Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Clin Immunol ; 43(4): 741-746, 2023 05.
Article in English | MEDLINE | ID: mdl-36648575

ABSTRACT

Leukocyte adhesion deficiency-III (LAD-III) is an extremely rare autosomal recessive syndrome caused by mutations in FERMT3, the gene encoding kindlin-3. The genetic alterations in this gene lead to abnormal expression or activity of kindlin-3 in leukocytes and platelets. Kindlin-3 acts as an important regulator of integrin activation. LAD-III has features of the bleeding syndrome of Glanzmann and also of leukocyte adhesion deficiency. In this study, we report on two families, one of Turkish and one of Syrian origin, with clinical features of LAD-III, loss of kindlin-3 protein expression, and a functional leukocyte defect. A novel, homozygous deletion in FERMT3 (c.921delC, p.Ser307Argfs*21) was found in the Turkish patient. The parents were carriers of the mutation, consistent with an autosomal recessive inheritance. A common c.1525C > T (p.Arg509*) mutation was found in the Syrian patient. In conclusion, beside the variant c.1525C > T in the FERMT3 gene, which was previously found in more than 15 patients in Anatolia, our study is the first to identify the novel homozygous variant c.921delC in the FERMT3 gene.


Subject(s)
Leukocyte-Adhesion Deficiency Syndrome , Humans , CD18 Antigens/metabolism , Cell Adhesion/genetics , Homozygote , Leukocyte-Adhesion Deficiency Syndrome/genetics , Sequence Deletion/genetics , Turkey
2.
J Clin Immunol ; 43(6): 1250-1258, 2023 08.
Article in English | MEDLINE | ID: mdl-37014583

ABSTRACT

BACKGROUND: Leukocyte and platelet integrin function defects are present in leukocyte adhesion deficiency type III (LAD-III) due to mutations in FERMT3. Additionally, osteoclast/osteoblast dysfunction develops in LAD-III. AIM: To discuss the distinguishing clinical, radiological, and laboratory features of LAD-III. METHODS: This study included the clinical, radiological, and laboratory characteristics of twelve LAD-III patients. RESULTS: The male/female ratio was 8/4. The parental consanguinity ratio was 100%. Half of the patients had a family history of patients with similar findings. The median age at presentation and diagnosis was 18 (1-60) days and 6 (1-20) months, respectively. The median leukocyte count on admission was 43,150 (30,900-75,700)/µL. The absolute eosinophil count was tested in 8/12 patients, and eosinophilia was found in 6/8 (75%). All patients had a history of sepsis. Other severe infections were pneumonia (66.6%), omphalitis (25%), osteomyelitis (16.6%), gingivitis/periodontitis (16%), chorioretinitis (8.3%), otitis media (8.3%), diarrhea (8.3%), and palpebral conjunctiva infection (8.3%). Four patients (33.3%) received hematopoietic stem cell transplantation (HSCT) from HLA-matched-related donors, and one deceased after HSCT. At initial presentation, 4 (33.3%) patients were diagnosed with other hematologic disorders, three patients (P5, P7, and P8) with juvenile myelomonocytic leukemia (JMML), and one (P2) with myelodysplastic syndrome (MDS). CONCLUSION: In LAD-III, leukocytosis, eosinophilia, and bone marrow findings may mimic pathologies such as JMML and MDS. In addition to non-purulent infection susceptibility, patients with LAD-III exhibit Glanzmann-type bleeding disorder. In LAD-III, absent integrin activation due to kindlin-3 deficiency disrupts osteoclast actin cytoskeleton organization. This results in defective bone resorption and osteopetrosis-like radiological changes. These are distinctive features compared to other LAD types.


Subject(s)
Leukocyte-Adhesion Deficiency Syndrome , Osteopetrosis , Humans , Male , Female , Osteopetrosis/diagnosis , Osteopetrosis/genetics , Leukocyte-Adhesion Deficiency Syndrome/diagnosis , Leukocyte-Adhesion Deficiency Syndrome/genetics , Integrins/physiology , Leukocytes/metabolism , Leukocytes/pathology
3.
J Cell Sci ; 134(8)2021 04 15.
Article in English | MEDLINE | ID: mdl-33912917

ABSTRACT

Integrin-mediated cell-extracellular matrix (ECM) interactions play crucial roles in a broad range of physiological and pathological processes. Kindlins are important positive regulators of integrin activation. The FERM-domain-containing kindlin family comprises three members, kindlin-1, kindlin-2 and kindlin-3 (also known as FERMT1, FERMT2 and FERMT3), which share high sequence similarity (identity >50%), as well as domain organization, but exhibit diverse tissue-specific expression patterns and cellular functions. Given the significance of kindlins, analysis of their atomic structures has been an attractive field for decades. Recently, the structures of kindlin and its ß-integrin-bound form have been obtained, which greatly advance our understanding of the molecular functions that involve kindlins. In particular, emerging evidence indicates that oligomerization of kindlins might affect their integrin binding and focal adhesion localization, positively or negatively. In this Review, we presented an update on the recent progress of obtaining kindlin structures, and discuss the implication for integrin activation based on kindlin oligomerization, as well as the possible regulation of this process.


Subject(s)
Membrane Proteins , Neoplasm Proteins , Cell Adhesion , Focal Adhesions , Integrins/genetics , Membrane Proteins/genetics , Neoplasm Proteins/genetics
4.
J Cell Sci ; 134(22)2021 11 15.
Article in English | MEDLINE | ID: mdl-34704600

ABSTRACT

Osteoclasts form special integrin-mediated adhesion structures called sealing zones that enable them to adhere to and resorb bone. Sealing zones consist of densely packed podosomes tightly interconnected by actin fibers. Their formation requires the presence of the hematopoietic integrin regulator kindlin-3 (also known as Fermt3). In this study, we investigated osteoclasts and their adhesion structures in kindlin-3 hypomorphic mice expressing only 5-10% of the kindlin-3 level of wild-type mice. Low kindlin-3 expression reduces integrin activity, results in impaired osteoclast adhesion and signaling, and delays cell spreading. Despite these defects, in vitro-generated kindlin-3-hypomorphic osteoclast-like cells arrange their podosomes into adhesion patches and belts, but their podosome and actin organization is abnormal. Remarkably, kindlin-3-hypomorphic osteoclasts form sealing zones when cultured on calcified matrix in vitro and on bone surface in vivo. However, functional assays, immunohistochemical staining and electron micrographs of bone sections showed that they fail to seal the resorption lacunae properly, which is required for secreted proteinases to digest bone matrix. This results in mild osteopetrosis. Our study reveals a new, hitherto understudied function of kindlin-3 as an essential organizer of integrin-mediated adhesion structures, such as sealing zones.


Subject(s)
Cytoskeletal Proteins , Osteoclasts , Osteopetrosis , Animals , Bone Matrix , Bone and Bones , Cytoskeletal Proteins/genetics , Integrins , Mice , Osteopetrosis/genetics
5.
Blood Cells Mol Dis ; 99: 102726, 2023 03.
Article in English | MEDLINE | ID: mdl-36696755

ABSTRACT

Leukocyte adhesion deficiency (LAD) is an immunodeficiency caused by defects in the adhesion of leukocytes (especially neutrophils) to the blood vessel wall. As a result, patients with LAD suffer from severe bacterial infections and impaired wound healing, accompanied by neutrophilia. In LAD-I, characterized directly after birth by delayed separation of the umbilical cord, mutations are found in ITGB2, the gene that encodes the ß subunit (CD18) of the ß2 integrins. In the rare LAD-II disease, the fucosylation of selectin ligands is disturbed, caused by mutations in SLC35C1, the gene that encodes a GDP-fucose transporter of the Golgi system. LAD-II patients lack the H and Lewis Lea and Leb blood group antigens. Finally, in LAD-III, the conformational activation of the hematopoietically expressed ß integrins is disturbed, leading to leukocyte and platelet dysfunction. This last syndrome is caused by mutations in FERMT3, encoding the kindlin-3 protein in all blood cells, involved in the regulation of ß integrin conformation. This article contains an update of the mutations that we consider to be relevant for the various forms of LAD.


Subject(s)
Leukocyte-Adhesion Deficiency Syndrome , Humans , Cell Adhesion/genetics , Leukocyte-Adhesion Deficiency Syndrome/genetics , CD18 Antigens/genetics , CD18 Antigens/metabolism , Leukocytes , Mutation
6.
Proc Natl Acad Sci U S A ; 117(39): 24326-24335, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32929022

ABSTRACT

Kindlin-3 (K3)-mediated integrin adhesion controls homing and bone marrow (BM) retention of normal hematopoietic cells. However, the role of K3 in leukemic stem cell (LSC) retention and growth in the remodeled tumor-promoting BM is unclear. We report that loss of K3 in a mouse model of chronic myeloid leukemia (CML) triggers the release of LSCs from the BM into the circulation and impairs their retention, proliferation, and survival in secondary organs, which curbs CML development, progression, and metastatic dissemination. We found de novo expression of cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) on CML-LSCs but not normal hematopoietic stem cells and this enabled us to specifically deplete K3 with a CTLA-4-binding RNA aptamer linked to a K3-siRNA (small interfering RNA) in CTLA-4+ LSCs in vivo, which mobilized LSCs in the BM, induced disease remission, and prolonged survival of mice with CML. Thus, disrupting interactions of LSCs with the BM environment is a promising strategy to halt the disease-inducing and relapse potential of LSCs.


Subject(s)
Bone Marrow/metabolism , Cytoskeletal Proteins/deficiency , Leukemia, Myeloid/metabolism , Neoplastic Stem Cells/metabolism , Animals , CTLA-4 Antigen/genetics , CTLA-4 Antigen/metabolism , Cell Movement , Cytoskeletal Proteins/genetics , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myeloid/genetics , Leukemia, Myeloid/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplastic Stem Cells/cytology , Stem Cell Niche , Tumor Microenvironment
7.
Exp Cell Res ; 399(2): 112456, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33417921

ABSTRACT

Identifying patient mutations driving skeletal development disorders has driven our understanding of bone development. Integrin adhesion deficiency disease is caused by a Kindlin-3 (fermitin family member 3) mutation, and its inactivation results in bleeding disorders and osteopenia. In this study, we uncover a role for Kindlin-3 in the differentiation of bone marrow mesenchymal stem cells (BMSCs) down the chondrogenic lineage. Kindlin-3 expression increased with chondrogenic differentiation, similar to RUNX2. BMSCs isolated from a Kindlin-3 deficient patient expressed chondrocyte markers, including SOX9, under basal conditions, which were further enhanced with chondrogenic differentiation. Rescue of integrin activation by a constitutively activated ß3 integrin construct increased adhesion to multiple extracellular matrices and reduced SOX9 expression to basal levels. Growth plates from mice expressing a mutated Kindlin-3 with the integrin binding site ablated demonstrated alterations in chondrocyte maturation similar to that seen with the human Kindlin-3 deficient BMSCs. These findings suggest that Kindlin-3 expression mirrors RUNX2 during chondrogenesis.


Subject(s)
Chondrogenesis/genetics , Cytoskeletal Proteins/genetics , Membrane Proteins/genetics , Mesenchymal Stem Cells/physiology , Neoplasm Proteins/genetics , Animals , Cell Differentiation/genetics , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Female , Humans , Male , Mesenchymal Stem Cells/metabolism , Mice , Mice, Transgenic , Mutation/physiology
8.
Pol J Pathol ; 72(3): 237-244, 2021.
Article in English | MEDLINE | ID: mdl-35048636

ABSTRACT

It is well known that metastasis is the most crucial factor in determining the fate of the patient. The prognosis of melanoma is very poor at the stage of metastasis. Recently, several genes and proteins, including kindlin3, dioxin receptor (AhR), RASSF6, and claudin-11, which were shown as possible prognostic biomarkers for human tumours, were described. In this study, we focused on these proteins in melanoma within a clinical setting. Forty-three primary melanomas (PMs), 17 metastatic melanomas (MMs), 15 melanocytic nevi (MN), and two melanoma cell lines were included in this retrospective study. All proteins were investigated using immunohistochemistry, and analysis was performed using a semi-quantitative immunoreactive score (IRS). The nevus group showed lower RASSF6 and AhR IRS levels than PMs. RASSF6 and kindlin-3 levels in the PMs with metastasis (MwM) and also in PMs showing lymphovascular invasion were significantly lower. The logistic regression model also proved that kindlin-3 expression was a significant independent predictor of metastasis. The current study supports the role of kindlin-3 and RASSF6 as prognostic biomarkers in melanoma. Besides the prognostic roles of these proteins, they are probably potential candidates for target-oriented therapies for melanoma metastasis blocking.


Subject(s)
Melanoma , Membrane Proteins , Neoplasm Proteins , Nevus , Skin Neoplasms , Apoptosis Regulatory Proteins , Biomarkers , Biomarkers, Tumor , Humans , Retrospective Studies , Melanoma, Cutaneous Malignant
9.
Int J Mol Sci ; 20(24)2019 Dec 06.
Article in English | MEDLINE | ID: mdl-31817594

ABSTRACT

Kindlins are important proteins for integrin signaling and regulation of the cytoskeleton, but we know little about their precise function and regulation in platelets during acute ischemic events. In this work, we investigated kindlin-3 protein levels in platelets isolated from patients with ST-elevation myocardial infarction (STEMI) compared to patients with non-ischemic chest pain. Platelets from twelve patients with STEMI and twelve patients with non-ischemic chest pain were isolated and analyzed for kindlin-3 protein levels and intracellular localization by immunoblotting and two-dimensional gel electrophoresis. Platelet proteome analysis by two-dimensional gel electrophoresis and protein sequencing identified kindlin-3 as a protein that is cleaved in platelets from patients with myocardial infarction. Kindlin-3 full-length protein was significantly decreased in patients with STEMI compared to patients with non-ischemic chest pain (1.0 ± 0.2 versus 0.28 ± 0.2, p < 0.05) by immunoblotting. Kindlin-3 showed a differential distribution and was primarily cleaved in the cytosolic and membrane compartment of platelets in myocardial infarction. Platelet activation with thrombin alone did not affect kindlin-3 protein levels. The present study demonstrates that kindlin-3 protein levels become significantly reduced in platelets of patients with myocardial infarction compared to controls. The results suggest that kindlin-3 cleavage in platelets is associated with the ischemic event of myocardial infarction.


Subject(s)
Blood Platelets/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , ST Elevation Myocardial Infarction/metabolism , Aged , Blood Platelets/drug effects , Electrophoresis, Gel, Two-Dimensional , Female , Humans , Immunoblotting , Male , Middle Aged , Thrombin/pharmacology
10.
J Biol Chem ; 291(19): 10363-71, 2016 May 06.
Article in English | MEDLINE | ID: mdl-26994136

ABSTRACT

Integrin-mediated rolling and firm cell adhesion are two critical steps in leukocyte trafficking. Integrin α4ß1 mediates a mixture of rolling and firm cell adhesion on vascular cell adhesion molecule-1 (VCAM-1) when in its resting state but only supports firm cell adhesion upon activation. The transition from rolling to firm cell adhesion is controlled by integrin activation. Kindlin-3 has been shown to bind to integrin ß tails and trigger integrin activation via inside-out signaling. However, the role of kindlin-3 in regulating resting α4ß1-mediated cell adhesion is not well characterized. Herein we demonstrate that kindlin-3 was required for the resting α4ß1-mediated firm cell adhesion but not rolling adhesion. Knockdown of kindlin-3 significantly decreased the binding of kindlin-3 to ß1 and down-regulated the binding affinity of the resting α4ß1 to soluble VCAM-1. Notably, it converted the resting α4ß1-mediated firm cell adhesion to rolling adhesion on VCAM-1 substrates, increased cell rolling velocity, and impaired the stability of cell adhesion. By contrast, firm cell adhesion mediated by Mn(2+)-activated α4ß1 was barely affected by knockdown of kindlin-3. Structurally, lack of kindlin-3 led to a more bent conformation of the resting α4ß1. Thus, kindlin-3 plays an important role in maintaining a proper conformation of the resting α4ß1 to mediate both rolling and firm cell adhesion. Defective kindlin-3 binding to the resting α4ß1 leads to a transition from firm to rolling cell adhesion on VCAM-1, implying its potential role in regulating the transition between integrin-mediated rolling and firm cell adhesion.


Subject(s)
Integrin alpha4beta1/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Signal Transduction/physiology , Cell Adhesion/physiology , Humans , Integrin alpha4beta1/genetics , K562 Cells , Manganese/metabolism , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Protein Binding , Shear Strength , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
11.
Exp Dermatol ; 24(6): 443-8, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25807898

ABSTRACT

EMMPRIN is known to promote tumor invasion through extracellular matrix (ECM) degradation. Here we report that EMMPRIN can regulate melanoma cell adhesion to the ECM through an interaction with ß1 integrin involving kindlin-3. In this study, EMMPRIN knockdown in the human melanoma cell line M10 using siRNA decreased cell invasion and significantly increased cell adhesion and spreading. A morphological change from a round to a spread shape was observed associated with enhanced phalloidin-labelled actin staining. In situ proximity ligation assay and co-immunoprecipitation revealed that EMMPRIN silencing increased the interaction of ß1 integrin with kindlin-3, a focal adhesion protein. This was associated with an increase in ß1 integrin activation and a decrease in the phosphorylation of the downstream integrin kinase FAK. Moreover, the expression at both the transcript and protein level of kindlin-3 and of ß1 integrin was inversely regulated by EMMPRIN. EMMPRIN did not regulate either talin expression or its interaction with ß1 integrin. These results are consistent with our in vivo demonstration that EMMPRIN inhibition increased ß1 integrin activation and its interaction with kindlin-3. To conclude, these findings reveal a new role of EMMPRIN in tumor cell migration through ß1 integrin/kindlin-3-mediated adhesion pathway.


Subject(s)
Basigin/physiology , Cell Adhesion/physiology , Integrin beta1/physiology , Melanoma/pathology , Membrane Proteins/physiology , Neoplasm Proteins/physiology , Skin Neoplasms/pathology , Animals , Basigin/drug effects , Basigin/genetics , Cell Line, Tumor , Cell Shape/physiology , Extracellular Matrix/physiology , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Heterografts , Humans , In Vitro Techniques , Melanoma/physiopathology , Mice , Mice, Nude , RNA, Small Interfering/pharmacology , Signal Transduction/physiology , Skin Neoplasms/physiopathology
12.
Arterioscler Thromb Vasc Biol ; 34(9): 1961-7, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24969775

ABSTRACT

OBJECTIVE: Kindlin-3 is a critical supporter of integrin function in platelets. Lack of expression of kindlin-3 protein in patients impairs integrin αIIbß3-mediated platelet aggregation. Although kindlin-3 has been categorized as an integrin-binding partner, the functional significance of the direct interaction of kindlin-3 with integrin αIIbß3 in platelets has not been established. Here, we evaluated the significance of the binding of kindlin-3 to integrin αIIbß3 in platelets in supporting integrin αIIbß3-mediated platelet functions. APPROACH AND RESULTS: We generated a strain of kindlin-3 knockin (K3KI) mice that express a kindlin-3 mutant that carries an integrin-interaction defective substitution. K3KI mice could survive normally and express integrin αIIbß3 on platelets similar to their wild-type counterparts. Functional analysis revealed that K3KI mice exhibited defective platelet function, including impaired integrin αIIbß3 activation, suppressed platelet spreading and platelet aggregation, prolonged tail bleeding time, and absence of platelet-mediated clot retraction. In addition, whole blood drawn from K3KI mice showed resistance to in vitro thrombus formation and, as a consequence, K3KI mice were protected from in vivo arterial thrombosis. CONCLUSIONS: These observations demonstrate that the direct binding of kindlin-3 to integrin αIIbß3 is involved in supporting integrin αIIbß3 activation and integrin αIIbß3-dependent responses of platelets and consequently contributes significantly to arterial thrombus formation.


Subject(s)
Blood Platelets/physiology , Carotid Artery Thrombosis/physiopathology , Cytoskeletal Proteins/physiology , Platelet Glycoprotein GPIIb-IIIa Complex/physiology , Amino Acid Substitution , Animals , Bleeding Time , Blood Platelets/ultrastructure , Carotid Artery Thrombosis/blood , Carotid Artery Thrombosis/chemically induced , Cell Shape , Chlorides/toxicity , Clot Retraction , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , Disease Models, Animal , Female , Ferric Compounds/toxicity , Gene Knock-In Techniques , Genes, Reporter , Male , Mice , Mice, Inbred C57BL , Microspheres , Platelet Activation , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Protein Interaction Mapping , Recombinant Fusion Proteins/metabolism
13.
Pediatr Blood Cancer ; 62(9): 1677-9, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25854317

ABSTRACT

Disabling mutations in integrin-mediated cell signaling have been a major focus of interest over the last decade for patients affected with leukocyte adhesion deficiency-III (LAD-III). In this study, we identified a new C>T point mutation in exon 13 in the FERMT3 gene in an infant diagnosed with LAD-III and showed that KINDLIN-3 expression is required for platelet aggregation and leukocyte function, but also osteoclast-mediated bone resorption. After allogeneic bone marrow transplant, all overt symptoms disappeared. This newly identified mutation along with its novel role in dysregulation of bone homeostasis extends our understanding of KINDLIN-3 in humans.


Subject(s)
Blood Platelets/physiology , Bone Resorption/genetics , Codon, Nonsense , Integrins/physiology , Leukocyte-Adhesion Deficiency Syndrome/genetics , Leukocytes/physiology , Membrane Proteins/genetics , Mutation, Missense , Neoplasm Proteins/genetics , Osteoclasts/physiology , Osteopetrosis/genetics , Point Mutation , Bone Marrow Transplantation , Bone Resorption/pathology , Cell Adhesion , Cell Nucleus/ultrastructure , Exons/genetics , Female , Hemorrhagic Disorders/genetics , Homeostasis , Humans , Infant, Newborn , Leukocyte-Adhesion Deficiency Syndrome/pathology , Leukocyte-Adhesion Deficiency Syndrome/therapy , Membrane Proteins/deficiency , Membrane Proteins/physiology , Neoplasm Proteins/deficiency , Neoplasm Proteins/physiology , Osteoclasts/ultrastructure , Osteopetrosis/pathology , Osteopetrosis/therapy , Platelet Aggregation/genetics , Remission Induction
14.
J Thromb Haemost ; 22(7): 2009-2017, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38266679

ABSTRACT

BACKGROUND: Kindlin-3 in platelets plays an essential role in supporting integrin αIIbß3 activation, platelet spreading, aggregation, and clot retraction by binding to the integrin ß3 cytoplasmic tail. However, the mechanism by which kindlin-3 mediates the crosstalk between integrin αIIbß3 and myosin in platelets remains unknown. OBJECTIVES: To examine the role of myosin light chain 6 (Myl6) in supporting integrin αIIbß3 activation in platelets. METHODS: Myl6fl/flPF4-Cre mice with a deficiency of Myl6 in the megakaryocyte lineage were generated, and integrin αIIbß3 activation in Myl6-deficient platelets was analyzed. RESULTS: We identified a novel kindlin-3 binding protein, Myl6, an essential light chain of myosin in platelets. Myl6fl/flPF4-Cre mice exhibited significant macrothrombocytopenia resulting from defective proplatelet formation. In the absence of Myl6, integrin αIIbß3 activation in platelets was significantly suppressed, and platelet aggregation was substantially impaired. Interestingly, the deficiency of Myl6 in platelets preferentially affected the binding of a multivalent ligand compared to a monovalent ligand to integrin αIIbß3 upon activation, indicating that Myl6 may contribute to the avidity modulation of integrin αIIbß3 by binding to kindlin-3. Furthermore, blood coagulation ability was impaired in Myl6fl/flPF4-Cre mice, and consistently, these mice exhibited defects in both hemostatic and thrombotic functions. CONCLUSION: In summary, these results suggest that Myl6, as a novel kindlin-3 binding partner, is required to support integrin αIIbß3 activation in platelets, which plays an important role in both hemostasis and thrombosis.


Subject(s)
Blood Platelets , Myosin Light Chains , Platelet Aggregation , Platelet Glycoprotein GPIIb-IIIa Complex , Protein Binding , Animals , Blood Platelets/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Myosin Light Chains/metabolism , Thrombocytopenia/blood , Platelet Activation , Mice, Knockout , Megakaryocytes/metabolism , Humans , Mice, Inbred C57BL , Mice , Signal Transduction , Thrombosis/metabolism , Thrombosis/blood , Thrombosis/genetics , Cytoskeletal Proteins
15.
Cell Rep ; 42(6): 112580, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37267105

ABSTRACT

Bidirectional control of integrin activation plays crucial roles in cell adhesive behaviors, but how integrins are specifically regulated by inside-out and outside-in signaling has not been fully understood. Here, we report distinct bidirectional regulation of major lymphocyte homing receptors LFA1 and α4ß7 in primary T cells. A small increase of Rap1 activation in L-selectin-mediated tether/rolling was boosted by the outside-in signaling from ICAM1-interacting LFA1 through subsecond, simultaneous activation of Rap1 GTPase and talin1, but not kindlin-3, resulting in increased capture and slowing. In contrast, none of them were required for tether/rolling by α4ß7 on MAdCAM1. High Rap1 activation with chemokines or the loss of Rap1-inactivating proteins Rasa3 and Sipa1 increased talin1/kindlin-3-dependent arrest with high-affinity binding of LFA1 to membrane-anchored ICAM1. However, despite increased affinity of α4ß7, activated Rap1 severely suppressed adhesion on MAdCAM1 under shear flow, indicating the critical importance of a sequential outside-in/inside-out signaling for α4ß7.


Subject(s)
Integrins , Lymphocyte Function-Associated Antigen-1 , T-Lymphocytes , Cell Adhesion/physiology , Chemokines/metabolism , Integrins/metabolism , Signal Transduction/physiology , T-Lymphocytes/metabolism
16.
Cells ; 11(11)2022 05 26.
Article in English | MEDLINE | ID: mdl-35681446

ABSTRACT

Integrin LFA1 is a cell adhesion receptor expressed exclusively in leukocytes, and plays crucial roles in lymphocyte trafficking, antigen recognition, and effector functions. Since the discovery that the adhesiveness of LFA1 can be dynamically changed upon stimulation, one challenge has been understanding how integrins are regulated by inside-out signaling coupled with macromolecular conformational changes, as well as ligand bindings that transduce signals from the extracellular domain to the cytoplasm in outside-in signaling. The small GTPase Rap1 and integrin adaptor proteins talin1 and kindlin-3 have been recognized as critical molecules for integrin activation. However, their cooperative regulation of integrin adhesiveness in lymphocytes requires further research. Recent advances in single-molecule imaging techniques have revealed dynamic molecular processes in real-time and provided insight into integrin activation in cellular environments. This review summarizes integrin regulation and discusses new findings regarding the bidirectionality of LFA1 activation and signaling processes in lymphocytes.


Subject(s)
Adaptor Proteins, Signal Transducing , Integrins , Adaptor Proteins, Signal Transducing/metabolism , Cell Adhesion , Integrins/metabolism , Leukocytes/metabolism , Signal Transduction/physiology
17.
Anticancer Res ; 42(3): 1217-1220, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35220211

ABSTRACT

BACKGROUND/AIM: Kindlins are essential integrin activators. Kindlin-1 and kindlin-2 are often concomitantly expressed in epithelial tumor cells and participate in regulating tumor malignancy. However, it remains unclear whether kindlin-3, the one expressed in immune cells, also plays a role in regulating tumor malignancy. MATERIALS AND METHODS: To examine the role of kindlin-3 in different immune cells in regulating solid tumor growth, a xenograft model of prostate cancer tumor growth in genetically modified kindlin-3 mice was employed. RESULTS: Disruption of crosstalk between kindlin-3 and integrins significantly promoted subcutaneous prostate cancer tumor growth in mice. Furthermore, deficiency of kindlin-3 in T cells and NK cells, but not myeloid cells and B cells, significantly enhanced prostate cancer tumor growth. CONCLUSION: Tumor-killing leukocytes require Kindlin-3 for suppressing cancerous tumor growth, thus providing a novel anticancer mechanism.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Cytoskeletal Proteins/metabolism , Killer Cells, Natural/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , Prostatic Neoplasms/metabolism , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell Proliferation , Cytoskeletal Proteins/genetics , Killer Cells, Natural/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Signal Transduction , Tumor Burden , Tumor Microenvironment
18.
Cells ; 11(9)2022 05 03.
Article in English | MEDLINE | ID: mdl-35563841

ABSTRACT

The use of cell-based reporter systems has provided valuable insights into the molecular mechanisms of integrin activation. However, current models have significant drawbacks because their artificially expressed integrins cannot be regulated by either physiological stimuli or endogenous signaling pathways. Here, we report the generation of a Hoxb8 cell line expressing human ß2 integrin that functionally replaced the deleted mouse ortholog. Hoxb8 cells are murine hematopoietic progenitor cells that can be efficiently differentiated into neutrophils and macrophages resembling their primary counterparts. Importantly, these cells can be stimulated by physiological stimuli triggering classical integrin inside-out signaling pathways, ultimately leading to ß2 integrin conformational changes that can be recorded by the conformation-specific antibodies KIM127 and mAb24. Moreover, these cells can be efficiently manipulated via the CRISPR/Cas9 technique or retroviral vector systems. Deletion of the key integrin regulators talin1 and kindlin3 or expression of ß2 integrins with mutations in their binding sites abolished both integrin extension and full activation regardless of whether only one or both activators no longer bind to the integrin. Moreover, humanized ß2 integrin Hoxb8 cells represent a valuable new model for rapidly testing the role of putative integrin regulators in controlling ß2 integrin activity in a physiological context.


Subject(s)
CD18 Antigens , Integrins , Animals , CD18 Antigens/metabolism , Homeodomain Proteins/metabolism , Integrins/metabolism , Mice , Neutrophils/metabolism , Signal Transduction/genetics
19.
Front Immunol ; 12: 792813, 2021.
Article in English | MEDLINE | ID: mdl-35154074

ABSTRACT

Integrins in effector T cells are crucial for cell adhesion and play a central role in cell-mediated immunity. Leukocyte adhesion deficiency (LAD) type III, a genetic condition that can cause death in early childhood, highlights the importance of integrin/kindlin interactions for immune system function. A TTT/AAA mutation in the cytoplasmic domain of the ß2 integrin significantly reduces kindlin-3 binding to the ß2 tail, abolishes leukocyte adhesion to intercellular adhesion molecule 1 (ICAM-1), and decreases T cell trafficking in vivo. However, how kindlin-3 affects integrin function in T cells remains incompletely understood. We present an examination of LFA-1/ICAM-1 bonds in both wild-type effector T cells and those with a kindlin-3 binding site mutation. Adhesion assays show that effector T cells carrying the kindlin-3 binding site mutation display significantly reduced adhesion to the integrin ligand ICAM-1. Using optical trapping, combined with back focal plane interferometry, we measured a bond rupture force of 17.85 ±0.63 pN at a force loading rate of 30.21 ± 4.35 pN/s, for single integrins expressed on wild-type cells. Interestingly, a significant drop in rupture force of bonds was found for TTT/AAA-mutant cells, with a measured rupture force of 10.08 ± 0.88pN at the same pulling rate. Therefore, kindlin-3 binding to the cytoplasmic tail of the ß2-tail directly affects catch bond formation and bond strength of integrin-ligand bonds. As a consequence of this reduced binding, CD8+ T cell activation in vitro is also significantly reduced.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Adhesion/immunology , Cytoskeletal Proteins/genetics , Intercellular Adhesion Molecule-1/metabolism , Lymphocyte Function-Associated Antigen-1/metabolism , Animals , Binding Sites , CD18 Antigens/immunology , CD18 Antigens/metabolism , Cytoskeletal Proteins/immunology , Cytoskeletal Proteins/metabolism , Intercellular Adhesion Molecule-1/immunology , Lymphocyte Activation/immunology , Lymphocyte Function-Associated Antigen-1/immunology , Mice , Mutation , Optical Tweezers
20.
J Thromb Haemost ; 19(12): 3126-3138, 2021 12.
Article in English | MEDLINE | ID: mdl-34411430

ABSTRACT

BACKGROUND: Kindlin-3 is essential for supporting the bidirectional signaling of integrin αIIbß3 in platelets by bridging the crosstalk between integrin αIIbß3 and the cytoplasmic signaling adaptors. OBJECTIVE: In this study, we identified a previously unrecognized paxillin binding site in the pleckstrin homology (PH) domain of kindlin-3 and verified its functional significance. METHODS: Structure-based approaches were employed to identify the paxillin binding site in the PH domain of kindlin-3. In addition, the bidirectional signaling of integrin αIIbß3 were evaluated in both human and mouse platelets. RESULTS: In brief, we found that a ß1-ß2 loop in the PH domain of kindlin-3, an important part of the canonical membrane phospholipid binding pocket, was also involved in mediating paxillin interaction. Interestingly, the binding sites of paxillin and membrane phospholipids in the PH domain of kindlin-3 were mutually exclusive. Specific disruption of paxillin binding to the PH domain by point mutations inhibited platelet spreading on immobilized fibrinogen while having no inhibition on soluble fibrinogen binding to stimulated platelets. In addition, a membrane-permeable peptide derived from the ß1-ß2 loop in the PH domain of kindlin-3 was capable of inhibiting platelet spreading and clot retraction, but it had no effect on soluble fibrinogen binding to platelets and platelet aggregation. Treatment with this peptide significantly reduced thrombus formation in mice. CONCLUSION: Taken together, these findings suggest that interaction between paxillin and the PH domain of kindlin-3 plays an important role in supporting integrin αIIbß3 outside-in signaling in platelets, thus providing a novel antithrombotic target.


Subject(s)
Blood Platelets , Platelet Glycoprotein GPIIb-IIIa Complex , Animals , Blood Platelets/metabolism , Clot Retraction , Cytoskeletal Proteins , Mice , Paxillin , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Pleckstrin Homology Domains
SELECTION OF CITATIONS
SEARCH DETAIL