Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Biol Chem ; 300(2): 105601, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38159860

ABSTRACT

Hepatocyte plays a principal role in preserving integrity of the liver homeostasis. Our recent study demonstrated that Kindlin-2, a focal adhesion protein that activates integrins and regulates cell-extracellular matrix interactions, plays an important role in regulation of liver homeostasis by inhibiting inflammation pathway; however, the molecular mechanism of how Kindlin-2 KO activates inflammation is unknown. Here, we show that Kindlin-2 loss largely downregulates the antioxidant glutathione-S-transferase P1 in hepatocytes by promoting its ubiquitination and degradation via a mechanism involving protein-protein interaction. This causes overproduction of intracellular reactive oxygen species and excessive oxidative stress in hepatocytes. Kindlin-2 loss upregulates osteopontin in hepatocytes partially because of upregulation of reactive oxygen species and consequently stimulates overproduction of inflammatory cytokines and infiltration in liver. The molecular and histological deteriorations caused by Kindlin-2 deficiency are markedly reversed by systemic administration of an antioxidant N-acetylcysteine in mice. Taken together, Kindlin-2 plays a pivotal role in preserving integrity of liver function.


Subject(s)
Cytoskeletal Proteins , Inflammation , Membrane Proteins , Oxidative Stress , Animals , Mice , Antioxidants/metabolism , Homeostasis , Inflammation/metabolism , Liver/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Reactive Oxygen Species/metabolism , Cytoskeletal Proteins/metabolism
2.
Exp Cell Res ; 436(2): 113974, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38346630

ABSTRACT

The extracellular matrix (ECM) mechanical properties regulate biological processes, such as fibroblast-myofibroblast transformation (FMT), which is a crucial component in pelvic organ prolapse (POP) development. The 'Kindlin-2' protein, expressed by fibroblasts, plays an important role in the development of the mesoderm, which is responsible for connective tissue formation; however, the role of Kindlin-2 in FMT remains to be explored. In this study, we aimed to explore the role of Kindlin-2 in FMT as it relates to POP. We found that ECM stiffness induces autophagy to translocate Kindlin-2 to the cytoplasm of L929 cells, where it interacts with and degrades MOB1, thereby facilitating Yes-associated protein (YAP) entry into the nucleus and influencing FMT progression. Stiffness-induced autophagy was inhibited when using an autophagy inhibitor, which blocked the translocation of Kindlin-2 to the cytoplasm and partially reversed high-stiffness-induced FMT. In patients with POP, we observed an increase in cytoplasmic Kindlin-2 and nuclear YAP levels. Similar changes in vaginal wall-associated proteins were observed in a mouse model of acute vaginal injury. In conclusion, Kindlin-2 is a key gene affecting ECM stiffness, which regulates FMT by inducing autophagy and may influence the development of POP.


Subject(s)
Cytoskeletal Proteins , Extracellular Matrix , Muscle Proteins , Myofibroblasts , Animals , Female , Humans , Mice , Cytoplasm/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/metabolism , Fibroblasts/metabolism , Myofibroblasts/metabolism , Cytoskeletal Proteins/metabolism , Muscle Proteins/metabolism
3.
J Biol Chem ; 299(6): 104774, 2023 06.
Article in English | MEDLINE | ID: mdl-37142218

ABSTRACT

Mitochondria are signaling organelles implicated in cancer, but the mechanisms are elusive. Here, we show that Parkin, an E3 ubiquitination (Ub) ligase altered in Parkinson's disease, forms a complex with the regulator of cell motility, Kindlin-2 (K2), at mitochondria of tumor cells. In turn, Parkin ubiquitinates Lys581 and Lys582 using Lys48 linkages, resulting in proteasomal degradation of K2 and shortened half-life from ∼5 h to ∼1.5 h. Loss of K2 inhibits focal adhesion turnover and ß1 integrin activation, impairs membrane lamellipodia size and frequency, and inhibits mitochondrial dynamics, altogether suppressing tumor cell-extracellular matrix interactions, migration, and invasion. Conversely, Parkin does not affect tumor cell proliferation, cell cycle transitions, or apoptosis. Expression of a Parkin Ub-resistant K2 Lys581Ala/Lys582Ala double mutant is sufficient to restore membrane lamellipodia dynamics, correct mitochondrial fusion/fission, and preserve single-cell migration and invasion. In a 3D model of mammary gland developmental morphogenesis, impaired K2 Ub drives multiple oncogenic traits of EMT, increased cell proliferation, reduced apoptosis, and disrupted basal-apical polarity. Therefore, deregulated K2 is a potent oncogene, and its Ub by Parkin enables mitochondria-associated metastasis suppression.


Subject(s)
Membrane Proteins , Ubiquitin-Protein Ligases , Cell Movement , Membrane Proteins/metabolism , Mitochondria/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Humans
4.
Development ; 148(10)2021 05 15.
Article in English | MEDLINE | ID: mdl-33999995

ABSTRACT

The focal adhesion protein Kindlin2 is essential for integrin activation, a process that is fundamental to cell-extracellular matrix adhesion. Kindlin 2 (Fermt2) is widely expressed in mouse embryos, and its absence causes lethality at the peri-implantation stage due to the failure to trigger integrin activation. The function of kindlin2 during embryogenesis has not yet been fully elucidated as a result of this early embryonic lethality. Here, we showed that kindlin2 is essential for neural crest (NC) formation in Xenopus embryos. Loss-of-function assays performed with kindlin2-specific morpholino antisense oligos (MOs) or with CRISPR/Cas9 techniques in Xenopus embryos severely inhibit the specification of the NC. Moreover, integrin-binding-deficient mutants of Kindlin2 rescued the phenotype caused by loss of kindlin2, suggesting that the function of kindlin2 during NC specification is independent of integrins. Mechanistically, we found that Kindlin2 regulates the fibroblast growth factor (FGF) pathway, and promotes the stability of FGF receptor 1. Our study reveals a novel function of Kindlin2 in regulating the FGF signaling pathway and provides mechanistic insights into the function of Kindlin2 during NC specification.


Subject(s)
Fibroblast Growth Factors/metabolism , Membrane Proteins/metabolism , Neural Crest/embryology , Xenopus Proteins/metabolism , Xenopus laevis/embryology , Animals , CRISPR-Cas Systems/genetics , Cell Line , Embryo, Nonmammalian/metabolism , Embryonic Development/genetics , Embryonic Development/physiology , Gene Expression Regulation, Developmental/genetics , Gene Knockout Techniques , HEK293 Cells , HeLa Cells , Humans , Integrins/metabolism , Membrane Proteins/genetics , Morpholinos/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Signal Transduction/genetics , Xenopus Proteins/genetics
5.
Cell Mol Life Sci ; 80(8): 223, 2023 Jul 22.
Article in English | MEDLINE | ID: mdl-37480504

ABSTRACT

Kindlin-2 is critical for development and homeostasis of key organs, including skeleton, liver, islet, etc., yet its role in modulating angiogenesis is unknown. Here, we report that sufficient KINDLIN-2 is extremely important for NOTCH-mediated physiological angiogenesis. The expression of KINDLIN-2 in HUVECs is significantly modulated by angiogenic factors such as vascular endothelial growth factor A or tumor necrosis factor α. A strong co-localization of CD31 and Kindlin-2 in tissue sections is demonstrated by immunofluorescence staining. Endothelial-cell-specific Kindlin-2 deletion embryos die on E10.5 due to hemorrhage caused by the impaired physiological angiogenesis. Experiments in vitro show that vascular endothelial growth factor A-induced multiple functions of endothelial cells, including migration, matrix proteolysis, morphogenesis and sprouting, are all strengthened by KINDLIN-2 overexpression and severely impaired in the absence of KINDLIN-2. Mechanistically, we demonstrate that KINDLIN-2 inhibits the release of Notch intracellular domain through binding to and maintaining the integrity of NOTCH1. The impaired angiogenesis and avascular retinas caused by KINDLIN-2 deficiency can be rescued by DAPT, an inhibitor of γ-secretase which releases the intracellular domain from NOTCH1. Moreover, we demonstrate that high glucose stimulated hyperactive angiogenesis by increasing KINDLIN-2 expression could be prevented by KINDLIN-2 knockdown, indicating Kindlin-2 as a potential therapeutic target in treatment of diabetic retinopathy. Our study for the first time demonstrates the significance of Kindlin-2 in determining Notch-mediated angiogenesis during development and highlights Kindlin-2 as the potential therapeutic target in angiogenic diseases, such as diabetic retinopathy.


Subject(s)
Diabetic Retinopathy , Humans , Cardiovascular Physiological Phenomena , Endothelial Cells , Morphogenesis , Vascular Endothelial Growth Factor A/genetics
6.
J Cell Sci ; 134(1)2021 01 13.
Article in English | MEDLINE | ID: mdl-33277381

ABSTRACT

Cell migration involves front-to-rear asymmetric focal adhesion (FA) dynamics, which facilitates trailing edge detachment and directional persistence. Here, we show that kindlin-2 is crucial for FA sliding and disassembly in migrating cells. Loss of kindlin-2 markedly reduced FA number and selectively impaired rear FA sliding and disassembly, resulting in defective rear retraction and reduced directional persistence during cell migration. Kindlin-2-deficient cells failed to develop serum-induced actomyosin-dependent tension at FAs. At the molecular level, kindlin-2 directly interacted with myosin light chain kinase (MYLK, hereafter referred to as MLCK), which was enhanced in response to serum stimulation. Serum deprivation inhibited rear FA disassembly, which was released in response to serum stimulation. Overexpression of the MLCK-binding kindlin-2 F0F1 fragment (amino acid residues 1-167), which inhibits the interaction of endogenous kindlin-2 with MLCK, phenocopied kindlin-2 deficiency-induced migration defects. Inhibition of MLCK, like loss of kindlin-2, also impaired trailing-edge detachment, rear FA disassembly and directional persistence. These results suggest a role of kindlin-2 in promoting actomyosin contractility at FAs, leading to increased rear FA sliding and disassembly, and directional persistence during cell migration.


Subject(s)
Focal Adhesions , Myosin-Light-Chain Kinase , Cell Adhesion , Cell Movement/genetics , Focal Adhesions/metabolism , Myosin-Light-Chain Kinase/metabolism , Phosphorylation
7.
Biochem Biophys Res Commun ; 614: 1-8, 2022 07 23.
Article in English | MEDLINE | ID: mdl-35567938

ABSTRACT

Diabetes mellitus has been a major public health problem worldwide, characterized by insulin resistance and dysfunction of ß-cells. A previous study showed that Kindlin-2 loss in ß-cells dramatically reduces insulin secretion and decreases ß-cell mass, resulting in severe diabetes-like phenotypes. It suggests that Kindlin-2 in ß-cells play an important role in regulating glucose homeostasis. However, the effect of Kindlin-2 on the function of ß-cells under chronic hyperglycemia in diabetes has not been explored. Here we report that Kindlin-2 overexpression ameliorates diabetes and improves insulin secretion in mice induced by streptozocin. In contrast, Kindlin-2 insufficiency exacerbates diabetes and promotes ß-cells dysfunction and inflammation in ß-cells induced by a high-fat diet (HFD). In vitro, Kindlin-2 overexpression prevented high-glucose (HG)-induced dysfunction in ß-cells. Kindlin-2 overexpression also decreased the expression of pro-inflammatory cytokines and NLRP3 inflammasome expression in ß-cells exposed to HG. Furthermore, the loss of Kindlin-2 aggravates the expression of inflammatory cytokines and NLRP3 induced by HG in ß-cells. Collectively, we demonstrate that Kindlin-2 protects against diabetes by inhibiting NLRP3 inflammasome activation.


Subject(s)
Cytoskeletal Proteins , Diabetes Mellitus, Experimental , Inflammasomes , Insulin-Secreting Cells , Animals , Cytokines/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Diabetes Mellitus, Experimental/metabolism , Glucose/metabolism , Inflammasomes/metabolism , Insulin-Secreting Cells/metabolism , Mice , Mice, Inbred C57BL , Muscle Proteins/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
8.
Exp Cell Res ; 408(1): 112813, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34492266

ABSTRACT

Keloids are benign skin tumors characterized by aggressive growth. To date, there is no exact treatment because little is known about its pathological mechanism. Therefore, it is important to investigate the mechanism of its occurrence and development to identify therapeutic targets. In this study, the expression of Kindlin-2 was higher in keloid fibroblasts (KFs) than in normal skin fibroblasts (NFs). In vitro experiments showed that knocking down Kindlin-2 in KFs could promote cell apoptosis and inhibit cell proliferation, cell migration and invasion, and contractile capability. Western blot results showed that the phosphorylation of Smad3 in KFs was inhibited after knocking down Kindlin-2, inhibiting the activation of the Smad pathway. Moreover, knocking down Kindlin-2 increased the expression of Fas and FasL in KFs, which demonstrated that knocking down Kindlin-2 promoted the activation of the exogenous apoptotic pathway of KFs and then facilitated apoptosis. The above results revealed that knocking down Kindlin-2 in KFs can inhibit the activation of the Smad pathway and promote the activation of the Fas/FasL exogenous apoptosis pathway, thereby altering the cytological function of KFs. Therefore, Kindlin-2 might play an important role in the occurrence and development of keloids and could become a new target to treat keloids.


Subject(s)
Cell Movement/physiology , Fibroblasts/metabolism , Keloid/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Adult , Cell Proliferation/physiology , Cells, Cultured , Extracellular Matrix/metabolism , Fas Ligand Protein/metabolism , Female , Fibroblasts/pathology , Humans , Keloid/pathology , Male , Signal Transduction/physiology , Transforming Growth Factor beta1/metabolism
9.
Beijing Da Xue Xue Bao Yi Xue Ban ; 54(5): 846-852, 2022 Oct 18.
Article in Zh | MEDLINE | ID: mdl-36241227

ABSTRACT

OBJECTIVE: To investigate the effects and mechanisms of Kindlin-2 on uterus development and reproductive capacity in female mice. METHODS: Cdh16-Cre tool mice and Kindlin-2flox/flox mice were used to construct the mouse model of uterus specific knockout of Kindlin-2, and the effects of Kindlin-2 deletion on uterine development and reproduction capacity of female mice were observed. High expression and knockdown of Kindlin-2 in endometrial cancer cell lines HEC-1 and Ish were used to detect the regulation of mammalian target of rapamycin (mTOR) signaling pathway. In addition, uterine proteins of the female mice with specific knockout of Kindlin-2 and female mice in the control group were extracted to detect the protein levels of key molecules of mTOR signaling pathway and Hippo signaling pathway. RESULTS: The mouse model of uterine specific knockout of Kindlin-2 was successfully constructed. The knockout efficiency of Kindlin-2 in mouse uterus was identified and verified by mouse tail polymerase chain reaction (PCR), Western blot protein identification, immunohistochemical staining (IHC) and other methods. Compared with the control group, the female mice with uterus specific deletion of Kindlin-2 lost weight, seriously impaired reproductive ability, and the number of newborn mice decreased, but the proportion of the female mice and male mice in the newborn mice did not change. Hematoxylin eosin staining (HE) experiment showed that the endometrium of Kindlin-2 knockout group was incomplete and the thickness of uterine wall became thinner. In terms of mechanism, the deletion of Kindlin-2 in endo-metrial cancer cell lines HEC-1 and Ish could downregulate the protein levels of mTOR, phosphorylated mTOR, adenosine monophosphate-activated protein kinase (AMPK), phosphorylated AMPK and phosphorylated ribosomal protein S6 (S6), and the mTOR signal pathway was inhibited. It was found that the specific deletion of Kindlin-2 could upregulate the protein levels of Mps one binding 1 (MOB1) and phosphorylated Yes-associated protein (YAP) in the uterus of the female mice, and the Hippo signal pathway was activated. CONCLUSION: Kindlin-2 inhibits the development of uterus by inhibiting mTOR signal pathway and activating Hippo signal pathway, thereby inhibiting the fertility of female mice.


Subject(s)
AMP-Activated Protein Kinases , Hippo Signaling Pathway , AMP-Activated Protein Kinases/metabolism , Adenosine Monophosphate/metabolism , Animals , Cadherins/metabolism , Cytoskeletal Proteins/metabolism , Endometrium/metabolism , Eosine Yellowish-(YS)/metabolism , Female , Hematoxylin/metabolism , Male , Mammals/metabolism , Mice , Muscle Proteins , Ribosomal Protein S6/metabolism , Sirolimus/metabolism , TOR Serine-Threonine Kinases/metabolism , YAP-Signaling Proteins
10.
Am J Respir Cell Mol Biol ; 65(1): 54-69, 2021 07.
Article in English | MEDLINE | ID: mdl-33761308

ABSTRACT

Pulmonary fibrosis is a progressive and fatal lung disease characterized by activation of lung fibroblasts and excessive deposition of collagen matrix. We show here that the concentrations of kindlin-2 and its binding partner PYCR1, a key enzyme for proline synthesis, are significantly increased in the lung tissues of human patients with pulmonary fibrosis. Treatment of human lung fibroblasts with TGF-ß1 markedly increased the expression of kindlin-2 and PYCR1, resulting in increased kindlin-2 mitochondrial translocation, formation of the kindlin-2-PYCR1 complex, and proline synthesis. The concentrations of the kindlin-2-PYCR1 complex and proline synthesis were markedly reduced in response to pirfenidone or nintedanib, two clinically approved therapeutic drugs for pulmonary fibrosis. Furthermore, depletion of kindlin-2 alone was sufficient to suppress TGF-ß1-induced increases of PYCR1 expression, proline synthesis, and fibroblast activation. Finally, using a bleomycin mouse model of pulmonary fibrosis, we show that ablation of kindlin-2 effectively reduced the concentrations of PYCR1, proline, and collagen matrix and alleviate the progression of pulmonary fibrosis in vivo. Our results suggest that kindlin-2 is a key promoter of lung fibroblast activation, collagen matrix synthesis, and pulmonary fibrosis, underscoring the therapeutic potential of targeting the kindlin-2 signaling pathway for control of this deadly lung disease.


Subject(s)
Cytoskeletal Proteins/metabolism , Fibroblasts/metabolism , Lung/metabolism , Muscle Proteins/metabolism , Pulmonary Fibrosis/metabolism , Signal Transduction , Animals , Bleomycin/adverse effects , Bleomycin/pharmacology , Cytoskeletal Proteins/genetics , Disease Models, Animal , Fibroblasts/pathology , Humans , Lung/pathology , Mice , Mice, Transgenic , Muscle Proteins/genetics , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology
11.
Amino Acids ; 53(12): 1835-1840, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34291342

ABSTRACT

Δ1-Pyrroline-5-carboxylate (P5C) reductase (PYCR or P5CR) catalyzes the conversion of P5C to L-proline (Pro) with concomitant oxidation of a cofactor, NADPH or NADH. Mammalian PYCR have been studied since 1950' and currently three isozymes of human PYCR, 1, 2, and L, have been identified and characterized and their roles in genetic diseases and cancer biology have been keenly investigated. These three isozymes are encoded by three different genes localized at three different chromosomes, and catalyze NAD(P)H-dependent reduction of P5C to Pro important for the transfer of oxidizing potential across the mitochondrion and cell. The review summarizes the current understanding of these three human PYCR isozymes and their roles in diseases with a focus on cancer.


Subject(s)
Isoenzymes/metabolism , Neoplasms/metabolism , Pyrroline Carboxylate Reductases/metabolism , Amino Acid Sequence , Animals , Humans , Mitochondria/metabolism , NAD/metabolism , NADP/metabolism , delta-1-Pyrroline-5-Carboxylate Reductase
12.
Int J Mol Sci ; 22(19)2021 Sep 30.
Article in English | MEDLINE | ID: mdl-34638957

ABSTRACT

The tumor microenvironment plays a critical role in defining the growth and malignancy of solid tumors. Extracellular matrix (ECM) proteins such as collagen, vitronectin, and fibronectin are major components of the tumor microenvironment. Tumor growth-promoting reciprocal interaction between ECM and cytoplasmic proteins is regulated by the cell surface receptors called integrins. This study investigated the mechanism by which integrin ß1 promotes pancreatic tumor growth. In MIA PaCa-2 pancreatic cancer cell line, the loss of integrin ß1 protein reduced the ability of cells to proliferate in a 3D matrix and compromised the ability to form a focal adhesion complex. Decreased expression of integrin α5 was observed in KO cells, which resulted in impaired cell spreading and adhesion on vitronectin and fibronectin. Reduced expression of the integrin-associated protein, kindlin-2 was also recorded. The downregulation of kindlin-2 decreased the phosphorylation of Smad2/3 by reducing the expression of TGF-ß receptor 2. These results unravel a new mechanism of integrin ß1 in tumor growth by modifying the expression of kindlin-2 and TGF-ß receptor 2 signaling.


Subject(s)
Carcinogenesis/metabolism , Cell Proliferation/genetics , Integrin beta1/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/metabolism , Receptor, Transforming Growth Factor-beta Type II/metabolism , Signal Transduction/genetics , Carcinogenesis/genetics , Cell Adhesion/genetics , Cell Line, Tumor , Cell Movement/genetics , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Humans , Integrin beta1/genetics , Pancreatic Neoplasms/pathology , Transfection
13.
J Cell Mol Med ; 24(22): 13081-13092, 2020 11.
Article in English | MEDLINE | ID: mdl-32954626

ABSTRACT

Liver steatosis is associated with increased ischaemia reperfusion (I/R) injury. Our previous studies have shown that irisin, an exercise-induced hormone, mitigates I/R injury via binding to αVß5 integrin. However, the effect of irisin on I/R injury in steatotic liver remains unknown. Kindlin-2 directly interacts with ß integrin. We therefore suggest that irisin protects against I/R injury in steatotic liver via a kindlin-2 dependent mechanism. To study this, hepatic steatosis was induced in male adult mice by feeding them with a 60% high-fat diet (HFD). At 12 weeks after HFD feeding, the mice were subjected to liver ischaemia by occluding partial (70%) hepatic arterial/portal venous blood for 60 minutes, which was followed by 24 hours reperfusion. Our results showed HFD exaggerated I/R-induced liver injury. Irisin (250 µg/kg) administration at the beginning of reperfusion attenuated liver injury, improved mitochondrial function, and reduced oxidative and endoplasmic reticulum stress in HFD-fed mice. However, kindlin-2 inhibition by RNAi eliminated irisin's direct effects on cultured hepatocytes. In conclusion, irisin attenuates I/R injury in steatotic liver via a kindlin-2 dependent mechanism.


Subject(s)
Cytoskeletal Proteins/metabolism , Fibronectins/metabolism , Liver Diseases/metabolism , Muscle Proteins/metabolism , Reperfusion Injury/metabolism , Animals , Diet, High-Fat , Disease Models, Animal , Endoplasmic Reticulum Stress , Fatty Liver/metabolism , Hepatocytes/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , RNA Interference , RNA, Small Interfering/metabolism
14.
J Cell Sci ; 131(20)2018 10 26.
Article in English | MEDLINE | ID: mdl-30254023

ABSTRACT

The integrin-associated adaptor proteins integrin-linked kinase (ILK) and kindlin-2 play central roles in integrin signaling and control of cell morphology. A direct ILK-kindlin-2 interaction is conserved across species and involves the F2PH subdomain of kindlin-2 and the pseudokinase domain (pKD) of ILK. However, complete understanding of the ILK-kindlin-2 interaction and its role in integrin-mediated signaling has been impeded by difficulties identifying the binding site for kindlin-2 on ILK. We used conservation-guided mapping to dissect the interaction between ILK and kindlin-2 and identified a previously unknown binding site for kindlin-2 on the C-lobe of the pKD of ILK. Mutations at this site inhibit binding to kindlin-2 while maintaining structural integrity of the pKD. Importantly, kindlin-binding-defective ILK mutants exhibit impaired focal adhesion localization and fail to fully rescue the spreading defects seen in ILK knockdown cells. Furthermore, kindlin-2 mutants with impaired ILK binding are also unable to fully support cell spreading. Thus, the interaction between ILK and kindlin-2 is critical for cell spreading and focal adhesion localization, representing a key signaling axis downstream of integrins.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Focal Adhesions/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/genetics , Protein Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Cell Adhesion , Humans , Membrane Proteins/genetics , Signal Transduction
15.
Chin J Cancer Res ; 32(1): 72-88, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32194307

ABSTRACT

OBJECTIVE: Recent studies have shown that tumor-associated macrophages (TAMs) play an important role in cancer invasion and metastasis. Our previous studies have reported that TAMs promote the invasion and metastasis of gastric cancer (GC) cells through the Kindlin-2 pathway. However, the mechanism needs to be clarified. METHODS: THP-1 monocytes were induced by PMA/interleukin (IL)-4/IL-13 to establish an efficient TAM model in vitro and M2 macrophages were isolated via flow cytometry. A dual luciferase reporter system and chromatin immunoprecipitation (ChIP) assay were used to investigate the mechanism of transforming growth factor ß2 (TGFß2) regulating Kindlin-2 expression. Immunohistochemistry was used to study the relationships among TAM infiltration in human GC tissues, Kindlin-2 protein expression, clinicopathological parameters and prognosis in human GC tissues. A nude mouse oncogenesis model was used to verify the invasion and metastasis mechanisms in vivo. RESULTS: We found that Kindlin-2 expression was upregulated at both mRNA and protein levels in GC cells cocultured with TAMs, associated with higher invasion rate. Kindlin-2 knockdown reduced the invasion rate of GC cells under coculture condition. TGFß2 secreted by TAMs regulated the expression of Kindlin-2 through the transcription factor NF-кB. TAMs thus participated in the progression of GC through the TGFß2/NF-κB/Kindlin-2 axis. Kindlin-2 expression and TAM infiltration were significantly positively correlated with TNM stage, and patients with high Kindlin-2 expression had significantly poorer overall survival than patients with low Kindlin-2 expression. Furthermore, Kindlin-2 promoted the invasion of GC cells in vivo. CONCLUSIONS: This study elucidates the mechanism of TAMs participating in GC cell invasion and metastasis through the TGFß2/NF-κB/Kindlin-2 axis, providing a possibility for new treatment options and approaches.

16.
J Cell Sci ; 130(20): 3532-3541, 2017 Oct 15.
Article in English | MEDLINE | ID: mdl-28864764

ABSTRACT

The integrin αVß3 is reported to promote angiogenesis in some model systems but not in others. Here, we used optogenetics to study the effects of αVß3 interaction with the intracellular adapter kindlin-2 (Fermt2) on endothelial cell functions potentially relevant to angiogenesis. Because interaction of kindlin-2 with αVß3 requires the C-terminal three residues of the ß3 cytoplasmic tail (Arg-Gly-Thr; RGT), optogenetic probes LOVpep and ePDZ1 were fused to ß3ΔRGT-GFP and mCherry-kindlin-2, respectively, and expressed in ß3 integrin-null microvascular endothelial cells. Exposure of the cells to 450 nm (blue) light caused rapid and specific interaction of kindlin-2 with αVß3 as assessed by immunofluorescence and total internal reflection fluorescence (TIRF) microscopy, and it led to increased endothelial cell migration, podosome formation and angiogenic sprouting. Analyses of kindlin-2 mutants indicated that interaction of kindlin-2 with other kindlin-2 binding partners, including c-Src, actin, integrin-linked kinase and phosphoinositides, were also likely necessary for these endothelial cell responses. Thus, kindlin-2 promotes αVß3-dependent angiogenic functions of endothelial cells through its simultaneous interactions with ß3 integrin and several other binding partners. Optogenetic approaches should find further use in clarifying spatiotemporal aspects of vascular cell biology.


Subject(s)
Endothelial Cells/physiology , Integrin alphaVbeta3/genetics , Animals , Cell Adhesion , Cell Movement , Cells, Cultured , Cytoskeletal Proteins/metabolism , Fibrinogen/metabolism , Gene Expression , Integrin alphaVbeta3/metabolism , Mice, Inbred C57BL , Muscle Proteins/metabolism , Neovascularization, Physiologic , Optogenetics , Protein Binding , Talin/metabolism
17.
BMC Dev Biol ; 18(1): 19, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30382829

ABSTRACT

BACKGROUND: Integrins are transmembrane receptors that mediate cell-extracellular matrix (ECM) and cell-cell adhesion and trophoblast cells undergo changes in integrin expression as they differentiate. However, the mechanism(s) of integrin activation leading to integrin-mediated signaling in trophoblast cell differentiation is unknown. The Fermitin family proteins are integrin activators that help mediate integrin-mediated signaling, but have never been studied in detail within the human placenta. Thus, we examined the spatiotemporal pattern of expression of Fermitin family homolog-2 (FERMT2) in human chorionic villi throughout gestation and its role in trophoblast-substrate adhesion and invasion. METHODS: Placental villous tissue was obtained from patients undergoing elective terminations by dilatation and curettage at weeks 8-12 (n = 10), weeks 13-14 (n = 8), as well as from term deliveries at weeks 37-40 (n = 6). Tissues were fixed, processed and sections utilized for immunofluorescence analysis of FERMT2 expression during gestation. Additionally, HTR8-SVneo human trophoblast cells were transfected by electroporation with FERMT2-specific siRNAs or non-targeting siRNAs (control) and used in cell-substrate adhesion as well as invasion assays. RESULTS: FERMT2 was more commonly expressed in the basal domain of villous cytotrophoblast cells and prominently localized around the periphery of individual extravillous trophoblast cells. siRNA-mediated knockdown of FERMT2 in HTR8-SVneo cells resulted in significantly decreased trophoblast-substrate attachment (p < 0.05) as well as significantly decreased trophoblast invasion (p < 0.05) relative to control cells. CONCLUSIONS: The detection of FERMT2 throughout extravillous trophoblast columns and the results of invasion assays demonstrated that this protein is likely an important regulator of integrin activation in extravillous cells to modulate migration and invasion.


Subject(s)
Cell Movement , Chorionic Villi/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Trophoblasts/cytology , Cell Adhesion , Cell Line , Humans , Immune Sera/metabolism , Integrin alpha6/metabolism , RNA, Small Interfering/metabolism , von Willebrand Factor/metabolism
18.
Cancer Cell Int ; 18: 166, 2018.
Article in English | MEDLINE | ID: mdl-30386175

ABSTRACT

BACKGROUND: Kindlin-2 is one of the Kindlin family members which are evolutionarily conserved focal adhesion proteins with integrin ß-binding affinity. Recently, accumulative studies have suggested that Kindlin-2 plays important roles in tumor biology. However, the prognostic significance of Kindlin-2 in patients with solid tumors remains controversial. Therefore, this study aimed to clarify the prognostic value of Kindlin-2 in solid tumors via meta-analysis. METHODS: A comprehensive search was performed in PubMed, Embase, Web of Science and EBSCO for all relevant studies reporting the prognostic significance of Kindlin-2 expression in solid cancer patients. The summary hazard ratio (HR) and corresponding 95% confidence interval (CI) were calculated to estimate the association between Kindlin-2 expression with survival of solid cancer patients. RESULTS: We included 14 eligible studies containing 1869 patients in our meta-analysis. The pooled results indicated that high Kindlin-2 expression was significantly associated with poor overall survival (OS) (pooled HR 1.66, 95% CI 1.44-1.92, P < 0.0001), disease-free survival (DFS)/recurrence-free survival (RFS)/progression-free survival (PFS) (pooled HR 1.73, 95% CI 1.16-2.57, P = 0.0067). For certain tumor types, high Kindlin-2 expression was significantly correlated with a poor outcome in patients with solid tumors, including pancreatic ductal adenocarcinoma (DFS/RFS/PFS), esophageal squamous cell carcinoma (OS, DFS/RFS/PFS), hepatocellular carcinoma (OS), clear cell renal cell carcinoma (OS), bladder cancer (OS, DFS/RFS/PFS), chondrosarcoma (OS), osteosarcoma (OS), gastric cancer (DFS/RFS/PFS), and glioma (OS). CONCLUSIONS: Our meta-analysis demonstrated that high Kindlin-2 expression might indicate poor outcome in patients with solid tumors and could serve as a prognostic biomarker for solid cancer patients.

19.
J Am Soc Nephrol ; 28(12): 3545-3562, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28775002

ABSTRACT

Alteration of podocyte behavior is critically involved in the development and progression of many forms of human glomerular diseases. The molecular mechanisms that control podocyte behavior, however, are not well understood. Here, we investigated the role of Kindlin-2, a component of cell-matrix adhesions, in podocyte behavior in vivo Ablation of Kindlin-2 in podocytes resulted in alteration of actin cytoskeletal organization, reduction of the levels of slit diaphragm proteins, effacement of podocyte foot processes, and ultimately massive proteinuria and death due to kidney failure. Through proteomic analyses and in vitro coimmunoprecipitation experiments, we identified Rho GDP-dissociation inhibitor α (RhoGDIα) as a Kindlin-2-associated protein. Loss of Kindlin-2 in podocytes significantly reduced the expression of RhoGDIα and resulted in the dissociation of Rac1 from RhoGDIα, leading to Rac1 hyperactivation and increased motility of podocytes. Inhibition of Rac1 activation effectively suppressed podocyte motility and alleviated the podocyte defects and proteinuria induced by the loss of Kindlin-2 in vivo Our results identify a novel Kindlin-2-RhoGDIα-Rac1 signaling axis that is critical for regulation of podocyte structure and function in vivo and provide evidence that it may serve as a useful target for therapeutic control of podocyte injury and associated glomerular diseases.


Subject(s)
Cytoskeletal Proteins/metabolism , Membrane Proteins/metabolism , Muscle Proteins/metabolism , Neoplasm Proteins/metabolism , Neuropeptides/metabolism , Podocytes/metabolism , rac1 GTP-Binding Protein/metabolism , rho Guanine Nucleotide Dissociation Inhibitor alpha/metabolism , Albuminuria/metabolism , Animals , Apoptosis , Cell Movement , Creatinine/analysis , Cytoskeletal Proteins/genetics , Disease Progression , Female , Fibrosis , Genotype , Humans , Kidney Glomerulus/pathology , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , Muscle Proteins/genetics , Neoplasm Proteins/genetics , RNA, Small Interfering/metabolism , Renal Insufficiency/pathology , Signal Transduction
20.
Zhonghua Wai Ke Za Zhi ; 56(8): 617-622, 2018 Aug 01.
Article in Zh | MEDLINE | ID: mdl-30107706

ABSTRACT

Objective: To investigate the effects of Kindlin-2 on malignant phenotypes of human gallbladder cancer cells and discuss the mechanisms. Methods: The expression level of Kindlin-2 in 30 cases of gallbladder cancer tissues and adjacent non-tumoral tissues collected from the First Affiliated Hospital of Zhengzhou University between September 2012 and May 2013 was assessed by real-time PCR and immunohistochemistry.Lentivirus-mediated Kindlin-2 overexpression was used in gallbladder cancer cell lines GBC-SD and SGC-996.Transwell assay and adhesion assay were investigated to explore the functional role of Kindlin-2 on gallbladder cancer cells.Western Blot was used to test the protein change of epithelial-mesenchymal transition(EMT) characteristics. The t-test was used to analyzed results. Results: The RNA and protein levels of Kindlin-2 in gallbladder cancer tissues were higher than in the non-tumoral tissues (t=4.372, P=0.001; t=7.477, P=0.000). The expression level of Kindlin-2 in gallbladder cancer tissues was correlated with Nevin stage(χ(2)=5.932, P=0.035). Compared with control groups, the cell-matrix adhesion ability of GBC-SD and SGC-996 with Kindlin-2 overexpression was obviously promoted(1.66±0.03 vs. 1.07±0.22, t=2.710, P=0.041; 2.66±0.24 vs. 1.03±0.02, t=6.610, P=0.020). The number of GBC-SD and SGC-996 cells with Kindlin-2 overexpression passing through the Transwell chamber matrix increased significantly compared with the control groups(116.1±13.9 vs. 54.7±8.4, t=3.781, P=0.019; 136.3±7.5 vs. 64.3±6.4, t=7.302, P=0.002). The wound healing rate of GBC-SD with Kindlin-2 overexpression at 12-hour and 24-hour was higher than that of the group ((42.9±2.2)% vs. (29.7±1.7)%, t=4.690, P=0.009; (65.0±2.4)% vs.(40.4±2.0)%, t=7.945, P=0.001). The wound healing rate of SGC-996 with Kindlin-2 overexpression at 12-hour and 24-hour was also higher than that of the group ((32.9±1.3)% vs. (24.1±1.5)%, t=4.518, P=0.011; (51.3±1.1)% vs. (39.2±1.1)%, t=8.001, P=0.001). The characteristics of EMT were induced in gallbladder cancer cells with Kindlin-2 overexpression, including the up-regulation of N-cadherin, Vemintin and the down-regulation of E-cadherin. Conclusion: The expression of Kindlin-2 is up-regulated in gallbladder cancer tissues and Kindlin-2 promoted the malignant phenotypes of gallbladder cancer cells partially by epithelial-mesenchymal transition.


Subject(s)
Epithelial-Mesenchymal Transition , Gallbladder Neoplasms , Membrane Proteins , Neoplasm Invasiveness , Neoplasm Proteins , Cell Line, Tumor , Cell Movement , Gallbladder Neoplasms/genetics , Gallbladder Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Humans , Membrane Proteins/physiology , Neoplasm Invasiveness/genetics , Neoplasm Proteins/physiology , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL