Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters











Publication year range
1.
EMBO J ; 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39026000

ABSTRACT

The cellular cortex provides crucial mechanical support and plays critical roles during cell division and migration. The proteins of the ERM family, comprised of ezrin, radixin, and moesin, are central to these processes by linking the plasma membrane to the actin cytoskeleton. To investigate the contributions of the ERM proteins to leukocyte migration, we generated single and triple ERM knockout macrophages. Surprisingly, we found that even in the absence of ERM proteins, macrophages still form the different actin structures promoting cell migration, such as filopodia, lamellipodia, podosomes, and ruffles. Furthermore, we discovered that, unlike every other cell type previously investigated, the single or triple knockout of ERM proteins does not affect macrophage migration in diverse contexts. Finally, we demonstrated that the loss of ERMs in macrophages does not affect the mechanical properties of their cortex. These findings challenge the notion that ERMs are universally essential for cortex mechanics and cell migration and support the notion that the macrophage cortex may have diverged from that of other cells to allow for their uniquely adaptive cortical plasticity.

2.
bioRxiv ; 2024 May 15.
Article in English | MEDLINE | ID: mdl-38798563

ABSTRACT

Osteoclasts are multinucleated cells unique in their ability to resorb bone. Osteoclastogenesis involves several steps of actin-driven rearrangements that participate not only in the cell-cell fusion process, but also in the formation of the sealing zone, the adhesive structure determining the resorption area. Despite the importance of these actin cytoskeleton-based processes, their precise mechanisms of regulation are still poorly characterized. Here, we found that moesin, a member of the Ezrin/Radixin/Moesin (ERM) protein family, is activated during osteoclast maturation and plays an instrumental role for both osteoclast fusion and function. In mouse and human osteoclast precursors, moesin is negatively regulated to potentiate their ability to fuse and degrade bone. Accordingly, we demonstrated that moesin depletion decreases membrane-to-cortex attachment and enhances formation of tunneling nanotubes (TNTs), F-actin-containing intercellular bridges that we revealed to trigger osteoclast fusion. In addition, via a ß3-integrin/RhoA/SLK pathway and independently of its role in fusion, moesin regulates the number and organization of sealing zones in mature osteoclast, and thus participates in the control of bone resorption. Supporting these findings, we found that moesin-deficient mice are osteopenic with a reduced density of trabecular bones and increased osteoclast abundance and activity. These findings provide a better understanding of the regulation of osteoclast biology, and open new opportunities to specifically target osteoclast activity in bone disease therapy.

3.
Cell Rep ; 42(8): 112996, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37598341

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.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , T-Lymphocytes, Regulatory , Mice , Animals , Interleukin-2/metabolism , Receptors, Chemokine/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Receptors, Interleukin-2/metabolism , Signal Transduction , Forkhead Transcription Factors/metabolism
4.
Front Cell Dev Biol ; 10: 908622, 2022.
Article in English | MEDLINE | ID: mdl-35721481

ABSTRACT

Integrins regulate the adhesion and migration of blood cells to ensure the proper positioning of these cells in the environment. Integrins detect physical and chemical stimuli in the extracellular matrix and regulate signaling pathways in blood cells that mediate their functions. Integrins are usually in a resting state in blood cells until agonist stimulation results in a high-affinity conformation ("integrin activation"), which is central to integrins' contribution to blood cells' trafficking and functions. In this review, we summarize the mechanisms of integrin activation in blood cells with a focus on recent advances understanding of mechanisms whereby Rap1 regulates talin1-integrin interaction to trigger integrin activation in lymphocytes, platelets, and neutrophils.

5.
J Exp Med ; 219(8)2022 08 01.
Article in English | MEDLINE | ID: mdl-35766979

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 αLß2 and α4ß7. 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.


Subject(s)
Integrins , Lymphoid Tissue , Protein Phosphatase 1 , T-Lymphocytes , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Adhesion/physiology , Colitis/immunology , Colitis/metabolism , Integrins/immunology , Integrins/metabolism , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Membrane Proteins/metabolism , Mice , Protein Phosphatase 1/immunology , Protein Phosphatase 1/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Talin/metabolism , rap1 GTP-Binding Proteins/immunology , rap1 GTP-Binding Proteins/metabolism
6.
J Immunol ; 208(6): 1378-1388, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35197328

ABSTRACT

Agonist-induced Rap1 GTP loading results in integrin activation involved in T cell trafficking and functions. MRL proteins Rap1-interacting adapter molecule (RIAM) and lamellipodin (LPD) are Rap1 effectors that can recruit talin1 to integrins, resulting in integrin activation. Recent work also implicates direct Rap1-talin1 interaction in integrin activation. Here, we analyze in mice the connections between Rap1 and talin1 that support integrin activation in conventional CD4+ T (Tconv) and CD25HiFoxp3+CD4+ regulatory T (Treg) cells. Talin1(R35E, R118E) mutation that disrupts both Rap1 binding sites results in a partial defect in αLß2, α4ß1, and α4ß7 integrin activation in both Tconv and Treg cells with resulting defects in T cell homing. Talin1(R35E,R118E) Tconv manifested reduced capacity to induce colitis in an adoptive transfer mouse model. Loss of RIAM exacerbates the defects in Treg cell function caused by the talin1(R35E,R118E) mutation, and deleting both MRL proteins in combination with talin1(R35E,R118E) phenocopy the complete lack of integrin activation observed in Rap1a/b-null Treg cells. In sum, these data reveal the functionally significant connections between Rap1 and talin1 that enable αLß2, α4ß1, and α4ß7 integrin activation in CD4+ T cells.


Subject(s)
Talin , rap1 GTP-Binding Proteins , Animals , Binding Sites , CD4-Positive T-Lymphocytes/metabolism , Integrins/metabolism , Mice , Talin/genetics , Talin/metabolism , rap1 GTP-Binding Proteins/metabolism
7.
J Clin Invest ; 131(13)2021 07 01.
Article in English | MEDLINE | ID: mdl-34043589

ABSTRACT

Cerebral cavernous malformations (CCMs) are common neurovascular lesions caused by loss-of-function mutations in 1 of 3 genes, including KRIT1 (CCM1), CCM2, and PDCD10 (CCM3), and generally regarded as an endothelial cell-autonomous disease. Here we reported that proliferative astrocytes played a critical role in CCM pathogenesis by serving as a major source of VEGF during CCM lesion formation. An increase in astrocyte VEGF synthesis is driven by endothelial nitric oxide (NO) generated as a consequence of KLF2- and KLF4-dependent elevation of eNOS in CCM endothelium. The increased brain endothelial production of NO stabilized HIF-1α in astrocytes, resulting in increased VEGF production and expression of a "hypoxic" program under normoxic conditions. We showed that the upregulation of cyclooxygenase-2 (COX-2), a direct HIF-1α target gene and a known component of the hypoxic program, contributed to the development of CCM lesions because the administration of a COX-2 inhibitor significantly prevented the progression of CCM lesions. Thus, non-cell-autonomous crosstalk between CCM endothelium and astrocytes propels vascular lesion development, and components of the hypoxic program represent potential therapeutic targets for CCMs.


Subject(s)
Astrocytes/physiology , Hemangioma, Cavernous, Central Nervous System/physiopathology , Animals , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/genetics , Astrocytes/pathology , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Disease Models, Animal , Disease Progression , Endothelial Cells/metabolism , Hemangioma, Cavernous, Central Nervous System/etiology , Hemangioma, Cavernous, Central Nervous System/pathology , Human Umbilical Vein Endothelial Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Knockout , Models, Neurological , Mutation , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Vascular Endothelial Growth Factor A/biosynthesis
8.
J Biol Chem ; 296: 100675, 2021.
Article in English | MEDLINE | ID: mdl-33865854

ABSTRACT

Interaction of talin with the cytoplasmic tails of integrin ß triggers integrin activation, leading to an increase of integrin affinity/avidity for extracellular ligands. In talin KO mice, loss of talin interaction with platelet integrin αIIbß3 causes a severe hemostatic defect, and loss of talin interaction with endothelial cell integrin αVß3 affects angiogenesis. In normal cells, talin is autoinhibited and localized in the cytoplasm. Here, we used an optogenetic platform to assess whether recruitment of full-length talin to the plasma membrane was sufficient to induce integrin activation. A dimerization module (Arabidopsis cryptochrome 2 fused to the N terminus of talin; N-terminal of cryptochrome-interacting basic helix-loop-helix domain ended with a CAAX box protein [C: cysteine; A: aliphatic amino acid; X: any C-terminal amino acid]) responsive to 450 nm (blue) light was inserted into Chinese hamster ovary cells and endothelial cells also expressing αIIbß3 or αVß3, respectively. Thus, exposure of the cells to blue light caused a rapid and reversible recruitment of Arabidopsis cryptochrome 2-talin to the N-terminal of cryptochrome-interacting basic helix-loop-helix domain ended with a CAAX box protein [C: cysteine; A: aliphatic amino acid; X: any C-terminal amino acid]-decorated plasma membrane. This resulted in ß3 integrin activation in both cell types, as well as increasing migration of the endothelial cells. However, membrane recruitment of talin was not sufficient for integrin activation, as membrane-associated Ras-related protein 1 (Rap1)-GTP was also required. Moreover, talin mutations that interfered with its direct binding to Rap1 abrogated ß3 integrin activation. Altogether, these results define a role for the plasma membrane recruitment of talin in ß3 integrin activation, and they suggest a nuanced sequence of events thereafter involving Rap1-GTP.


Subject(s)
Cell Membrane/metabolism , Cytoplasm/metabolism , Endothelial Cells/metabolism , Optogenetics , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Talin/metabolism , rap1 GTP-Binding Proteins/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Mice , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Protein Binding , Talin/genetics , rap1 GTP-Binding Proteins/genetics
9.
Structure ; 29(4): 305-307, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33798425

ABSTRACT

In this issue of Structure, Cho et al. (2020) identified an intermolecular interaction between two RIAM pleckstrin homology (PH) domains that masks the phosphoinositide-binding site, and that phosphorylation by Src unmasks the PH domain. This provides an explanation of how RIAM plasma membrane translocation is regulated to promote integrin activation.


Subject(s)
Adaptor Proteins, Signal Transducing , Pleckstrin Homology Domains , Adaptor Proteins, Signal Transducing/metabolism , Integrins/metabolism , Membrane Proteins/metabolism , Phosphorylation
10.
J Exp Med ; 218(2)2021 02 01.
Article in English | MEDLINE | ID: mdl-33104169

ABSTRACT

Integrin activation mediates lymphocyte trafficking and immune functions. Conventional T cell (Tconv cell) integrin activation requires Rap1-interacting adaptor molecule (RIAM). Here, we report that Apbb1ip-/- (RIAM-null) mice are protected from spontaneous colitis due to IL-10 deficiency, a model of inflammatory bowel disease (IBD). Protection is ascribable to reduced accumulation and homing of Tconv cells in gut-associated lymphoid tissue (GALT). Surprisingly, there are abundant RIAM-null regulatory T cells (T reg cells) in the GALT. RIAM-null T reg cells exhibit normal homing to GALT and lymph nodes due to preserved activation of integrins αLß2, α4ß1, and α4ß7. Similar to Tconv cells, T reg cell integrin activation and immune function require Rap1; however, lamellipodin (Raph1), a RIAM paralogue, compensates for RIAM deficiency. Thus, in contrast to Tconv cells, RIAM is dispensable for T reg cell integrin activation and suppressive function. In consequence, inhibition of RIAM can inhibit spontaneous Tconv cell-mediated autoimmune colitis while preserving T reg cell trafficking and function.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Integrins/immunology , Integrins/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Colitis/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymphocyte Activation/immunology , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Mice , Mice, Inbred C57BL
11.
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
12.
Blood ; 136(10): 1180-1190, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32518959

ABSTRACT

Ras-related protein 1 (Rap1) is a major convergence point of the platelet-signaling pathways that result in talin-1 binding to the integrin ß cytoplasmic domain and consequent integrin activation, platelet aggregation, and effective hemostasis. The nature of the connection between Rap1 and talin-1 in integrin activation is an important remaining gap in our understanding of this process. Previous work identified a low-affinity Rap1-binding site in the talin-1 F0 domain that makes a small contribution to integrin activation in platelets. We recently identified an additional Rap1-binding site in the talin-1 F1 domain that makes a greater contribution than F0 in model systems. Here we generated mice bearing point mutations, which block Rap1 binding without affecting talin-1 expression, in either the talin-1 F1 domain (R118E) alone, which were viable, or in both the F0 and F1 domains (R35E,R118E), which were embryonic lethal. Loss of the Rap1-talin-1 F1 interaction in platelets markedly decreases talin-1-mediated activation of platelet ß1- and ß3-integrins. Integrin activation and platelet aggregation in mice whose platelets express only talin-1(R35E, R118E) are even more impaired, resembling the defect seen in platelets lacking both Rap1a and Rap1b. Although Rap1 is important in thrombopoiesis, platelet secretion, and surface exposure of phosphatidylserine, loss of the Rap1-talin-1 interaction in talin-1(R35E, R118E) platelets had little effect on these processes. These findings show that talin-1 is the principal direct effector of Rap1 GTPases that regulates platelet integrin activation in hemostasis.


Subject(s)
Integrin beta1/metabolism , Integrin beta3/metabolism , Point Mutation , Talin/physiology , Thrombopoiesis , rap GTP-Binding Proteins/physiology , rap1 GTP-Binding Proteins/physiology , Animals , Female , Integrin beta1/genetics , Integrin beta3/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Activation , Platelet Aggregation , Protein Domains , Signal Transduction
13.
J Cell Biol ; 218(6): 1799-1809, 2019 06 03.
Article in English | MEDLINE | ID: mdl-30988001

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.


Subject(s)
Integrins/metabolism , Lipids/physiology , Talin/metabolism , Telomere-Binding Proteins/metabolism , Animals , Binding Sites , CHO Cells , Cricetinae , Cricetulus , Humans , Mutation , Protein Binding , Protein Conformation , Protein Domains , Shelterin Complex , Talin/chemistry , Talin/genetics , Telomere-Binding Proteins/genetics
14.
Blood ; 133(3): 193-204, 2019 01 17.
Article in English | MEDLINE | ID: mdl-30442679

ABSTRACT

Cerebral cavernous malformations (CCMs) are common brain vascular dysplasias that are prone to acute and chronic hemorrhage with significant clinical sequelae. The pathogenesis of recurrent bleeding in CCM is incompletely understood. Here, we show that central nervous system hemorrhage in CCMs is associated with locally elevated expression of the anticoagulant endothelial receptors thrombomodulin (TM) and endothelial protein C receptor (EPCR). TM levels are increased in human CCM lesions, as well as in the plasma of patients with CCMs. In mice, endothelial-specific genetic inactivation of Krit1 (Krit1 ECKO ) or Pdcd10 (Pdcd10 ECKO ), which cause CCM formation, results in increased levels of vascular TM and EPCR, as well as in enhanced generation of activated protein C (APC) on endothelial cells. Increased TM expression is due to upregulation of transcription factors KLF2 and KLF4 consequent to the loss of KRIT1 or PDCD10. Increased TM expression contributes to CCM hemorrhage, because genetic inactivation of 1 or 2 copies of the Thbd gene decreases brain hemorrhage in Pdcd10 ECKO mice. Moreover, administration of blocking antibodies against TM and EPCR significantly reduced CCM hemorrhage in Pdcd10 ECKO mice. Thus, a local increase in the endothelial cofactors that generate anticoagulant APC can contribute to bleeding in CCMs, and plasma soluble TM may represent a biomarker for hemorrhagic risk in CCMs.


Subject(s)
Anticoagulants/metabolism , Apoptosis Regulatory Proteins/physiology , Cerebral Hemorrhage/diagnosis , Endothelium, Vascular/pathology , Hemangioma, Cavernous, Central Nervous System/complications , KRIT1 Protein/physiology , Membrane Proteins/physiology , Protein C/metabolism , Proto-Oncogene Proteins/physiology , Thrombomodulin/blood , Adult , Animals , Blood Coagulation , Case-Control Studies , Cerebral Hemorrhage/blood , Cerebral Hemorrhage/etiology , Endothelial Protein C Receptor/metabolism , Endothelium, Vascular/metabolism , Hemangioma, Cavernous, Central Nervous System/metabolism , Hemangioma, Cavernous, Central Nervous System/physiopathology , Humans , Kruppel-Like Factor 4 , Mice , Mice, Knockout , Signal Transduction , Young Adult
15.
Blood Adv ; 2(18): 2358-2368, 2018 09 25.
Article in English | MEDLINE | ID: mdl-30242097

ABSTRACT

Activation of platelet glycoprotein IIb-IIIa (GPIIb-IIIa; integrin αIIbß3) leads to high-affinity fibrinogen binding and platelet aggregation during hemostasis. Whereas GTP-bound Rap1 GTPase promotes talin 1 binding to the ß3 cytoplasmic domain to activate platelet GPIIb-IIIa, the Rap1 effector that regulates talin association with ß3 in platelets is unknown. Rap1 binding to the talin 1 F0 subdomain was proposed to forge the talin 1-Rap1 link in platelets. Here, we report a talin 1 point mutant (R35E) that significantly reduces Rap1 affinity without a significant effect on its structure or expression. Talin 1 head domain (THD) (R35E) was of similar potency to wild-type THD in activating αIIbß3 in Chinese hamster ovary cells. Coexpression with activated Rap1b increased activation, and coexpression with Rap1GAP1 reduced activation caused by transfection of wild-type THD or THD(R35E). Furthermore, platelets from Tln1R35E/R35E mice showed similar GPIIb-IIIa activation to those from wild-type littermates in response to multiple agonists. Tln1R35E/R35E platelets exhibited slightly reduced platelet aggregation in response to low doses of agonists; however, there was not a significant hemostatic defect, as judged by tail bleeding times. Thus, the Rap1-talin 1 F0 interaction has little effect on platelet GPIIb-IIIa activation and hemostasis and cannot account for the dramatic effects of loss of Rap1 activity on these platelet functions.


Subject(s)
Blood Platelets/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/agonists , Protein Interaction Domains and Motifs , Talin/metabolism , rap1 GTP-Binding Proteins/metabolism , Animals , Blood Cell Count , CHO Cells , Cricetulus , Female , Male , Mice , Mice, Transgenic , Models, Molecular , Mutation , Platelet Function Tests , Protein Conformation , Talin/chemistry , Talin/genetics
16.
J Cell Biol ; 217(4): 1453-1465, 2018 04 02.
Article in English | MEDLINE | ID: mdl-29535192

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 ß3 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 α4ß7, 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 α4ß7, thereby inhibiting rolling lymphocyte arrest, a key step in transmigration. Furthermore, the α4ß7(L721P) mutation blocks lymphocyte homing to and development of the GALT. These studies show that impairing the ability of an integrin ß TMD to transmit talin-induced TMD topology inhibits agonist-induced physiological integrin activation and biological function in development.


Subject(s)
Gastrointestinal Tract/metabolism , Integrin beta Chains/metabolism , Integrins/metabolism , Lymphocytes/metabolism , Lymphoid Tissue/metabolism , Animals , Cell Adhesion , Female , Gastrointestinal Tract/cytology , Gastrointestinal Tract/immunology , HEK293 Cells , Humans , Integrin beta Chains/chemistry , Integrin beta Chains/genetics , Integrins/chemistry , Integrins/genetics , Jurkat Cells , Leukocyte Rolling , Lymphocyte Activation , Lymphocytes/immunology , Lymphoid Tissue/cytology , Male , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Protein Binding , Protein Interaction Domains and Motifs , Signal Transduction , Structure-Activity Relationship , Talin/genetics , Talin/metabolism
17.
J Exp Med ; 214(11): 3331-3346, 2017 Nov 06.
Article in English | MEDLINE | ID: mdl-28970240

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.


Subject(s)
Genetic Therapy/methods , Hemangioma, Cavernous, Central Nervous System/metabolism , Hemangioma, Cavernous, Central Nervous System/therapy , KRIT1 Protein/metabolism , Thrombospondin 1/metabolism , Animals , Cells, Cultured , Endothelial Cells/metabolism , Gene Expression Profiling/methods , HEK293 Cells , Hemangioma, Cavernous, Central Nervous System/genetics , Humans , KRIT1 Protein/genetics , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , RNA Interference , Thrombospondin 1/genetics
18.
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
19.
Blood ; 128(4): 479-87, 2016 07 28.
Article in English | MEDLINE | ID: mdl-27207789

ABSTRACT

Integrin adhesion receptors mediate the adhesion of blood cells, such as leukocytes, to other cells, such as endothelial cells. Integrins also are critical for anchorage of hematopoietic precursors to the extracellular matrix. Blood cells can dynamically regulate the affinities of integrins for their ligands ("activation"), an event central to their functions. Here we review recent progress in understanding the mechanisms of integrin activation with a focus on the functions of blood cells. We discuss how talin binding to the integrin ß cytoplasmic domain, in conjunction with the plasma membrane, induces long-range allosteric rearrangements that lead to integrin activation. Second, we review our understanding of how signaling events, particularly those involving Rap1 small guanosine triphosphate (GTP)hydrolases, can regulate the talin-integrin interaction and resulting activation. Third, we review recent findings that highlight the role of the Rap1-GTP-interacting adapter molecule (RIAM), encoded by the APBB1IP gene, in leukocyte integrin activation and consequently in leukocyte trafficking.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Movement/physiology , Integrins/metabolism , Leukocytes/metabolism , Membrane Proteins/metabolism , Talin/metabolism , Telomere-Binding Proteins/metabolism , Humans , Leukocytes/cytology , Protein Domains , Shelterin Complex
20.
Nat Commun ; 6: 8492, 2015 Sep 30.
Article in English | MEDLINE | ID: mdl-26419705

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.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carrier Proteins/metabolism , Cell Movement , Cells/metabolism , Integrins/metabolism , Membrane Proteins/metabolism , Talin/metabolism , Adaptor Proteins, Signal Transducing/genetics , Carrier Proteins/genetics , Cells/cytology , Humans , Integrins/genetics , Membrane Proteins/genetics , Protein Binding , Talin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL