Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Med ; 28(1): 152, 2022 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-36510147

RESUMO

BACKGROUND: Acute renal injury (AKI) secondary to ischemia reperfusion (IR) injury continues to be a significant perioperative problem and there is no effective treatment. Mindin belongs to the mindin/F-spondin family and involves in inflammation, proliferation, and cell apoptosis. Previous studies have explored the biological functions of mindin in liver and brain ischemic injury, but its role in AKI is unknown. METHOD: To investigate whether mindin has a pathogenic role, mindin knockout (KO) and wild-type (WT) mice were used to establish renal IR model. After 30 min of ischemia and 24 h of reperfusion, renal histology, serum creatinine, and inflammatory response were examined to assess kidney injury. In vitro, proinflammatory factors and inflammatory signaling pathways were measured in mindin overexpression or knockdown and vector cells after hypoxia/reoxygenation (HR). RESULTS: Following IR, the kidney mindin level was increased in WT mice and deletion of mindin provided significant protection for mice against IR-induced renal injury as manifested by attenuated the elevation of serum creatinine and blood urea nitrogen along with less severity for histological alterations. Mindin deficiency significantly suppressed inflammatory cell infiltration, TNF-α and MCP-1 production following renal IR injury. Mechanistic studies revealed that mindin deficiency inhibits TLR4/JNK/NF-κB signaling activation. In vitro, the expression levels of TNF-α and MCP-1 were increased in mindin overexpression cells compared with vector cells following HR. Moreover, TLR4/JNK/NF-κB signaling activation was elevated in the mindin overexpression cells in response to HR stimulation while mindin knockdown inhibited the activation of TLR4/JNK/ NF-κB signaling after HR in vitro. Further study showed that mindin protein interacted directly with TLR4 protein. And more, mindin protein was confirmed to be expressed massively in renal tubule tissues of human hydronephrosis patients. CONCLUSION: These data demonstrate that mindin is a critical modulator of renal IR injury through regulating inflammatory responses. TLR4/JNK/NF-κB signaling most likely mediates the biological function of mindin in this model of renal ischemia.


Assuntos
NF-kappa B , Traumatismo por Reperfusão , Camundongos , Humanos , Animais , NF-kappa B/metabolismo , Fator de Necrose Tumoral alfa , Creatinina , Traumatismo por Reperfusão/metabolismo , Rim/metabolismo , Hipóxia , Isquemia , Camundongos Endogâmicos C57BL
2.
J Cell Mol Med ; 24(15): 8391-8404, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32614521

RESUMO

Mindin is important in broad spectrum of immune responses. On the other hand, we previously reported that mindin attenuated human colon cancer development by blocking angiogenesis through Egr-1-mediated regulation. However, the mice original mindin directly suppressed the syngenic colorectal cancer (CRC) growth in our recent study and we aimed to further define the role of mindin during CRC development in mice. We established the mouse syngeneic CRC CMT93 and CT26 WT cell lines with stable mindin knock-down or overexpression. These cells were also subcutaneously injected into C57BL/6 and BALB/c mice as well as established a colitis-associated colorectal cancer (CAC) mouse model treated with lentiviral-based overexpression and knocked-down of mindin. Furthermore, we generated mindin knockout mice using a CRISPR-Cas9 system with CAC model. Our data showed that overexpression of mindin suppressed cell proliferation in both of CMT93 and CT26 WT colon cancer cell lines, while the silencing of mindin promoted in vitro cell proliferation via the ERK and c-Fos pathways and cell cycle control. Moreover, the overexpression of mindin significantly suppressed in vivo tumour growth in both the subcutaneous transplantation and the AOM/DSS-induced CAC models. Consistently, the silencing of mindin reversed these in vivo observations. Expectedly, the tumour growth was promoted in the CAC model on mindin-deficient mice. Thus, mindin plays a direct tumour suppressive function during colon cancer progression and suggesting that mindin might be exploited as a therapeutic target for CRC.


Assuntos
Neoplasias do Colo/genética , Proteínas da Matriz Extracelular/genética , Genes Supressores de Tumor/fisiologia , Sistema de Sinalização das MAP Quinases/genética , Transdução de Sinais/genética , Animais , Ciclo Celular/genética , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/genética , Colite/genética , Colite/patologia , Colo/patologia , Neoplasias do Colo/patologia , Modelos Animais de Doenças , Progressão da Doença , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Células RAW 264.7
3.
J Cell Mol Med ; 24(10): 5740-5750, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32253812

RESUMO

Renal fibrosis acts as a clinical predictor in patients with chronic kidney disease and is characterized by excessive extracellular matrix (ECM) accumulation. Our previous study suggested that mindin can function as a mediator for liver steatosis pathogenesis. However, the role of mindin in renal fibrosis remains obscure. Here, tumour necrosis factor (TGF)-ß-treated HK-2 cells and global mindin knockout mouse were induced with renal ischaemia reperfusion injury (IRI) to test the relationship between mindin and renal fibrosis. In vitro, mindin overexpression promoted p65-the hub subunit of the NF-κB signalling pathway-translocation from the cytoplasm into the nucleus, resulting in NF-κB pathway activation in TGF-ß-treated HK-2 cells. Meanwhile, mindin activated the TGF-ß/Smad pathway, thereby causing fibrotic-related protein expression in vitro. Mindin-/- mice exhibited less kidney lesions than controls, with small renal tubular expansion, inflammatory cell infiltration, as well as collagen accumulation, following renal IRI. Mechanistically, mindin-/- mice suppressed p65 translocation and deactivated NF-κB pathway. Simultaneously, mindin disruption inhibited the TGF-ß/Smad pathway, alleviating the expression of ECM-related proteins. Hence, mindin may be a novel target of renal IRI in the treatment of renal fibrogenesis.


Assuntos
Proteínas da Matriz Extracelular/deficiência , Nefropatias/etiologia , Nefropatias/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Biomarcadores , Biópsia , Modelos Animais de Doenças , Fibrose , Técnicas de Silenciamento de Genes , Inflamação/etiologia , Inflamação/metabolismo , Nefropatias/patologia , Masculino , Camundongos
4.
J Cell Mol Med ; 23(5): 3402-3416, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30869196

RESUMO

Mindin has a broad spectrum of roles in the innate immune system, including in macrophage migration, antigen phagocytosis and cytokine production. Mindin functions as a pattern-recognition molecule for microbial pathogens. However, the underlying mechanisms of mindin-mediated phagocytosis and its exact membrane receptors are not well established. Herein, we generated mindin-deficient mice using the CRISPR-Cas9 system and show that peritoneal macrophages from mindin-deficient mice were severely defective in their ability to phagocytize E  coli. Phagocytosis was enhanced when E  coli or fluorescent particles were pre-incubated with mindin, indicating that mindin binds directly to bacteria or non-pathogen particles and promotes phagocytosis. We defined that 131 I-labelled mindin binds with integrin Mac-1 (CD11b/CD18), the F-spondin (FS)-fragment of mindin binds with the αM -I domain of Mac-1 and that mindin serves as a novel ligand of Mac-1. Blockade of the αM -I domain of Mac-1 using either a neutralizing antibody or si-Mac-1 efficiently blocked mindin-induced phagocytosis. Furthermore, mindin activated the Syk and MAPK signalling pathways and promoted NF-κB entry into the nucleus. Our data indicate that mindin binds with the integrin Mac-1 to promote macrophage phagocytosis through Syk activation and NF-κB p65 translocation, suggesting that the mindin/Mac-1 axis plays a critical role during innate immune responses.


Assuntos
Proteínas da Matriz Extracelular/metabolismo , Antígeno de Macrófago 1/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo , Fagocitose , Receptores de Reconhecimento de Padrão/metabolismo , Quinase Syk/metabolismo , Fator de Transcrição RelA/metabolismo , Animais , Sequência de Bases , Núcleo Celular/metabolismo , Células HEK293 , Humanos , Antígeno de Macrófago 1/química , Camundongos , Camundongos Knockout , Fosforilação , Ligação Proteica , Domínios Proteicos , Transporte Proteico , Células RAW 264.7
5.
Clin Sci (Lond) ; 132(11): 1199-1213, 2018 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-29695588

RESUMO

Mindin, which is a highly conserved extracellular matrix protein, has been documented to play pivotal roles in regulating angiogenesis, inflammatory processes, and immune responses. The aim of the present study was to assess whether mindin contributes to the development of atherosclerosis. A significant up-regulation of Mindin expression was observed in the serum, arteries and atheromatous plaques of ApoE-/- mice after high-fat diet treatment. Mindin-/-ApoE-/- mice and macrophage-specific mindin overexpression in ApoE-/- mice (Lyz2-mindin-TG) were generated to evaluate the effect of mindin on the development of atherosclerosis. The Mindin-/-ApoE-/- mice exhibited significantly ameliorated atherosclerotic burdens in the entire aorta and aortic root and increased atherosclerotic plaque stability. Moreover, bone marrow transplantation further demonstrated that mindin deficiency in macrophages was largely responsible for the alleviated atherogenesis. The Lyz2-mindin-TG mice exhibited the opposite phenotype. Mindin deficiency enhanced foam cell formation by increasing the expression of cholesterol effectors, including ABCA1 and ABCG1. The mechanistic study indicated that mindin ablation promoted LXR-ß expression via a direct interaction. Importantly, LXR-ß inhibition largely reversed the ameliorating effect of mindin deficiency on foam cell formation and ABCA1 and ABCG1 expression. The present study demonstrated that mindin deficiency serves as a novel mediator that protects against foam cell formation and atherosclerosis by directly interacting with LXR-ß.


Assuntos
Aterosclerose/prevenção & controle , Proteínas da Matriz Extracelular/deficiência , Receptores X do Fígado/metabolismo , Macrófagos/metabolismo , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Aterosclerose/metabolismo , Aterosclerose/patologia , Transplante de Medula Óssea , Dieta Hiperlipídica , Proteínas da Matriz Extracelular/biossíntese , Proteínas da Matriz Extracelular/fisiologia , Células Espumosas/patologia , Hiperlipidemias/metabolismo , Mediadores da Inflamação/metabolismo , Receptores X do Fígado/antagonistas & inibidores , Macrófagos/patologia , Masculino , Camundongos Knockout para ApoE , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patologia , Regulação para Cima/fisiologia
6.
Clin Sci (Lond) ; 132(14): 1509-1512, 2018 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-30037838

RESUMO

The hallmark features of atherosclerosis include accumulation of low-density lipoprotein (LDL) carrying cholesterol in the vessel wall, formation of lipid-laden foam cells, and the creation of a pro-inflammatory microenvironment. To date, no effective treatments are clinically available for increasing cholesterol efflux from vascular macrophages and inducing reverse cholesterol transport (RCT). In an article published recently in Clinical Science (vol 132, issue 6, 1199-1213), Zhang and colleagues identified the extracellular matrix protein mindin/spondin 2 as a positive regulator of atherosclerosis. Genetic knockout of mindin in apolipoprotein-E (apoE)-/- mice attenuated atherosclerosis, foam cell formation, and inflammation within the vessel wall. Conversely, selective overexpression of mindin in macrophages in apoE-/- mice was sufficient to promote the greater severity of atherosclerosis. Interestingly, foam cell formation was closely associated with the expression of cholesterol transporters (ABCA1 and ACBG1) that facilitate cholesterol efflux. Liver X receptor (LXR)-ß is a key modulator of cholesterol transporter expression and formed direct interactions with mindin. Furthermore, the protective effects of mindin deficiency on foam cell formation were blocked by inhibition of LXR-ß. This article highlights a novel role of mindin in modulating foam cell formation and atherosclerosis development in mice through direct regulation of LXR-ß. Thus far, direct targetting of LXR-ß via pharmacological agonists has proven to be problematic due to the lack of subtype selective inhibitors and associated adverse effects. Indirect targetting of LXR-ß, therefore, via mindin inhibition offers a new therapeutic strategy for increasing LXR-ß induced cholesterol efflux, reducing foam cell formation, and preventing or treating atherosclerosis.


Assuntos
Aterosclerose , Células Espumosas , Transportador 1 de Cassete de Ligação de ATP , Animais , Apolipoproteínas E , Proteínas da Matriz Extracelular , Receptores X do Fígado , Macrófagos , Camundongos
7.
Acta Cardiol ; 73(6): 544-549, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29265929

RESUMO

PURPOSE: Mindin was associated with diabetic nephropathy, podocyte injury, colitis, allergic asthma, liver ischaemia and reperpusion injury and ischaemic brain injury. On the other hand, it was reported as a protective factor against obesity, cardiac hypertrophy, fibrosis and remodelling. Fragmented QRS complexes (fQRS) are markers of altered ventricular depolarisation owing to a prior myocardial scar and fibrosis. In this study, we aimed to investigate mindin levels in hypertensive patients with left ventricular hypertrophy and fQRS on electrocardiography. METHODS: This observational case-control study enrolled 70 (36 female) hypertensive patients with fQRS and 38 (23 female) hypertensive control patients. All patients were evaluated by transthoracic echocardiography. Mindin levels were measured by the enzyme-linked immunosorbent assay (ELISA). Clinical, echocardiographic and laboratory data were compared between patient and control groups. RESULTS: There was no significant difference between patient and control groups in terms of clinical, echocardiographic and routine laboratory parameters. The mindin levels were significantly higher in the patient group than controls (11.3 (7.21-19.31) vs 4.15 (2.86-6.34); p < .001). Multiple logistic regression analyses defined increased mindin levels as an independent predictor for the presence of fQRS (Odds ratio: 1.733; p = .034). Mindin levels >6.74 predicted the presence of fQRS with a sensitifity of 84.3% and specificity of 79.9% on receiver operating characteristic (ROC) curve analysis (The area under the curve:0.889; Confidence Interval: 0.827-0.951; p < .001). CONCLUSION: Mindin expressin is upregulated in hypertensive patients with fQRS complexes. In contrary to previous studies, increased mindin levels may be associated with myocardial fibrosis.


Assuntos
Eletrocardiografia/métodos , Proteínas da Matriz Extracelular/sangue , Hipertensão/sangue , Hipertrofia Ventricular Esquerda/sangue , Biomarcadores/sangue , Estudos de Casos e Controles , Ecocardiografia , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Hipertensão/complicações , Hipertensão/fisiopatologia , Hipertrofia Ventricular Esquerda/diagnóstico , Hipertrofia Ventricular Esquerda/etiologia , Masculino , Pessoa de Meia-Idade , Curva ROC
8.
J Hepatol ; 63(5): 1198-211, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26165142

RESUMO

BACKGROUND & AIMS: Hepatic ischemia/reperfusion (I/R) injury often occurs during liver surgery and may cause liver failure. Our previous studies revealed that Mindin is involved in the pathogenesis of ischemic stroke. However, the function of Mindin in hepatic I/R injury remains unknown. METHODS: Partial hepatic warm ischemia was induced in parallel in global Mindin knockout mice (Mindin KO), hepatocyte-specific Mindin knockdown mice, hepatocyte-specific Mindin transgenic mice (Mindin TG), myeloid cell-specific Mindin TG mice (LysM-Mindin TG), and their corresponding controls, followed by reperfusion. Hepatic histology, serum aminotransferase, inflammatory cytokines, and hepatocyte apoptosis and proliferation were examined to assess liver injury. The molecular mechanisms of Mindin function were explored in vivo and in vitro. RESULTS: Mindin KO and hepatocyte-specific Mindin knockdown mice exhibited less liver damage than controls, with smaller necrotic areas and lower serum transaminase levels. Mindin deficiency significantly suppressed inflammatory cell infiltration, cytokine and chemokine production, and hepatocyte apoptosis, but increased hepatocyte proliferation following hepatic I/R injury. In contrast, the opposite pathological and biochemical changes were observed in hepatocyte-specific Mindin TG mice, whereas no significant changes in liver damage were found in LysM-Mindin TG mice compared to non-transgenic controls. Mechanistically, Akt signaling was activated in livers of Mindin KO mice but was suppressed in Mindin TG mice. Most importantly, Akt inhibitor treatment blocked the protective effect of Mindin deficiency on hepatic I/R injury. CONCLUSIONS: Mindin is a novel modulator of hepatic I/R injury through regulating inflammatory responses, as well as hepatocyte apoptosis and proliferation via inactivation of the Akt signaling pathway.


Assuntos
Proteínas da Matriz Extracelular/deficiência , Proteínas da Matriz Extracelular/genética , Regulação da Expressão Gênica , Hepatopatias/genética , Fígado/irrigação sanguínea , RNA/genética , Traumatismo por Reperfusão/genética , Animais , Apoptose , Western Blotting , Células Cultivadas , Modelos Animais de Doenças , Citometria de Fluxo , Marcação In Situ das Extremidades Cortadas , Fígado/metabolismo , Fígado/patologia , Hepatopatias/etiologia , Hepatopatias/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/metabolismo , Transdução de Sinais
9.
Nephrology (Carlton) ; 20(10): 721-726, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25973958

RESUMO

AIM: We aimed to determine whether serum SPON2 is a useful biomarker in the detection of Diabetic Nephropathy (DN) and to compare serum SPON2 levels with 24-hour urinary albumin excretion rate (UAER) in patients with DN at different stages. METHODS: The cohort included 80 adult patients with T2D and 20 healthy controls. The patients with T2D were divided into four groups according to UAER and serum creatinine (sCr) levels. Group 1 consisted of patients with normoalbuminuria (n = 20), Group 2 with microalbuminuria (n = 20), Group 3 with macroalbuminuria (n = 20) and Group 4 with albuminuria and sCr > 1.5 mg/dL (n = 20). RESULTS: There were no significant differences between the groups in terms of demographic data, C-reactive protein, HbA1c, lipids, serum uric acid levels and leukocyte counts. SPON2 levels were observed to increase linearly with increasing severity of diabetic nephropathy levels. The SPON2 levels of Group 4 were significantly higher than Group 1 and the controls, and SPON2 levels of Group 3 were significantly higher than Group 1. Blood urea nitrogen, creatinine and UAER were significantly positively correlated with SPON2; serum total protein and calcium levels were negatively correlated with SPON2 in patients with DN. CONCLUSION: We observed a linear and significant increase in SPON2 levels of patients with T2D as the stage of DN increased, but serum SPON2 level was not as effective as microalbuminuria in reflecting nephropathy. Also, serum SPON2 level was not as good as urine and tissue levels of SPON2 in detection of renal damage in DN.

10.
mBio ; 14(2): e0291922, 2023 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-36779788

RESUMO

Mindin is a secreted extracellular matrix protein that is involved in regulating cellular events through interacting with integrin. Studies have demonstrated its role in host immunity, including phagocytosis, cell migration, and cytokine production. However, the function of Mindin in the host-virus interaction is largely unknown. In the present study, we report that Mindin facilitates virus infection by activating lipid utilization in an arthropod, kuruma shrimp (Marsupenaeus japonicus). Shrimp Mindin facilitates white spot syndrome virus infection by facilitating viral entry and replication. By activating autophagy, Mindin induces lipid droplet consumption, the hydrolysis of triglycerides into free fatty acids, and ATP production, ultimately providing energy for virus infection. Moreover, integrin is essential for Mindin-mediated autophagy and lipid utilization. Therefore, by revealing the mechanism by which Mindin facilitates virus infection through regulating lipid metabolism, the present study reveals the significance of Mindin in the host-virus interaction. IMPORTANCE White spot syndrome virus (WSSV) is an enveloped double-stranded DNA virus that has had a serious influence on worldwide shrimp farming in the last 30 years. We have demonstrated that WSSV hijacks host autophagy and lipid metabolism for reproduction in kuruma shrimp (Marsupenaeus japonicus). These findings revealed the mechanism by which WSSV exploits host machinery for its infection and provided serial targets for WSSV prevention and control in shrimp farming.


Assuntos
Vírus da Síndrome da Mancha Branca 1 , Animais , Vírus da Síndrome da Mancha Branca 1/genética , Autofagia , Crustáceos , Proteínas da Matriz Extracelular , Lipídeos
11.
Cell Rep ; 40(12): 111390, 2022 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-36130502

RESUMO

Preservation of a small population of cancer stem cells (CSCs) within a heterogeneous carcinoma serves as a paradigm to understand how select cells in a tissue maintain their undifferentiated status. In both embryogenesis and cancer, Snail has been correlated with stemness, but the molecular underpinning of this phenomenon remains largely ill-defined. In models of cutaneous squamous cell carcinoma (cSCC), we discovered a non-epithelial-mesenchymal transition function for the transcription factor Snail in maintaining the stemness of epidermal keratinocytes. Snail-expressing cells secrete the matricellular protein Mindin, which functions in an autocrine fashion to activate a Src-STAT3 pathway to reinforce their stem/progenitor phenotype. This pathway is activated by the engagement of Mindin with the leukocyte-specific integrin, CD11b (ITGAM), which is also unexpectedly expressed by epidermal keratinocytes. Interestingly, disruption of this signaling module in human cSCC attenuates tumorigenesis, suggesting that targeting Mindin would be a promising therapeutic approach to hinder cancer recurrence.


Assuntos
Carcinoma de Células Escamosas , Neoplasias Cutâneas , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Células Epiteliais/metabolismo , Proteínas da Matriz Extracelular , Humanos , Integrinas/metabolismo , Proteínas de Neoplasias , Recidiva Local de Neoplasia/patologia , Células-Tronco Neoplásicas/metabolismo , Neoplasias Cutâneas/patologia , Fatores de Transcrição da Família Snail/metabolismo
12.
Cancers (Basel) ; 13(3)2021 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-33498862

RESUMO

Advanced prostate cancer preferential metastasis to bone is associated with osteomimicry. MINDIN is a secreted matrix protein upregulated in prostate tumors that overexpresses bone-related genes during prostate cancer progression. Na+/H+ exchanger regulatory factor (NHERF-1) is a scaffold protein that has been involved both in tumor regulation and osteogenesis. We hypothesize that NHERF-1 modulation is a mechanism used by MINDIN to promote prostate cancer progression. We analyzed the expression of NHERF-1 and MINDIN in human prostate samples and in a premetastatic prostate cancer mouse model, based on the implantation of prostate adenocarcinoma TRAMP-C1 (transgenic adenocarcinoma of the mouse prostate) cells in immunocompetent C57BL/6 mice. The relationship between NHERF-1 and MINDIN and their effects on cell proliferation, migration, survival and osteomimicry were evaluated. Upregulation of MINDIN and downregulation of NHERF-1 expression were observed both in human prostate cancer samples and in the TRAMP-C1 model. MINDIN silencing restored NHERF-1 expression to control levels in the mouse model. Stimulation with MINDIN reduced NHERF-1 expression and triggered its mobilization from the plasma membrane to the cytoplasm in TRAMP-C1 cells. MINDIN-dependent downregulation of NHERF-1 promoted tumor cell migration and proliferation without affecting osteomimicry and adhesion. We propose that MINDIN downregulates NHERF-1 expression leading to promotion of processes involved in prostate cancer progression.

13.
J Med Biochem ; 39(1): 108-111, 2020 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-32547325

RESUMO

BACKGROUND: It has been shown that decreased expression and activity of extracellular matrix protein mindin correlate with various types of cancers including breast, colon and lung cancers. The aim of the presented study was to investigate the serum mindin levels in prostate cancer. METHODS: Mindin concentrations in serum were measured in 56 patients with prostate cancer (mean age 68 years) and in control group of 29 healthy men (mean age 64 years) using commercially available enzymatic immunoassay (Cusabio, WuHan, China). The patients were divided with respect to the severity of the disease into two groups according to the EAU guidelines (stage 1, 2 - less severe tumours, stage 3, 4 - severe tumours). RESULTS: Serum mindin concentrations were significantly elevated in the group of healthy individuals unlike in the patients with prostate cancer (2.12 ng/mL vs 0.78 ng/mL, with P=0.0007, AUC=0.705). Patients with less severe tumours (stage 1, 2) and severe tumours (stage 3, 4) had significantly decreased levels of S-mindin as well (P=0.0037), although the difference in serum mindin concentrations between the patients with less severe and severe tumours was not significant. CONCLUSIONS: Concentrations of mindin were decreased in patients with prostate cancer and reduced in patients with less severe prostate cancer as well. Mindin appears to be a promising diagnostic marker useful in the diagnosis of prostate cancer.

14.
Endocr Relat Cancer ; 27(7): 441-456, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32357310

RESUMO

Bone metastases are common in advanced prostate cancer patients, but mechanisms by which specific pro-metastatic skeletal niches are formed before tumor cell homing are unclear. We aimed to analyze the effects of proteins secreted by primary prostate tumors on the bone microenvironment before the settlement and propagation of metastases. Here, using an in vivo pre-metastatic prostate cancer model based on the implantation of prostate adenocarcinoma TRAMP-C1 cells in immunocompetent C57BL/6 mice, we identify MINDIN as a prostate tumor secreted protein that induces bone microstructural and bone remodeling gene expression changes before tumor cell homing. Associated with these changes, increased tumor cell adhesion to the endosteum ex vivo and to osteoblasts in vitro was observed. Furthermore, MINDIN promoted osteoblast proliferation and mineralization and monocyte expression of osteoclast markers. ß-catenin signaling pathway revealed to mediate MINDIN actions on osteoblast gene expression but failed to affect MINDIN-induced adhesion to prostate tumor cells or monocyte differentiation to osteoclasts. Our study evidences that MINDIN secretion by primary prostate tumors creates a favorable bone environment for tumor cell homing before metastatic spread.


Assuntos
Proteínas da Matriz Extracelular/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias da Próstata/metabolismo , beta Catenina/metabolismo , Animais , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Transfecção
15.
Biochim Biophys Acta Gen Subj ; 1864(8): 129632, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32416197

RESUMO

BACKGROUND: Mindin (spondin2), a secretory protein related to neural development and immunity, is a member of thrombospondin type I repeat (TSR) superfamily proteins, and has a unique glycosylation of C-mannosylation in its structure. However, it remains unclear whether C-mannosylation plays a functional role in the biosynthesis of mindin in cells. METHODS: Protein C-mannosylation was analyzed by mass spectrometry. Mindin expression was examined by immunoblot and immunofluorescence analyses in COS-7 cells transfected with the expression vectors for wild type (mindin-WT) or C-mannosylation-defective mutant of mindin (mindin-mutF). The redox status was examined in mindin by using 4-acetoamide-4'-maleimidylstilbene-2,2'-disulfonate. RESULTS: When mindin cDNA was expressed in COS-7 cells, C-mannosylation of mindin was confirmed at Trp257 by mass spectrometry. In cells expressing a mindin-mutF, secretion of the mutant was significantly inhibited compared with mindin-WT. In immunofluorescence analysis, mindin-mutF was accumulated in the endoplasmic reticulum (ER), whereas mindin-WT was detected in the Golgi. In addition, mindin-mutF showed an enhanced interaction with calreticulin, an ER-resident chaperone, in cells. In cells, reduced forms were increased in mindin-mutF, compared with a mostly oxidized form of mindin-WT. In the presence of chemical chaperones such as dimethylsulfoxide or 4-phenylbutyrate, inhibited secretion of mindin-mutF was ameliorated in cells, although redox-dependent folding was not affected. CONCLUSIONS: C-Mannosylation of mindin facilitates its secretion especially through modulating disulfide bond formation in mindin in cells. GENERAL SIGNIFICANCE: These results suggest that C-mannosylation plays a functional role in the redox-dependent folding and transport of TSR superfamily proteins in cells.


Assuntos
Proteínas da Matriz Extracelular/metabolismo , Manose/metabolismo , Animais , Células COS , Células Cultivadas , Chlorocebus aethiops , Retículo Endoplasmático/metabolismo , Glicosilação , Camundongos , Chaperonas Moleculares/metabolismo , Células NIH 3T3 , Coelhos
16.
Exp Neurol ; 247: 506-16, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23360804

RESUMO

BACKGROUND: Stroke is the second leading cause of death among adults worldwide. Mindin is an ECM protein that plays important roles in regulating inflammation, angiogenesis and neuronal outgrowth. The role of mindin in the context of brain ischemia has not been examined. METHODS AND RESULTS: Transient occlusion of the middle cerebral artery was performed on mindin knockout (KO) mice, mice that carried a neuron-specific constitutively active mindin transgene (TG) and the appropriate controls. The outcome of the ischemia was evaluated by examination of the infarct and edema volumes and by neurological score assessments. The brains were collected 24 h or 3 days following the induced stroke. Compared with the control mice, the mindin KO mice exhibited lower infarct volumes and better outcomes in the neurological tests. Mindin-deficient mice exhibited low expression levels of stroke-induced inflammatory mediators, an attenuated recruitment of inflammatory cells, and inhibited activation of NF-κB. The neuronal apoptosis levels were also lower in the brains of the mindin KO mice than in those of the control mice. The mice that expressed a neuron-specific, constitutively active mindin transgene exhibited effects following the cerebral ischemic injury that were the opposite of those that were observed in the mindin KO mice. Moreover, Akt signaling activation was elevated in the ischemic brains of mindin KO mice. CONCLUSIONS: Mindin KO mice exhibited minor infarctions, an attenuated inflammatory response and low levels of neuronal apoptosis following an ischemic insult. These data demonstrate that mindin is a critical mediator of ischemic brain injury in an experimental stroke model. Akt signaling most likely mediates the biological function of mindin in this model of cerebral ischemia.


Assuntos
Lesões Encefálicas/etiologia , Proteínas da Matriz Extracelular/metabolismo , Regulação da Expressão Gênica/genética , Infarto da Artéria Cerebral Média/complicações , Animais , Infarto Encefálico/etiologia , Lesões Encefálicas/patologia , Ciclo-Oxigenase 2/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Proteínas da Matriz Extracelular/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Proteína Oncogênica v-akt/metabolismo , Fator de Crescimento Derivado de Plaquetas/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA