Medical and Health Sciences ,Biochemistry & Molecular Biology ,Biological sciences ,Biomedical and clinical sciences ,Chemical sciences - Abstract
Cationic membrane-proximal amino acids determine the topology of membrane proteins by interacting with anionic lipids that are restricted to the intracellular membrane leaflet. This mechanism implies that anionic lipids interfere with electrostatic interactions of membrane proteins. The integrin αIIbβ3 transmembrane (TM) complex is stabilized by a membrane-proximal αIIb(Arg(995))-β3(Asp(723)) interaction; here, we examine the influence of anionic lipids on this complex. Anionic lipids compete for αIIb(Arg(995)) contacts with β3(Asp(723)) but paradoxically do not diminish the contribution of αIIb(Arg(995))-β3(Asp(723)) to TM complex stability. Overall, anionic lipids in annular positions stabilize the αIIbβ3 TM complex by up to 0.50 ± 0.02 kcal/mol relative to zwitterionic lipids in a headgroup structure-dependent manner. Comparatively, integrin receptor activation requires TM complex destabilization of 1.5 ± 0.2 kcal/mol, revealing a sizeable influence of lipid composition on TM complex stability. We implicate changes in lipid headgroup accessibility to small molecules (physical membrane characteristics) and specific but dynamic protein-lipid contacts in this TM helix-helix stabilization. Thus, anionic lipids in ubiquitous annular positions can benefit the stability of membrane proteins while leaving membrane-proximal electrostatic interactions intact.
- Published
- 2015
38. Integrin activation
- Author
-
Ginsberg, Mark H
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Underpinning research ,1.1 Normal biological development and functioning ,Inflammatory and immune system ,Allosteric Regulation ,Cell Adhesion ,Humans ,Integrins ,Protein Binding ,Protein Structure ,Tertiary ,Talin ,Cell adhesion ,Integrin ,Nanodisc ,Signal transduction ,Transmembrane domain ,Biochemistry & Molecular Biology ,Biochemistry and cell biology - Abstract
Integrin-mediated cell adhesion is important for development, immune responses, hemostasis and wound healing. Integrins also function as signal transducing receptors that can control intracellular pathways that regulate cell survival, proliferation, and cell fate. Conversely, cells can modulate the affinity of integrins for their ligands a process operationally defined as integrin activation. Analysis of activation of integrins has now provided a detailed molecular understanding of this unique form of "inside-out" signal transduction and revealed new paradigms of how transmembrane domains (TMD) can transmit long range allosteric changes in transmembrane proteins. Here, we will review how talin and mediates integrin activation and how the integrin TMD can transmit these inside out signals.
- Published
- 2014
39. CD98hc (SLC3A2) Loss Protects Against Ras-Driven Tumorigenesis by Modulating Integrin-Mediated Mechanotransduction
- Author
-
Estrach, Soline, Lee, Sin-Ae, Boulter, Etienne, Pisano, Sabrina, Errante, Aurélia, Tissot, Floriane S, Cailleteau, Laurence, Pons, Catherine, Ginsberg, Mark H, and Féral, Chloé C
- Subjects
Cancer ,Aetiology ,2.1 Biological and endogenous factors ,Acyltransferases ,Adaptor Proteins ,Signal Transducing ,Animals ,Carcinogenesis ,Cell Cycle Proteins ,Cell Proliferation ,Cells ,Cultured ,Extracellular Matrix ,Fusion Regulatory Protein 1 ,Heavy Chain ,Integrins ,Mechanotransduction ,Cellular ,Mice ,Phosphoproteins ,Signal Transduction ,Skin ,Transcription Factors ,Tumor Microenvironment ,YAP-Signaling Proteins ,ras Proteins ,rho-Associated Kinases ,Oncology and Carcinogenesis ,Oncology & Carcinogenesis - Abstract
CD98hc (SLC3A2) is the heavy chain component of the dimeric transmembrane glycoprotein CD98, which comprises the large neutral amino acid transporter LAT1 (SLC7A5) in cells. Overexpression of CD98hc occurs widely in cancer cells and is associated with poor prognosis clinically, but its exact contributions to tumorigenesis are uncertain. In this study, we showed that genetic deficiency of CD98hc protects against Ras-driven skin carcinogenesis. Deleting CD98hc after tumor induction was also sufficient to cause regression of existing tumors. Investigations into the basis for these effects defined two new functions of CD98hc that contribute to epithelial cancer beyond an intrinsic effect of CD98hc on tumor cell proliferation. First, CD98hc increased the stiffness of the tumor microenvironment. Second, CD98hc amplified the capacity of cells to respond to matrix rigidity, an essential factor in tumor development. Mechanistically, CD98hc mediated this stiffness sensing by increasing Rho kinase (ROCK) activity, resulting in increased transcription mediated by YAP/TAZ, a nuclear relay for mechanical signals. Our results suggest that CD98hc contributes to carcinogenesis by amplifying a positive feedback loop, which increases both extracellular matrix stiffness and resulting cellular responses. This work supports a rationale to explore the use of CD98hc inhibitors as cancer therapeutics.
- Published
- 2014
40. Cerebral cavernous malformations form an anticoagulant vascular domain in humans and mice
- Author
-
Lopez-Ramirez, Miguel Alejandro, Pham, Angela, Girard, Romuald, Wyseure, Tine, Hale, Preston, Yamashita, Atsuki, Koskimäki, Janne, Polster, Sean, Saadat, Laleh, Romero, Ignacio A., Esmon, Charles T., Lagarrigue, Frederic, Awad, Issam A., Mosnier, Laurent O., and Ginsberg, Mark H.
- Published
- 2019
- Full Text
- View/download PDF
41. High mutation detection rates in cerebral cavernous malformation upon stringent inclusion criteria: one‐third of probands are minors
- Author
-
Spiegler, Stefanie, Najm, Juliane, Liu, Jian, Gkalympoudis, Stephanie, Schröder, Winnie, Borck, Guntram, Brockmann, Knut, Elbracht, Miriam, Fauth, Christine, Ferbert, Andreas, Freudenberg, Leonie, Grasshoff, Ute, Hellenbroich, Yorck, Henn, Wolfram, Hoffjan, Sabine, Hüning, Irina, Korenke, G Christoph, Kroisel, Peter M, Kunstmann, Erdmute, Mair, Martina, Munk-Schulenburg, Susanne, Nikoubashman, Omid, Pauli, Silke, Rudnik-Schöneborn, Sabine, Sudholt, Irene, Sure, Ulrich, Tinschert, Sigrid, Wiednig, Michaela, Zoll, Barbara, Ginsberg, Mark H, and Felbor, Ute
- Subjects
Stroke ,Genetic Testing ,Neurosciences ,Rare Diseases ,Brain Disorders ,Clinical Research ,Genetics ,2.1 Biological and endogenous factors ,Aetiology ,Age at disease onset ,CCM1 ,CCM2 ,CCM3 ,cerebral cavernous malformation ,HEG1 ,mutation detection rate ,predictive testing ,Medicinal and Biomolecular Chemistry ,Clinical Sciences - Abstract
Cerebral cavernous malformations (CCM) are prevalent vascular malformations occurring in familial autosomal dominantly inherited or isolated forms. Once CCM are diagnosed by magnetic resonance imaging, the indication for genetic testing requires either a positive family history of cavernous lesions or clinical symptoms such as chronic headaches, epilepsy, neurological deficits, and hemorrhagic stroke or the occurrence of multiple lesions in an isolated case. Following these inclusion criteria, the mutation detection rates in a consecutive series of 105 probands were 87% for familial and 57% for isolated cases. Thirty-one novel mutations were identified with a slight shift towards proportionally more CCM3 mutations carriers than previously published (CCM1: 60%, CCM2: 18%, CCM3: 22%). In-frame deletions and exonic missense variants requiring functional analyses to establish their pathogenicity were rare: An in-frame deletion within the C-terminal FERM domain of CCM1 resulted in decreased protein expression and impaired binding to the transmembrane protein heart of glass (HEG1). Notably, 20% of index cases carrying a CCM mutation were below age 10 and 33% below age 18 when referred for genetic testing. Since fulminant disease courses during the first years of life were observed in CCM1 and CCM3 mutation carriers, predictive testing of minor siblings became an issue.
- Published
- 2014
42. SnapShot: Talin and the Modular Nature of the Integrin Adhesome
- Author
-
Ye, Feng, Lagarrigue, Frederic, and Ginsberg, Mark H
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Amino Acid Sequence ,Animals ,Cell Adhesion ,Integrins ,Molecular Sequence Data ,Talin ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
Integrin αβ transmembrane heterodimers play central roles in metazoan development and physiology by mediating adhesion and by transmitting forces and biochemical signals across the plasma membrane. In this SnapShot, we present a simplified "modular" view of the integrin adhesome, centered on the talin-integrin interaction, and provide examples of how this view can help to unravel the adhesome's remarkable functional diversity and plasticity.
- Published
- 2014
43. Platelet Function
- Author
-
Ginsberg, Mark H., primary, Plow, Edward F., additional, and Marguerie, Gérard A., additional
- Published
- 2020
- Full Text
- View/download PDF
44. The Mechanism of Kindlin-Mediated Activation of Integrin αIIbβ3
- Author
-
Ye, Feng, Petrich, Brian G, Anekal, Praju, Lefort, Craig T, Kasirer-Friede, Ana, Shattil, Sanford J, Ruppert, Raphael, Moser, Markus, Fässler, Reinhard, and Ginsberg, Mark H
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Hematology ,5.1 Pharmaceuticals ,Development of treatments and therapeutic interventions ,Animals ,Blood Platelets ,Cytoskeletal Proteins ,HEK293 Cells ,Humans ,Ligands ,Membrane Proteins ,Mice ,Neoplasm Proteins ,Platelet Glycoprotein GPIIb-IIIa Complex ,Protein Structure ,Tertiary ,Recombinant Proteins ,Medical and Health Sciences ,Psychology and Cognitive Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences ,Psychology - Abstract
Increased ligand binding to cellular integrins ("activation") plays important roles in processes such as development, cell migration, extracellular matrix assembly, tumor metastasis, hemostasis, and thrombosis. Integrin activation encompasses both increased integrin monomer affinity and increased receptor clustering and depends on integrin-talin interactions. Loss of kindlins results in reduced activation of integrins. Kindlins might promote talin binding to integrins through a cooperative mechanism; however, kindlins do not increase talin association with integrins. Here, we report that, unlike talin head domain (THD), kindlin-3 has little effect on the affinity of purified monomeric αIIbβ3, and it does not enhance activation by THD. Furthermore, studies with ligands of varying valency show that kindlins primarily increase cellular αIIbβ3 avidity rather than monomer affinity. In platelets or nucleated cells, loss of kindlins markedly reduces αIIbβ3 binding to multivalent but not monovalent ligands. Finally, silencing of kindlins reduces the clustering of ligand-occupied αIIbβ3 as revealed by total internal reflection fluorescence and electron microscopy. Thus, in contrast to talins, kindlins have little primary effect on integrin αIIbβ3 affinity for monovalent ligands and increase multivalent ligand binding by promoting the clustering of talin-activated integrins.
- Published
- 2013
45. Tiam1 interaction with the PAR complex promotes talin-mediated Rac1 activation during polarized cell migration
- Author
-
Wang, Shujie, Watanabe, Takashi, Matsuzawa, Kenji, Katsumi, Akira, Kakeno, Mai, Matsui, Toshinori, Ye, Feng, Sato, Kazuhide, Murase, Kiyoko, Sugiyama, Ikuko, Kimura, Kazushi, Mizoguchi, Akira, Ginsberg, Mark H, Collard, John G, and Kaibuchi, Kozo
- Subjects
Underpinning research ,1.1 Normal biological development and functioning ,Generic health relevance ,Adaptor Proteins ,Signal Transducing ,Animals ,COS Cells ,Cell Adhesion ,Cell Communication ,Cell Cycle Proteins ,Cell Line ,Tumor ,Cell Movement ,Cell Polarity ,Chlorocebus aethiops ,Guanine Nucleotide Exchange Factors ,HEK293 Cells ,HeLa Cells ,Humans ,Integrins ,Membrane Proteins ,Protein Kinase C ,RNA Interference ,RNA ,Small Interfering ,Signal Transduction ,T-Lymphoma Invasion and Metastasis-inducing Protein 1 ,Talin ,Vero Cells ,rac1 GTP-Binding Protein ,Hela Cells ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology - Abstract
Migrating cells acquire front-rear polarity with a leading edge and a trailing tail for directional movement. The Rac exchange factor Tiam1 participates in polarized cell migration with the PAR complex of PAR3, PAR6, and atypical protein kinase C. However, it remains largely unknown how Tiam1 is regulated and contributes to the establishment of polarity in migrating cells. We show here that Tiam1 interacts directly with talin, which binds and activates integrins to mediate their signaling. Tiam1 accumulated at adhesions in a manner dependent on talin and the PAR complex. The interactions of talin with Tiam1 and the PAR complex were required for adhesion-induced Rac1 activation, cell spreading, and migration toward integrin substrates. Furthermore, Tiam1 acted with talin to regulate adhesion turnover. Thus, we propose that Tiam1, with the PAR complex, binds to integrins through talin and, together with the PAR complex, thereby regulates Rac1 activity and adhesion turnover for polarized migration.
- Published
- 2012
46. Structural basis of the junctional anchorage of the cerebral cavernous malformations complex
- Author
-
Gingras, Alexandre R, Liu, Jian J, and Ginsberg, Mark H
- Subjects
Brain Disorders ,Rare Diseases ,Cardiovascular ,Underpinning research ,1.1 Normal biological development and functioning ,Animals ,Cardiovascular System ,Cells ,Cultured ,Disease Models ,Animal ,Endothelial Cells ,HEK293 Cells ,Hemangioma ,Cavernous ,Central Nervous System ,Humans ,Intercellular Junctions ,KRIT1 Protein ,Membrane Glycoproteins ,Microtubule-Associated Proteins ,Models ,Molecular ,Protein Conformation ,Proto-Oncogene Proteins ,Signal Transduction ,Zebrafish ,Zebrafish Proteins ,rho-Associated Kinases ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology - Abstract
The products of genes that cause cerebral cavernous malformations (CCM1/KRIT1, CCM2, and CCM3) physically interact. CCM1/KRIT1 links this complex to endothelial cell (EC) junctions and maintains junctional integrity in part by inhibiting RhoA. Heart of glass (HEG1), a transmembrane protein, associates with KRIT1. In this paper, we show that the KRIT1 band 4.1, ezrin, radixin, and moesin (FERM) domain bound the HEG1 C terminus (K(d) = 1.2 µM) and solved the structure of this assembly. The KRIT1 F1 and F3 subdomain interface formed a hydrophobic groove that binds HEG1(Tyr(1,380)-Phe(1,381)), thus defining a new mode of FERM domain-membrane protein interaction. This structure enabled design of KRIT1(L717,721A), which exhibited a >100-fold reduction in HEG1 affinity. Although well folded and expressed, KRIT1(L717,721A) failed to target to EC junctions or complement the effects of KRIT1 depletion on zebrafish cardiovascular development or Rho kinase activation in EC. These data establish that this novel FERM-membrane protein interaction anchors CCM1/KRIT1 at EC junctions to support cardiovascular development.
- Published
- 2012
47. Talin activates integrins by altering the topology of the β transmembrane domain
- Author
-
Kim, Chungho, Ye, Feng, Hu, Xiaohui, and Ginsberg, Mark H
- Subjects
Animals ,CHO Cells ,Cricetinae ,HEK293 Cells ,Humans ,Integrin beta Chains ,Protein Structure ,Tertiary ,Signal Transduction ,Talin ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology - Abstract
Talin binding to integrin β tails increases ligand binding affinity (activation). Changes in β transmembrane domain (TMD) topology that disrupt α-β TMD interactions are proposed to mediate integrin activation. In this paper, we used membrane-embedded integrin β3 TMDs bearing environmentally sensitive fluorophores at inner or outer membrane water interfaces to monitor talin-induced β3 TMD motion in model membranes. Talin binding to the β3 cytoplasmic domain increased amino acid side chain embedding at the inner and outer borders of the β3 TMD, indicating altered topology of the β3 TMD. Talin's capacity to effect this change depended on its ability to bind to both the integrin β tail and the membrane. Introduction of a flexible hinge at the midpoint of the β3 TMD decoupled the talin-induced change in intracellular TMD topology from the extracellular side and blocked talin-induced activation of integrin αIIbβ3. Thus, we show that talin binding to the integrin β TMD alters the topology of the TMD, resulting in integrin activation.
- Published
- 2012
48. RLIP76 (RalBP1) Is an R-Ras Effector That Mediates Adhesion-Dependent Rac Activation and Cell Migration
- Author
-
Goldfinger, Lawrence E., Shabanowitz, Jeffrey, Hunt, Donald F., and Ginsberg, Mark H.
- Published
- 2006
49. A mechanism of Rap1-induced stabilization of endothelial cell–cell junctions
- Author
-
Liu, Jian J, Stockton, Rebecca A, Gingras, Alexandre R, Ablooglu, Ararat J, Han, Jaewon, Bobkov, Andrey A, and Ginsberg, Mark H
- Subjects
Cardiovascular ,Brain Disorders ,Underpinning research ,1.1 Normal biological development and functioning ,Animals ,Cardiovascular System ,Endothelial Cells ,Gene Expression ,Genetic Vectors ,HEK293 Cells ,Humans ,Intercellular Junctions ,KRIT1 Protein ,Microtubule-Associated Proteins ,Microtubules ,Monomeric GTP-Binding Proteins ,Muscle Proteins ,Protein Conformation ,Protein Structure ,Tertiary ,Proto-Oncogene Proteins ,RNA ,Small Interfering ,Signal Transduction ,Structure-Activity Relationship ,Umbilical Veins ,Zebrafish ,Zebrafish Proteins ,rap1 GTP-Binding Proteins ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology - Abstract
Activation of Rap1 small GTPases stabilizes cell--cell junctions, and this activity requires Krev Interaction Trapped gene 1 (KRIT1). Loss of KRIT1 disrupts cardiovascular development and causes autosomal dominant familial cerebral cavernous malformations. Here we report that native KRIT1 protein binds the effector loop of Rap1A but not H-Ras in a GTP-dependent manner, establishing that it is an authentic Rap1-specific effector. By modeling the KRIT1-Rap1 interface we designed a well-folded KRIT1 mutant that exhibited a ~40-fold-reduced affinity for Rap1A and maintained other KRIT1-binding functions. Direct binding of KRIT1 to Rap1 stabilized endothelial cell-cell junctions in vitro and was required for cardiovascular development in vivo. Mechanistically, Rap1 binding released KRIT1 from microtubules, enabling it to locate to cell--cell junctions, where it suppressed Rho kinase signaling and stabilized the junctions. These studies establish that the direct physical interaction of Rap1 with KRIT1 enables the translocation of microtubule-sequestered KRIT1 to junctions, thereby supporting junctional integrity and cardiovascular development.
- Published
- 2011
50. $\alpha_4\beta_1-Dependent$ Adhesion Strengthening under Mechanical Strain Is Regulated by Paxillin Association with the $\alpha_4-Cytoplasmic$ Domain
- Author
-
Alon, Ronen, Feigelson, Sara W., Manevich, Eugenia, Rose, David M., Schmitz, Julia, Overby, Darryl R., Winter, Eitan, Grabovsky, Valentin, Shinder, Vera, Matthews, Benjamin D., Sokolovsky-Eisenberg, Maya, Ingber, Donald E., Benoit, Martin, and Ginsberg, Mark H.
- Published
- 2005
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.