Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 187(16): 4289-4304.e26, 2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-38942015

RESUMEN

Cellular homeostasis is intricately influenced by stimuli from the microenvironment, including signaling molecules, metabolites, and pathogens. Functioning as a signaling hub within the cell, mitochondria integrate information from various intracellular compartments to regulate cellular signaling and metabolism. Multiple studies have shown that mitochondria may respond to various extracellular signaling events. However, it is less clear how changes in the extracellular matrix (ECM) can impact mitochondrial homeostasis to regulate animal physiology. We find that ECM remodeling alters mitochondrial homeostasis in an evolutionarily conserved manner. Mechanistically, ECM remodeling triggers a TGF-ß response to induce mitochondrial fission and the unfolded protein response of the mitochondria (UPRMT). At the organismal level, ECM remodeling promotes defense of animals against pathogens through enhanced mitochondrial stress responses. We postulate that this ECM-mitochondria crosstalk represents an ancient immune pathway, which detects infection- or mechanical-stress-induced ECM damage, thereby initiating adaptive mitochondria-based immune and metabolic responses.


Asunto(s)
Matriz Extracelular , Homeostasis , Mitocondrias , Respuesta de Proteína Desplegada , Matriz Extracelular/metabolismo , Animales , Mitocondrias/metabolismo , Humanos , Factor de Crecimiento Transformador beta/metabolismo , Dinámicas Mitocondriales , Ratones , Transducción de Señal , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/inmunología
2.
J Biol Chem ; 300(2): 105607, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38159864

RESUMEN

A mouse model was used to investigate the role of the hyaluronidase, transmembrane protein 2 (TMEM2), on the progression of Graves' orbital (GO) disease. We established a GO mouse model through immunization with a plasmid expressing the thyroid stimulating hormone receptor. Orbital fibroblasts (OFs) were subsequently isolated from both GO and non-GO mice for comprehensive in vitro analyses. The expression of TMEM2 was assessed using qRT-PCR, Western blot and immunohistochemistry in vivo. Disease pathology was evaluated by H&E staining and Masson's trichrome staining in GO mouse tissues. Our investigation revealed a notable reduction in TMEM2 expression in GO mouse orbital tissues. Through overexpression and knockdown assays, we demonstrated that TMEM2 suppresses inflammatory cytokines and reactive oxygen species production. TMEM2 also inhibits the formation of lipid droplets in OFs and the expression of adipogenic factors. Further incorporating Gene Set Enrichment Analysis of relevant GEO datasets and subsequent in vitro cell experiments, robustly confirmed that TMEM2 overexpression was associated with a pronounced upregulation of the JAK/STAT signaling pathway. In vivo, TMEM2 overexpression reduced inflammatory cell infiltration, adipogenesis, and fibrosis in orbital tissues. These findings highlight the varied regulatory role of TMEM2 in GO pathogenesis. Our study reveals that TMEM2 plays a crucial role in mitigating inflammation, suppressing adipogenesis, and reducing fibrosis in GO. TMEM2 has potential as a therapeutic target and biomarker for treating or alleviating GO. These findings advance our understanding of GO pathophysiology and provide opportunities for targeted interventions to modulate TMEM2 for therapeutic purposes.


Asunto(s)
Oftalmopatía de Graves , Transducción de Señal , Animales , Ratones , Adipogénesis , Células Cultivadas , Fibroblastos/metabolismo , Fibrosis , Oftalmopatía de Graves/genética , Oftalmopatía de Graves/metabolismo , Ratones Endogámicos , Especies Reactivas de Oxígeno/metabolismo
3.
J Biol Chem ; 299(9): 105120, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37527776

RESUMEN

Transmembrane protein 2 (TMEM2) was originally identified as a membrane-anchored protein of unknown function. We previously demonstrated that TMEM2 can degrade hyaluronan (HA). Furthermore, we showed that induced global knockout of Tmem2 in adult mice results in rapid accumulation of incompletely degraded HA in bodily fluids and organs, supporting the identity of TMEM2 as a cell surface hyaluronidase. In spite of these advances, no direct evidence has been presented to demonstrate the intrinsic hyaluronidase activity of TMEM2. Here, we directly establish the catalytic activity of TMEM2. The ectodomain of TMEM2 (TMEM2ECD) was expressed as a His-tagged soluble protein and purified by affinity and size-exclusion chromatography. Both human and mouse TMEM2ECD robustly degrade fluorescein-labeled HA into 5 to 10 kDa fragments. TMEM2ECD exhibits this HA-degrading activity irrespective of the species of TMEM2 origin and the position of epitope tag insertion. The HA-degrading activity of TMEM2ECD is more potent than that of HYAL2, a hyaluronidase which, like TMEM2, has been implicated in cell surface HA degradation. Finally, we show that TMEM2ECD can degrade not only fluorescein-labeled HA but also native high-molecular weight HA. In addition to these core findings, our study reveals hitherto unrecognized confounding factors, such as the quality of reagents and the choice of assay systems, that could lead to erroneous conclusions regarding the catalytic activity of TMEM2. In conclusion, our results demonstrate that TMEM2 is a legitimate functional hyaluronidase. Our findings also raise cautions regarding the choice of reagents and methods for performing degradation assays for hyaluronidases.


Asunto(s)
Hialuronoglucosaminidasa , Proteínas de la Membrana , Animales , Humanos , Ratones , Membrana Celular/metabolismo , Fluoresceínas , Ácido Hialurónico/metabolismo , Hialuronoglucosaminidasa/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo
4.
J Biol Chem ; 299(6): 104826, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37196767

RESUMEN

Cutaneous hyaluronan (HA) is depolymerized to intermediate sizes in the extracellular matrix, and further fragmented in the regional lymph nodes. Previously, we showed that the HA-binding protein involved in HA depolymerization (HYBID), also known as KIAA1199/CEMIP, is responsible for the first step of HA depolymerization. Recently, mouse transmembrane 2 (mTMEM2) with high structural similarity to HYBID was proposed to be a membrane-bound hyaluronidase. However, we showed that the knockdown of human TMEM2 (hTMEM2) conversely promoted HA depolymerization in normal human dermal fibroblasts (NHDFs). Therefore, we examined the HA-degrading activity and function of hTMEM2 using HEK293T cells. We found that human HYBID and mTMEM2, but not hTMEM2, degraded extracellular HA, indicating that hTMEM2 does not function as a catalytic hyaluronidase. Analysis of the HA-degrading activity of chimeric TMEM2 in HEK293T cells suggested the importance of the mouse GG domain. Therefore, we focused on the amino acid residues that are conserved in active mouse and human HYBID and mTMEM2 but are substituted in hTMEM2. The HA-degrading activity of mTMEM2 was abolished when its His248 and Ala303 were simultaneously replaced by the corresponding residues of inactive hTMEM2 (Asn248 and Phe303). In NHDFs, enhancement of hTMEM2 expression by proinflammatory cytokines decreased HYBID expression and increased hyaluronan synthase 2-dependent HA production. The effects of proinflammatory cytokines were abrogated by hTMEM2 knockdown. A decreased HYBID expression by interleukin-1ß and transforming growth factor-ß was canceled by hTMEM2 knockdown. In conclusion, these results indicate that hTMEM2 is not a catalytic hyaluronidase, but a regulator of HA metabolism.


Asunto(s)
Ácido Hialurónico , Hialuronoglucosaminidasa , Animales , Humanos , Ratones , Citocinas , Células HEK293 , Hialuronano Sintasas/genética , Ácido Hialurónico/metabolismo , Hialuronoglucosaminidasa/genética , Hialuronoglucosaminidasa/metabolismo
5.
Mol Biol Rep ; 51(1): 417, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38483660

RESUMEN

BACKGROUND: Bronchial epithelial cells are at the front line of viral infections. Toll-like receptor 3 (TLR3) cascade causes the expression of interferon (IFN)-ß and IFN-stimulated genes (ISGs), which in turn induce an antiviral response. Members of the transmembrane protein (TMEM) family are expressed in various cell types. Although the prognostic value of TMEM2 in various cancers has been reported, its association with infectious diseases remains unknown. In this study, we investigated the effects of TMEM2 on antiviral immunity in BEAS-2B bronchial epithelial cells. METHODS AND RESULTS: TMEM2 protein was found in the cytoplasm of normal human bronchial epithelial cells and differed between organs using immunohistochemistry. Cultured BEAS-2B cells were transfected with TMEM2 siRNA, followed by administration of TLR3 ligand polyinosinic-polycytidylic acid (poly IC) or recombinant human (r(h)) IFN-ß. The expression of TMEM2, IFN-ß, ISG56, C-X-C motif chemokine ligand 10 (CXCL10) and hyaluronan were evaluated appropriately by western blotting, quantitative reverse transcription-polymerase chain reaction and enzyme-linked immunosorbent assay. TMEM2 expression was not altered by poly IC stimulation. Knockdown of TMEM2 increased poly IC-induced expression of IFN-ß, CXCL10, and ISG56, while IFN-ß-induced expression of ISG56 and CXCL10 were not changed by TMEM2 knockdown. The hyaluronan concentration in the medium was decreased by either TMEM2 knockdown or poly IC, but additive or synergistic effects were not observed. CONCLUSIONS: TMEM2 knockdown enhanced TLR3-mediated IFN-ß, CXCL10, and ISG56 expression in BEAS-2B cells. This implies that TMEM2 suppresses antiviral immune responses and prevents tissue injury in bronchial epithelial cells.


Asunto(s)
Ácido Hialurónico , Receptor Toll-Like 3 , Humanos , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/metabolismo , Ligandos , Poli I-C/farmacología , Células Epiteliales/metabolismo , Células Cultivadas , Quimiocina CXCL10/genética
6.
J Biol Chem ; 297(5): 101281, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34624311

RESUMEN

As a major component of the extracellular matrix, hyaluronan (HA) plays an important role in defining the biochemical and biophysical properties of tissues. In light of the extremely rapid turnover of HA and the impact of this turnover on HA biology, elucidating the molecular mechanisms underlying HA catabolism is key to understanding the in vivo functions of this unique polysaccharide. Here, we show that TMEM2, a recently identified cell surface hyaluronidase, plays an essential role in systemic HA turnover. Employing induced global Tmem2 knockout mice (Tmem2iKO), we determined the effects of Tmem2 ablation not only on the accumulation of HA in bodily fluids and organs, but also on the process of HA degradation in vivo. Within 3 weeks of tamoxifen-induced Tmem2 ablation, Tmem2iKO mice exhibit pronounced accumulation of HA in circulating blood and various organs, reaching levels as high as 40-fold above levels observed in control mice. Experiments using lymphatic and vascular injection of fluorescent HA tracers demonstrate that ongoing HA degradation in the lymphatic system and the liver is significantly impaired in Tmem2iKO mice. We also show that Tmem2 is strongly expressed in endothelial cells in the subcapsular sinus of lymph nodes and in the liver sinusoid, two primary sites implicated in systemic HA turnover. Our results establish TMEM2 as a physiologically relevant hyaluronidase with an essential role in systemic HA catabolism in vivo, acting primarily on the surface of endothelial cells in the lymph nodes and liver.


Asunto(s)
Células Endoteliales/enzimología , Regulación Enzimológica de la Expresión Génica , Ácido Hialurónico/metabolismo , Hialuronoglucosaminidasa/biosíntesis , Proteínas de la Membrana/biosíntesis , Animales , Ácido Hialurónico/genética , Hialuronoglucosaminidasa/genética , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados
7.
J Biol Chem ; 296: 100481, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33647313

RESUMEN

The extracellular matrix (ECM) plays an important role in maintaining tissue homeostasis and poses a significant physical barrier to in vivo cell migration. Accordingly, as a means of enhancing tissue invasion, tumor cells use matrix metalloproteinases to degrade ECM proteins. However, the in vivo ECM is comprised not only of proteins but also of a variety of nonprotein components. Hyaluronan (HA), one of the most abundant nonprotein components of the interstitial ECM, forms a gel-like antiadhesive barrier that is impenetrable to particulate matter and cells. Mechanisms by which tumor cells penetrate the HA barrier have not been addressed. Here, we demonstrate that transmembrane protein 2 (TMEM2), the only known transmembrane hyaluronidase, is the predominant mediator of contact-dependent HA degradation and subsequent integrin-mediated cell-substrate adhesion. We show that a variety of tumor cells are able to eliminate substrate-bound HA in a tightly localized pattern corresponding to the distribution of focal adhesions (FAs) and stress fibers. This FA-targeted HA degradation is mediated by TMEM2, which itself is localized at site of FAs. TMEM2 depletion inhibits the ability of tumor cells to attach and migrate in an HA-rich environment. Importantly, TMEM2 directly binds at least two integrins via interaction between extracellular domains. Our findings demonstrate a critical role for TMEM2-mediated HA degradation in the adhesion and migration of cells on HA-rich ECM substrates and provide novel insight into the early phase of FA formation.


Asunto(s)
Ácido Hialurónico/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Adhesión Celular/fisiología , Línea Celular Tumoral , Membrana Celular/metabolismo , Movimiento Celular/fisiología , Matriz Extracelular/metabolismo , Adhesiones Focales/metabolismo , Adhesiones Focales/fisiología , Humanos , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/fisiología , Hialuronoglucosaminidasa/metabolismo , Integrinas/metabolismo , Proteínas de la Membrana/fisiología , Ratones
8.
Biochem Biophys Res Commun ; 624: 102-111, 2022 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-35940122

RESUMEN

Hyaluronic acid is a main extracellular matrix component in the central nervous system (CNS), which provides structural support under physical and physiological conditions to maintain cellular homeostasis. However, hyaluronic acid and its degradation products are present within focal demyelinating lesions in multiple sclerosis (MS) patients and autoimmune encephalomyelitis (EAE) mouse models. Differentiated plasma membranes called myelin membranes are generated by oligodendrocytes (also called oligodendroglial cells), which are glial cells that wrap neuronal axons in the CNS. Despite these positive or negative relationships of hyaluronic acid with oligodendroglial cell differentiation and/or myelination, it remains unclear whether and how hyaluronic acid affects oligodendroglial cells. Here, we showed that hyaluronic acid and the cognate receptor CD44 are directly involved in inhibiting morphological differentiation in FBD-102b cells, which are differentiation models of oligodendroglial precursor cells, and primary oligodendroglial precursor cells. Their phenotype changes were supported by decreased oligodendroglial cell differentiation, myelin marker protein expression levels, and Akt kinase phosphorylation levels as a marker kinase. Furthermore, the effects of hyaluronic acid required transmembrane protein 2 (TMEM2), a cell surface hyaluronidase. These results suggest that hyaluronic acid and the CD44 receptor, acting through TMEM2, contribute to inhibiting morphological differentiation of oligodendroglial cells, providing a mechanism underlying cell physiological and possible pathological effects responsible for hyaluronic acid.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Ácido Hialurónico , Animales , Diferenciación Celular/fisiología , Encefalomielitis Autoinmune Experimental/metabolismo , Receptores de Hialuranos , Ácido Hialurónico/metabolismo , Ácido Hialurónico/farmacología , Proteínas de la Membrana/metabolismo , Ratones , Proteínas de la Mielina/metabolismo , Vaina de Mielina/metabolismo , Oligodendroglía/metabolismo
9.
Exp Cell Res ; 400(1): 112517, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33582094

RESUMEN

To investigate mechanisms that TMEM2 activation inhibits hepatitis B virus (HBV) infection in hepatocarcinoma (HCC) cells, co-immunoprecipitation (Co-IP) and mass spectrometry were used in screening interacting proteins for TMEM2. Levels of casein kinase 2 subunit α3 (CSNK2A3) in HCC cells were found to be inhibited or overexpressed using siRNAs and pcDNA3.1-CSNK2A3, respectively. Effect of CSNK2A3 expression on cell proliferation was analyzed using MTS, while its effect on HBV infection was measured using ddPCR and IHC. Western blotting and JAK inhibitor ruxolitinib were also used to determine whether TMEM2-regulated CSNK2A3 expression and HBV infection were affected by JAK-STAT signaling. Co-IP and mass spectrometry results showed that CSNK2A3 interacts with TMEM2. Moreover, overexpression of CSNK2A3 significantly inhibited cell proliferation, while inhibition of CSNK2A3 promoted proliferation of HCC cells. In addition, overexpression of CSNK2A3 was observed to significantly enhance HBV infection, while siRNA knockdown of CSNK2A3 inhibited HBV infection. Notably, effect of CSNK2A3 overexpression on HBV infection was suppressed by TMEM2 overexpression. Further mechanistic analyses have revealed that TMEM2 could antagonize the effects of CSNK2A3 on cell proliferation and HBV infection via JAK-STAT pathway activation. In conclusion, TMEM2 has been determined to bind to CSNK2A3 to inhibit HBV infection via activation of the JAK-STAT pathway.


Asunto(s)
Quinasa de la Caseína II/metabolismo , Epilepsia del Lóbulo Temporal/prevención & control , Hepatitis B/prevención & control , Janus Quinasa 1/metabolismo , Proteínas de la Membrana/metabolismo , Factores de Transcripción STAT/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Carcinoma Hepatocelular/complicaciones , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/virología , Estudios de Casos y Controles , Quinasa de la Caseína II/genética , Epilepsia del Lóbulo Temporal/genética , Epilepsia del Lóbulo Temporal/metabolismo , Epilepsia del Lóbulo Temporal/patología , Hepatitis B/metabolismo , Hepatitis B/patología , Hepatitis B/virología , Virus de la Hepatitis B/fisiología , Humanos , Janus Quinasa 1/genética , Neoplasias Hepáticas/complicaciones , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/virología , Proteínas de la Membrana/genética , PPAR gamma/genética , PPAR gamma/metabolismo , Factores de Transcripción STAT/genética , Células Tumorales Cultivadas
10.
Biochem Biophys Res Commun ; 539: 77-82, 2021 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-33422943

RESUMEN

In the skin, the metabolism of hyaluronan (HA) is highly regulated. Aging leads to chronic low-grade inflammation, which is characterized by elevated levels of pro-inflammatory cytokines; however, the relationship between inflammation and HA metabolism is not clear. Herein, we investigated the effects of a mixture of pro-inflammatory cytokines containing TNF-α, IL-1ß, and IL-6 on HA metabolism in human skin fibroblasts. Treatment with the cytokine mixture for 24 h suppressed HA depolymerization via downregulation of HYBID (HA-binding protein involved in HA depolymerization/KIAA1199/CEMIP) and promoted HA synthesis via upregulation of HAS2 in human skin fibroblasts. Moreover, HAS2-dependent HA synthesis was driven mainly by IL-1ß with partial contribution from TNF-α. Transmembrane protein 2 (TMEM2/CEMIP2), which was previously reported as a candidate hyaluronidase, was upregulated by the cytokine mixture, suggesting that TMEM2 might not function as a hyaluronidase in human skin fibroblasts. Furthermore, the effects of the cytokine mixture on HA metabolism were observed in fibroblasts after 8 days of treatment with cytokines during three passages. Thus, we have shown that HYBID-mediated HA metabolism is negatively regulated by the pro-inflammatory cytokine mixture, providing novel insights into the relationship between inflammation and HA metabolism in the skin.


Asunto(s)
Receptores de Hialuranos/metabolismo , Hialuronano Sintasas/metabolismo , Ácido Hialurónico/química , Hialuronoglucosaminidasa/antagonistas & inhibidores , Interleucina-1beta/farmacología , Piel/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Hialuronano Sintasas/genética , Ácido Hialurónico/metabolismo , Piel/metabolismo , Piel/patología , Envejecimiento de la Piel/efectos de los fármacos , Envejecimiento de la Piel/genética , Envejecimiento de la Piel/patología
11.
Pancreatology ; 20(7): 1479-1485, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32948431

RESUMEN

BACKGROUND: Abnormal metabolism of hyaluronan (HA), a major component of extracellular matrix, is a hallmark of cancer. Our previous studies have shown the importance of enzymes responsible for HA degradation in the aggressive phenotype of pancreatic ductal adenocarcinoma (PDAC). In the present study, we investigated the expression and function of transmembrane protein 2 (TMEM2), a recently identified HA-degrading enzyme, in PDAC. MATERIALS & METHODS: We used immunohistochemistry to investigate expression patterns of TMEM2 in archival tissues obtained from 100 patients with PDAC who underwent surgical resection from 1982 to 2012. The correlations between TMEM2 expression and clinicopathological variables, including survival, were determined using univariate and multivariate analyses. The effect of TMEM2 on proliferation and migratory ability (measured using transwell cell migration assay) of PDAC cells was determined by TMEM2 knockdown with small-interfering RNA (siRNA). RESULTS: Immunohistochemical analysis revealed high expression of TMEM2 in 22 (22%) of 100 patients. The overall survival was significantly shorter in patients with high TMEM2 expression than in those with low expression (P = 0.013). Multivariate analysis identified high TMEM2 expression as an independent factor predicting poor prognosis (P = 0.011). Unexpectedly, knockdown of TMEM2 resulted in increased migratory ability of PDAC cells, which was associated with increased expression of KIAA1199, a potent HA-degrading enzyme shown to enhance cell migration. CONCLUSION: TMEM2 overexpression is associated with poor prognosis in PDAC patients. Targeted disruption of this molecule, however, could enhance the aggressiveness of PDAC cells through a possible interaction with KIAA1199.


Asunto(s)
Carcinoma Ductal Pancreático/enzimología , Hialuronoglucosaminidasa/biosíntesis , Proteínas de la Membrana/biosíntesis , Neoplasias Pancreáticas/enzimología , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor , Carcinoma Ductal Pancreático/diagnóstico , Carcinoma Ductal Pancreático/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular , Femenino , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Hialuronoglucosaminidasa/genética , Estimación de Kaplan-Meier , Masculino , Proteínas de la Membrana/genética , Persona de Mediana Edad , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/genética , Valor Predictivo de las Pruebas , Pronóstico , ARN Interferente Pequeño/farmacología , Análisis de Supervivencia
12.
Biosci Biotechnol Biochem ; 84(8): 1576-1584, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32326855

RESUMEN

Transmembrane 2 (TMEM2) gene inhibits chronic hepatitis-B virus (HBV) infection, while the underlying molecular mechanisms remain unknown. Transcriptome alterations in HepG2 cells following TMEM2 overexpression or silencing by shRNA were analyzed by next-generation sequencing. Both overexpression and knockdown of the TMEM2 gene caused wide-spread changes in gene expression in HepG2 cells. Differentially expressed genes caused by altered TMEM2 gene expression were associated with multiple biological processes linked with viral infection and various signaling pathways. KEGG analysis revealed that many of the differentially expressed genes were enriched in the PI3K/AKT signaling pathway. Moreover, we show that genes related to the PI3K/AKT signaling pathway, such as SYK, FLT4, AKT3, FLT1, and IL6, are biological targets regulated by TMEM2 in HepG2 cells. This is the first transcriptome-wide study in which TMEM2-regulated genes in HepG2 cells have been screened. Our findings elucidate the molecular events associated with TMEM2-mediated hepatocyte pathogenesis in chronic HBV infection.


Asunto(s)
Proteínas de la Membrana/genética , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Transducción de Señal/genética , Transcriptoma , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Células Hep G2 , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Proteínas de la Membrana/agonistas , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/metabolismo , Anotación de Secuencia Molecular , Fosfatidilinositol 3-Quinasas/metabolismo , Plásmidos/química , Plásmidos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Quinasa Syk/genética , Quinasa Syk/metabolismo , Transfección , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo
13.
J Biol Chem ; 292(18): 7304-7313, 2017 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-28246172

RESUMEN

Hyaluronan (HA) is an extremely large polysaccharide (glycosaminoglycan) involved in many cellular functions. HA catabolism is thought to involve the initial cleavage of extracellular high-molecular-weight (HMW) HA into intermediate-size HA by an extracellular or cell-surface hyaluronidase, internalization of intermediate-size HA, and complete degradation into monosaccharides in lysosomes. Despite considerable research, the identity of the hyaluronidase responsible for the initial HA cleavage in the extracellular space remains elusive. HYAL1 and HYAL2 have properties more consistent with lysosomal hyaluronidases, whereas CEMIP/KIAA1199, a recently identified HA-binding molecule that has HA-degrading activity, requires the participation of the clathrin-coated pit pathway of live cells for HA degradation. Here we show that transmembrane protein 2 (TMEM2), a mammalian homolog of a protein playing a role in zebrafish endocardial cushion development, is a cell-surface hyaluronidase. Live immunostaining and surface biotinylation assays confirmed that mouse TMEM2 is expressed on the cell surface in a type II transmembrane topology. TMEM2 degraded HMW-HA into ∼5-kDa fragments but did not cleave chondroitin sulfate or dermatan sulfate, indicating its specificity to HA. The hyaluronidase activity of TMEM2 was Ca2+-dependent; the enzyme's pH optimum is around 6-7, and unlike CEMIP/KIAA1199, TMEM2 does not require the participation of live cells for its hyaluronidase activity. Moreover, TMEM2-expressing cells could eliminate HA immobilized on a glass surface in a contact-dependent manner. Together, these data suggest that TMEM2 is the long-sought-after hyaluronidase that cleaves extracellular HMW-HA into intermediate-size fragments before internalization and degradation in the lysosome.


Asunto(s)
Membrana Celular/enzimología , Regulación Enzimológica de la Expresión Génica/fisiología , Ácido Hialurónico/metabolismo , Hialuronoglucosaminidasa/biosíntesis , Proteínas de la Membrana/biosíntesis , Animales , Calcio/metabolismo , Línea Celular , Membrana Celular/genética , Humanos , Ácido Hialurónico/genética , Hialuronoglucosaminidasa/genética , Concentración de Iones de Hidrógeno , Proteínas de la Membrana/genética , Ratones
14.
Biochem Biophys Res Commun ; 505(1): 74-80, 2018 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-30241936

RESUMEN

We have previously reported that HYBID (hyaluronan (HA)-binding protein involved in HA depolymerization/KIAA1199/CEMIP) is a specific HA-binding protein that is essential for HA depolymerization in skin and synovial fibroblasts. HA is incorporated into cells in the presence of HYBID and clathrin, degraded in endosomes, and excreted into the extracellular space. However, it is not yet clear whether HYBID itself catalytically cleaves HA. A recent report on transmembrane protein 2 (TMEM2)-a novel cell surface hyaluronidase-prompted us to investigate whether TMEM2 is essential for HYBID-mediated HA depolymerization. In the present study, we found that transforming growth factor beta 1 (TGF-ß1), which suppressed HA depolymerization with a concomitant decrease in HYBID expression, upregulated TMEM2 expression conversely in human skin fibroblasts. TMEM2 expression was not affected by histamine, which significantly increased HA depolymerization accompanied by an increase in HYBID expression. We confirmed a similar response in two other cell lines: KEL FIB keloid fibroblasts and HT1080 fibrosarcoma cells. TGF-ß1 was the only inducer of TMEM2 expression among growth factors including epidermal growth factor (EGF), basic fibroblast growth factor (bFGF), and platelet-derived growth factor-BB (PDGF-BB), which suppressed HYBID expression. Moreover, HYBID knockdown completely suppressed HA depolymerization, whereas TMEM2 knockdown unexpectedly enhanced it. These findings clearly indicate that HYBID is indispensable, but TMEM2 is not involved in the HYBID-mediated HA depolymerization system as a catalytic hyaluronidase in human skin fibroblasts.


Asunto(s)
Fibroblastos/metabolismo , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas/metabolismo , Línea Celular , Línea Celular Tumoral , Fibroblastos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Receptores de Hialuranos/genética , Hialuronoglucosaminidasa , Proteínas de la Membrana/genética , Polimerizacion , Proteínas/genética , Interferencia de ARN , Piel/citología , Sinoviocitos/efectos de los fármacos , Sinoviocitos/metabolismo , Factor de Crecimiento Transformador beta/farmacología
15.
Am J Cancer Res ; 14(6): 3010-3035, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39005682

RESUMEN

Pancreatic adenocarcinoma (PAAD), known as one of the deadliest cancers, is characterized by a complex tumor microenvironment, primarily comprised of cancer-associated fibroblasts (CAFs) in the extracellular matrix. These CAFs significantly alter the matrix by interacting with hyaluronic acid (HA) and the enzyme hyaluronidase, which degrades HA - an essential process for cancer progression and spread. Despite the critical role of this interaction, the specific functions of CAFs and hyaluronidase in PAAD development are not fully understood. Our study investigates this interaction and assesses NSC777201, a new anti-cancer compound targeting hyaluronidase. This research utilized computational methods to analyze gene expression data from the Gene Expression Omnibus (GEO) database, specifically GSE172096, comparing gene expression profiles of cancer-associated and normal fibroblasts. We conducted in-house sequencing of pancreatic cancer cells treated with NSC777201 to identify differentially expressed genes (DEGs) and performed functional enrichment and pathway analysis. The identified DEGs were further validated using the TCGA-PAAD and Human Protein Atlas (HPA) databases for their diagnostic, prognostic, and survival implications, accompanied by Ingenuity Pathway Analysis (IPA) and molecular docking of NSC777201, in-vitro, and preclinical in-vivo validations. The result revealed 416 DEGs associated with CAFs and 570 DEGs related to NSC777201 treatment, with nine overlapping DEGs. A key finding was the transmembrane protein TMEM2, which strongly correlated with FAP, a CAF marker, and was associated with higher-risk groups in PAAD. NSC777201 treatment showed inhibition of TMEM2, validated by rescue assay, indicating the importance of targeting TMEM2. Further analyses, including IPA, demonstrated that NSC777201 regulates CAF cell senescence, enhancing its therapeutic potential. Both in-vitro and in-vivo studies confirmed the inhibitory effect of NSC777201 on TMEM2 expression, reinforcing its role in targeting PAAD. Therefore, TMEM2 has been identified as a theragnostic biomarker in PAAD, influenced by CAF activity and HA accumulation. NSC777201 exhibits significant potential in targeting and potentially reversing critical processes in PAAD progression, demonstrating its efficacy as a promising therapeutic agent.

16.
Heliyon ; 9(6): e16559, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37292284

RESUMEN

Glioblastoma multiforme (GBM) is the most common intracranial malignant tumor and is notorious for its poor prognosis. An important element in the short overall survival of GBM patients is the lack of understanding the pathogenesis and progression of tumor and deficiency biomarkers that can be used for early diagnosis and therapeutic sensitivity monitoring. Studies have shown that transmembrane protein 2 (TMEM2) is participated in tumorigenesis of various human tumors, including rectal and breast cancers. Although Qiuyi Jiang et al. have reported that TMEM2 combined with IDH1/2 and 1p19q can predict the survival time of glioma patients based on bioinformatics, its expression and biological role of glioma remain unclear. In our study, we investigated the effect of TMEM2 expression level on glioma malignancy in public datasets and an independent internal dataset. We revealed TEMM2 expression was higher in GBM tissues than in non-tumor brain tissues (NBT). Moreover, the increase in TMEM2 expression level was closely related to tumor malignancy. The survival analysis showed that TMEM2 high expression reduces survival time in all glioma patients, including GBM and LGG patients. Subsequent experiments demonstrated that knockdown TMEM2 inhibited proliferation of GBM cells. In addition, we analyzed TMEM2 mRNA levels in different GBM subtypes, and demonstrated that TMEM2 expression was upregulated in mesenchymal subtype. Meanwhile, bioinformatics analysis and transwell assay indicated that knockdown TMEM2 suppressed epithelial-mesenchymal transition (EMT) in GBM. Importantly, Kaplan-Meier analysis demonstrated that TMEM2 high expression reduced the treatment response to TMZ in GBM patients. Knockdown of TMEM2 alone did not reduce apoptosis GBM cells, but significant apoptotic cells were observed in the group treated with additional TMZ. These studies may contribute to improving the accuracy of early diagnosis and evaluating the effectiveness of TMZ treatment in GBM patients.

17.
Cell Signal ; 109: 110730, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37244634

RESUMEN

This study clarified the possible molecular mechanisms by which the miR-139-5p/SOX4/TMEM2 axis affected angiogenesis and tumorigenesis of ovarian cancer (OC) based on GEO microarray datasets and experimental support. The expression of miR-139-5p and SOX4 was examined in clinical OC samples. Human umbilical vein endothelial cells (HUVECs) and human OC cell lines were included in vitro experiments. Tube formation assay was conducted in HUVECs. The expression of SOX4, SOX4, and VEGF in OC cells was identified using Western blot and immunohistochemistry. Luciferase assays were conducted to validate the targeting relationship between miR-139-5p and SOX4 and between SOX4 and TMEM2. A RIP assay assessed the binding of SOX4 and miR-139-5p. The impact of miR-139-5p and SOX4 on OC tumorigenesis in vivo was evaluated in nude mice. SOX4 was up-regulated, while miR-139-5p was down-regulated in OC tissues and cells. Ectopic miR-139-5p expression or SOX4 knockdown inhibited angiogenesis and tumorigenicity of OC. By targeting SOX4 in OC, miR-139-5p lowered VEGF expression, angiogenesis, and TMEM2 expression. The miR-139-5p/SOX4/TMEM2 axis also reduced VEGF expression and angiogenesis, which might curtail OC growth in vivo. Collectively, miR-139-5p represses VEGF expression and angiogenesis by targeting the transcription factor SOX4 and down-regulating TMEM2 expression, thereby impeding OC tumorigenesis.


Asunto(s)
MicroARNs , Neoplasias Ováricas , Ratones , Animales , Humanos , Femenino , MicroARNs/metabolismo , Ratones Desnudos , Factor A de Crecimiento Endotelial Vascular/genética , Proliferación Celular/fisiología , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Carcinogénesis/genética , Transformación Celular Neoplásica , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Análisis por Micromatrices , Línea Celular Tumoral , Factores de Transcripción SOXC/genética
18.
Biomolecules ; 12(2)2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-35204753

RESUMEN

Hyaluronan (HA) comprises a fundamental component of the extracellular matrix and participates in a variety of biological processes. Half of the total amount of HA in the human body is present in the skin. HA exhibits a dynamic turnover; its half-life in the skin is less than one day. Nevertheless, the specific participants in the catabolism of HA in the skin have not yet been described in detail, despite the essential role of HA in cutaneous biology. A deeper knowledge of the processes involved will act to support the development of HA-based topical and implantable materials and enhance the understanding of the various related pathological cutaneous conditions. This study aimed to characterize the distribution and activity of hyaluronidases and the other proteins involved in the degradation of HA in healthy human full-thickness skin, the epidermis and the dermis. Hyaluronidase activity was detected for the first time in healthy human skin. The degradation of HA occurred in lysates at an acidic pH. HA gel zymography revealed a single band corresponding to approximately 50 kDa. This study provided the first comprehensive view of the distribution of canonic HA-degrading proteins (HYAL1 and HYAL2) in human skin employing IHF and IHC. Furthermore, contrary to previous assumptions TMEM2, a novel hyaluronidase, as well as CEMIP, a protein involved in HA degradation, were localized in the human epidermis, as well as in the dermis.


Asunto(s)
Ácido Hialurónico , Hialuronoglucosaminidasa , Matriz Extracelular/metabolismo , Humanos , Ácido Hialurónico/química , Hialuronoglucosaminidasa/metabolismo , Proteínas/metabolismo , Piel/metabolismo
19.
DNA Cell Biol ; 40(11): 1381-1395, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34735293

RESUMEN

Gliomas are common intracranial tumors with high morbidity and mortality in adults. Transmembrane protein 2 (TMEM2) is involved in the malignant behavior of solid tumors. TMEM2 regulates cell adhesion and metastasis as well as intercellular communication by degrading nonprotein components of the extracellular matrix. This study aimed to evaluate the relationship between TMEM2 expression levels and glioma subtypes or patient prognosis. Our findings revealed that TMEM2 expression was abnormally upregulated in high-grade glioma. Moreover, combining TMEM2, the status of isocitrate dehydrogenase (IDH) and 1p19q, we subdivided molecular subtypes with significant differences in survival. Patients in the MT-codel-low subgroup had better prognosis than those in the WT-no-codel-high subgroup, who fared the worst. Additionally, correlation analysis of TMEM2 and immune cell infiltration indicated an altered tumor microenvironment (TME) and cell redistribution in the TMEM2 high-expression subtype. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis revealed that focal adhesion and PI3K-Akt signaling pathways were enriched in the TMEM2-expressing group. In conclusion, aberrant TMEM2 expression can be used as an independent prognostic marker for refining glioma molecular subtyping and accurate prognosis. These findings will improve rational decision making to provide individualized therapy for patients with glioma.


Asunto(s)
Glioma/genética , Proteínas de la Membrana/genética , Biomarcadores de Tumor/genética , Neoplasias Encefálicas/patología , China , Cromosomas Humanos Par 1/genética , Cromosomas Humanos Par 19/genética , Biología Computacional/métodos , Bases de Datos Genéticas , Glioma/clasificación , Glioma/metabolismo , Humanos , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/metabolismo , Proteínas de la Membrana/metabolismo , Mutación , Pronóstico , Microambiente Tumoral
20.
Technol Cancer Res Treat ; 19: 1533033820977523, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33251982

RESUMEN

MicroRNAs (miRNAs) are emerging as critical mediators in tumors, including triple-negative breast cancer (TNBC). The role of miR-518a-3p in TNBC was investigated to identify potential therapeutic target. Data from KM Plotter database (www.kmplot.com) showed that high miR-518a-3p expression was significantly associated with overall survival of patients with TNBC (p = 0.04). The expression of miR-518a-3p was dysregulated in TNBC cells. Functional assays revealed that over-expression of miR-518a-3p inhibited cell invasion and migration of TNBC. Additionally, miR-518a-3p could target TMEM2 (transmembrane protein 2), and decreased protein and mRNA expression of TMEM2 in TNBC cells. Knockdown of TMEM2 suppressed cell invasion and migration through inhibiting phospho (p)-JAK1 (Janus kinase 1) and p-STAT (signal transducer and activator of transcription protein) 1/2. Moreover, over-expression of TMEM2 counteracted the suppressive effect of miR-518a-3p on TNBC invasion and migration through promoting the levels of p-JAK1 and p-STAT1/2. In conclusion, miR-518a-3p negatively regulates the JAK/STAT pathway via targeting TMEM2 and suppresses invasion and migration in TNBC, suggesting that miR-518a-3p may be a potential therapeutic target in TNBC.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Proteínas de la Membrana/genética , MicroARNs/genética , Interferencia de ARN , Neoplasias de la Mama Triple Negativas/genética , Biomarcadores de Tumor , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Pronóstico , Análisis de Supervivencia , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/mortalidad , Neoplasias de la Mama Triple Negativas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA