Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1869(7): 159515, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38844203

RESUMEN

Although our current knowledge of the molecular crosstalk between the ER stress, the unfolded protein response (UPR), and lipid homeostasis remains limited, there is increasing evidence that dysregulation of either protein or lipid homeostasis profoundly affects the other. Most research regarding UPR signaling in human diseases has focused on the causes and consequences of disrupted protein folding. The UPR itself consists of very complex pathways that function to not only maintain protein homeostasis, but just as importantly, modulate lipid biogenesis to allow the ER to adjust and promote cell survival. Lipid dysregulation is known to activate many aspects of the UPR, but the complexity of this crosstalk remains a major research barrier. ER lipid disequilibrium and lipotoxicity are known to be important contributors to numerous human pathologies, including insulin resistance, liver disease, cardiovascular diseases, neurodegenerative diseases, and cancer. Despite their medical significance and continuous research, however, the molecular mechanisms that modulate lipid synthesis during ER stress conditions, and their impact on cell fate decisions, remain poorly understood. Here we summarize the current view on crosstalk and connections between altered lipid metabolism, ER stress, and the UPR.

2.
Cell Mol Biol Lett ; 28(1): 86, 2023 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-37880612

RESUMEN

BACKGROUND: Membrane rafts play a crucial role in the regulation of many important biological processes. Our previous data suggest that specific interactions of flotillins with MPP1 are responsible for membrane raft domain organization and regulation in erythroid cells. Interaction of the flotillin-based protein network with specific membrane components underlies the mechanism of raft domain formation and regulation, including in cells with low expression of MPP1. METHODS: We sought to identify other flotillin partners via the immobilized recombinant flotillin-2-based affinity approach and mass spectrometry technique. The results were further confirmed via immunoblotting and via co-immunoprecipitation. In order to study the effect of the candidate protein on the physicochemical properties of the plasma membrane, the gene was knocked down via siRNA, and fluorescence lifetime imaging microscopy and spot-variation fluorescence correlation spectroscopy was employed. RESULTS: EFR3A was identified as a candidate protein that interacts with flotillin-2. Moreover, this newly discovered interaction was demonstrated via overlay assay using recombinant EFR3A and flotillin-2. EFR3A is a stable component of the detergent-resistant membrane fraction of HeLa cells, and its presence was sensitive to the removal of cholesterol. While silencing the EFR3A gene, we observed decreased order of the plasma membrane of living cells or giant plasma membrane vesicles derived from knocked down cells and altered mobility of the raft probe, as indicated via fluorescence lifetime imaging microscopy and spot-variation fluorescence correlation spectroscopy. Moreover, silencing of EFR3A expression was found to disturb epidermal growth factor receptor and phospholipase C gamma phosphorylation and affect epidermal growth factor-dependent cytosolic Ca2+ concentration. CONCLUSIONS: Altogether, our results suggest hitherto unreported flotillin-2-EFR3A interaction, which might be responsible for membrane raft organization and regulation. This implies participation of this interaction in the regulation of multiple cellular processes, including those connected with cell signaling which points to the possible role in human health, in particular human cancer biology.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Microdominios de Membrana , Proteínas de la Membrana , Humanos , Membrana Celular/metabolismo , Factor de Crecimiento Epidérmico , Células HeLa , Unión Proteica , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de la Membrana/metabolismo
3.
Int J Mol Sci ; 23(24)2022 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-36555705

RESUMEN

Cell migration is an essential part of the complex and multistep process that is the development of cancer, a disease that is the second most common cause of death in humans. An important factor promoting the migration of cancer cells is TNF-α, a pro-inflammatory cytokine that, among its many biological functions, also plays a major role in mediating the expression of MMP9, one of the key regulators of cancer cell migration. It is also known that TNF-α is able to induce the Warburg effect in some cells by increasing glucose uptake and enhancing the expression and activity of lactate dehydrogenase subunit A (LDHA). Therefore, the aim of the present study was to investigate the interrelationship between the TNF-α-induced promigratory activity of cancer cells and their glucose metabolism status, using esophageal cancer cells as an example. By inhibiting LDHA activity with sodium oxamate (SO, also known as aminooxoacetic acid sodium salt or oxamic acid sodium salt) or siRNA-mediated gene silencing, we found using wound healing assay and gelatin zymography that LDHA downregulation impairs TNF-α-dependent tumor cell migration and significantly reduces TNF-α-induced MMP9 expression. These effects were associated with disturbances in the activation of the ERK1/2 signaling pathway, as we observed by Western blotting. We also reveal that in esophageal cancer cells, SO effectively reduces the production of lactic acid, which, as we have shown, synergizes the stimulating effect of TNF-α on MMP9 expression. In conclusion, our findings identified LDHA as a regulator of TNF-α-induced cell migration in esophageal cancer cells by the ERK1/2 signaling pathway, suggesting that LDHA inhibitors that limit the migration of cancer cells caused by the inflammatory process may be considered as an adjunct to standard therapy in esophageal cancer patients.


Asunto(s)
Neoplasias Esofágicas , Factor de Necrosis Tumoral alfa , Humanos , Lactato Deshidrogenasa 5 , Factor de Necrosis Tumoral alfa/farmacología , L-Lactato Deshidrogenasa/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/farmacología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular
4.
Cancers (Basel) ; 14(21)2022 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-36358740

RESUMEN

In 2022, prostate cancer (PCa) is estimated to be the most commonly diagnosed cancer in men in the United States-almost 270,000 American men are estimated to be diagnosed with PCa in 2022. This review compares and contrasts in vivo models of PCa with regards to the altered genes, signaling pathways, and stages of tumor progression associated with each model. The main type of model included in this review are genetically engineered mouse models, which include conditional and constitutive knockout model. 2D cell lines, 3D organoids and spheroids, xenografts and allografts, and patient derived models are also included. The major applications, advantages and disadvantages, and ease of use and cost are unique to each type of model, but they all make it easier to translate the tumor progression that is seen in the mouse prostate to the human prostate. Although both human and mouse prostates are androgen-dependent, the fact that the native, genetically unaltered prostate in mice cannot give rise to carcinoma is an especially critical component of PCa models. Thanks to the similarities between the mouse and human genome, our knowledge of PCa has been expanded, and will continue to do so, through models of PCa.

5.
Cancers (Basel) ; 14(7)2022 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-35406619

RESUMEN

Having the capability to proteolyze diverse structural and signaling proteins, matrix metalloproteinase 9 (MMP9), one of the best-studied secretory endopeptidases, has been identified as a crucial mediator of processes closely associated with tumorigenesis, such as the extracellular matrix reorganization, epithelial to mesenchymal transition, cell migration, new blood vessel formation, and immune response. In this review, we present the current state of knowledge on MMP9 and its role in cancer growth in the context of cell adhesion/migration, cancer-related inflammation, and tumor microenvironment formation. We also summarize recent achievements in the development of selective MMP9 inhibitors and the limitations of using them as anticancer drugs.

6.
Int J Mol Sci ; 22(4)2021 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-33572115

RESUMEN

Inhibition of the protein neddylation process by the small-molecule inhibitor MLN4924 has been recently indicated as a promising direction for cancer treatment. However, the knowledge of all biological consequences of MLN4924 for cancer cells is still incomplete. Here, we report that MLN4924 inhibits tumor necrosis factor-alpha (TNF-α)-induced matrix metalloproteinase 9 (MMP9)-driven cell migration. Using real-time polymerase chain reaction (PCR) and gelatin zymography, we found that MLN4924 inhibited expression and activity of MMP9 at the messenger RNA (mRNA) and protein levels in both resting cells and cells stimulated with TNF-α, and this inhibition was closely related to impaired cell migration. We also revealed that MLN4924, similar to TNF-α, induced phosphorylation of inhibitor of nuclear factor kappa B-alpha (IκB-α). However, contrary to TNF-α, MLN4924 did not induce IκB-α degradation in treated cells. In coimmunoprecipitation experiments, nuclear IκB-α which formed complexes with nuclear factor kappa B p65 subunit (NFκB/p65) was found to be highly phosphorylated at Ser32 in the cells treated with MLN4924, but not in the cells treated with TNF-α alone. Moreover, in the presence of MLN4924, nuclear NFκB/p65 complexes were found to be enriched in c-Jun and cyclin dependent kinase inhibitor 1 A (CDKN1A/p21) proteins. In these cells, NFκB/p65 was unable to bind to the MMP9 gene promoter, which was confirmed by the chromatin immunoprecipitation (ChIP) assay. Taken together, our findings identified MLN4924 as a suppressor of TNF-α-induced MMP9-driven cell migration in esophageal squamous cell carcinoma (ESCC), likely acting by affecting the nuclear ubiquitin-proteasome system that governs NFκB/p65 complex formation and its DNA binding activity in regard to the MMP9 promoter, suggesting that inhibition of neddylation might be a new therapeutic strategy to prevent invasion/metastasis in ESCC patients.


Asunto(s)
Ciclopentanos/farmacología , Neoplasias Esofágicas/tratamiento farmacológico , Carcinoma de Células Escamosas de Esófago/tratamiento farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Metaloproteinasa 9 de la Matriz/genética , Pirimidinas/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Ciclopentanos/uso terapéutico , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/genética , Carcinoma de Células Escamosas de Esófago/patología , Humanos , Proteína NEDD8/metabolismo , Inhibidor NF-kappaB alfa , Invasividad Neoplásica/patología , Invasividad Neoplásica/prevención & control , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Procesamiento Proteico-Postraduccional/genética , Pirimidinas/uso terapéutico , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Enzimas Activadoras de Ubiquitina/antagonistas & inhibidores , Enzimas Activadoras de Ubiquitina/metabolismo
7.
Ann Transl Med ; 8(14): 902, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32793746

RESUMEN

The invasive nature of many cancer cells involves the formation of F-actin-based, lipid-raft-enriched membrane protrusions known as invadopodia or, more broadly, invadosomes. Invadopodia are specialized adhesive structures arising from ventral cell surface within cell-extracellular matrix (ECM) contacts and concentrate high proteolytic activities that allow cells to overcome the dense scaffold of local microenvironment, comprising a natural barrier to cell spreading. This degradative activity distinguishes invadopodia from other adhesive structures like focal adhesions, lamellipodia or filopodia, and is believed to drive cancer progression.

9.
Cell Commun Signal ; 17(1): 120, 2019 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-31530281

RESUMEN

BACKGROUND: Prostate cancer development involves various mechanisms, which are poorly understood but pointing to epithelial mesenchymal transition (EMT) as the key mechanism in progression to metastatic disease. ABI1, a member of WAVE complex and actin cytoskeleton regulator and adaptor protein, acts as tumor suppressor in prostate cancer but the role of ABI1 in EMT is not clear. METHODS: To investigate the molecular mechanism by which loss of ABI1 contributes to tumor progression, we disrupted the ABI1 gene in the benign prostate epithelial RWPE-1 cell line and determined its phenotype. Levels of ABI1 expression in prostate organoid tumor cell lines was evaluated by Western blotting and RNA sequencing. ABI1 expression and its association with prostate tumor grade was evaluated in a TMA cohort of 505 patients and metastatic cell lines. RESULTS: Low ABI1 expression is associated with biochemical recurrence, metastasis and death (p = 0.038). Moreover, ABI1 expression was significantly decreased in Gleason pattern 5 vs. pattern 4 (p = 0.0025) and 3 (p = 0.0012), indicating an association between low ABI1 expression and highly invasive prostate tumors. Disruption of ABI1 gene in RWPE-1 cell line resulted in gain of an invasive phenotype, which was characterized by a loss of cell-cell adhesion markers and increased migratory ability of RWPE-1 spheroids. Through RNA sequencing and protein expression analysis, we discovered that ABI1 loss leads to activation of non-canonical WNT signaling and EMT pathways, which are rescued by re-expression of ABI1. Furthermore, an increase in STAT3 phosphorylation upon ABI1 inactivation and the evidence of a high-affinity interaction between the FYN SH2 domain and ABI1 pY421 support a model in which ABI1 acts as a gatekeeper of non-canonical WNT-EMT pathway activation downstream of the FZD2 receptor. CONCLUSIONS: ABI1 controls prostate tumor progression and epithelial plasticity through regulation of EMT-WNT pathway. Here we discovered that ABI1 inhibits EMT through suppressing FYN-STAT3 activation downstream from non-canonical WNT signaling thus providing a novel mechanism of prostate tumor suppression.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Carcinogénesis/genética , Proteínas del Citoesqueleto/deficiencia , Proteínas del Citoesqueleto/genética , Transición Epitelial-Mesenquimal/genética , Técnicas de Inactivación de Genes , Neoplasias de la Próstata/patología , Vía de Señalización Wnt/genética , Cadherinas/metabolismo , Adhesión Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Receptores Frizzled/metabolismo , Humanos , Masculino , Clasificación del Tumor , Fenotipo , Recurrencia , Factor de Transcripción STAT3/metabolismo , Regulación hacia Arriba/genética , beta Catenina/metabolismo
10.
Folia Histochem Cytobiol ; 57(2): 43-55, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31099889

RESUMEN

Biological membranes are organized in various microdomains, one of the best known being called membrane rafts. The major function of these is thought to organize signaling partners into functional complexes. An important protein found in membrane raft microdomains of erythroid and other blood cells is MPP1 (membrane palmitoylated protein 1)/p55. MPP1 (p55) belongs to the MAGUK (membrane-associated guanylate kinase homolog) family and it is a major target of palmitoylation in the red blood cells (RBCs) membrane. The well-known function of this protein is to participate in formation of the junctional complex of the erythrocyte mem-brane skeleton. However, its function as a "raft organizer" is not well understood. In this review we focus on recent reports concerning MPP1 participation in membrane rafts organization in erythroid cells, including its role in signal transduction. Currently it is not known whether MPP1 could have a similar role in cell types other than erythroid lineage. We present also preliminary data regarding the expression level of MPP1 gene in several non-erythroid cell lines.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Membrana Celular/metabolismo , Eritrocitos/metabolismo , Microdominios de Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Sanguíneas/genética , Colesterol/metabolismo , Humanos , Fluidez de la Membrana/fisiología , Lípidos de la Membrana/metabolismo , Proteínas de la Membrana/genética , Unión Proteica
12.
Blood ; 132(19): 2053-2066, 2018 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-30213875

RESUMEN

Although the pathogenesis of primary myelofibrosis (PMF) and other myeloproliferative neoplasms (MPNs) is linked to constitutive activation of the JAK-STAT pathway, JAK inhibitors have neither curative nor MPN-stem cell-eradicating potential, indicating that other targetable mechanisms are contributing to the pathophysiology of MPNs. We previously demonstrated that Abelson interactor 1 (Abi-1), a negative regulator of Abelson kinase 1, functions as a tumor suppressor. Here we present data showing that bone marrow-specific deletion of Abi1 in a novel mouse model leads to development of an MPN-like phenotype resembling human PMF. Abi1 loss resulted in a significant increase in the activity of the Src family kinases (SFKs), STAT3, and NF-κB signaling. We also observed impairment of hematopoietic stem cell self-renewal and fitness, as evidenced in noncompetitive and competitive bone marrow transplant experiments. CD34+ hematopoietic progenitors and granulocytes from patients with PMF showed decreased levels of ABI1 transcript as well as increased activity of SFKs, STAT3, and NF-κB. In aggregate, our data link the loss of Abi-1 function to hyperactive SFKs/STAT3/NF-κB signaling and suggest that this signaling axis may represent a regulatory module involved in the molecular pathophysiology of PMF.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Médula Ósea/patología , Proteínas del Citoesqueleto/genética , Eliminación de Gen , Mielofibrosis Primaria/genética , Mielofibrosis Primaria/patología , Animales , Médula Ósea/metabolismo , Autorrenovación de las Células , Células Cultivadas , Regulación hacia Abajo , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , FN-kappa B/metabolismo , Mielofibrosis Primaria/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Familia-src Quinasas/metabolismo
13.
Cell Signal ; 47: 27-36, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29588220

RESUMEN

Metastasis is the leading cause of mortality in patients with highly invasive cancers and, as such, is a major problem for medicine. It has been increasingly recognized that cancer-related inflammation plays an important role in promoting invasion and the metastatic process in which cell motility and upregulation of proteolytic enzymes are crucial events. TNFα is a proinflammatory cytokine known to stimulate synthesis of MMP9, a zinc- and calcium-dependent endopeptidase contributing to the regulation of ECM remodeling and cell signaling. However, the precise molecular mechanism of TNFα-induced MMP9 gene expression in cancers is still not fully understood. This study shows that TNFα-induced cell migration and invasion involve ERK1/2-dependent up-regulation of CDKN1A/p21 expression in highly aggressive breast cancer cells and that CDKN1A/p21 plays an important regulatory role in TNFα-induced MMP9 gene expression, indicating an unknown function of CDKN1A/p21 as a regulator of proteolytic activity in cancer cells.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/antagonistas & inhibidores , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Femenino , Humanos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación/efectos de los fármacos , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología
14.
J Urol ; 196(6): 1788-1795, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27177425

RESUMEN

PURPOSE: We describe a novel method of urethral stricture treatment using liquid buccal mucosal grafts to augment direct vision internal urethrotomy. MATERIALS AND METHODS: A rabbit stricture model was used to test this method. In phase 1 the concept of endoscopic liquid buccal mucosal graft implantation was tested by performing direct vision internal urethrotomy in 3 rabbits with immediate intraurethral injection of autologous liquid buccal mucosal grafts suspended in fibrin glue. Animals were sacrificed at 2 to 3 weeks and the urethras were examined for the presence of buccal mucosa engraftment. In phase 2 strictures were induced by electroresection in 9 rabbits divided into 2 groups, including 1) 6 rabbits treated with direct vision internal urethrotomy and liquid buccal mucosal grafts, and 2) 3 controls that underwent direct vision internal urethrotomy and injection of fibrin glue only. Two treated and 1 control animals were sacrificed at 8, 16 and 24 weeks each. Prior to sacrifice the animals underwent retrograde urethrograms and urethroscopy. Histological specimens were examined for the presence of buccal mucosal engraftment. RESULTS: In phase 1, 2 of the 3 rabbits demonstrated engraftment of buccal mucosa in the urethra after injection of liquid buccal mucosal grafts. In phase 2 all 6 treated animals demonstrated engraftment with resolution/improvement of strictures on retrograde urethrograms and urethroscopy. Controls had no buccal engraftment and showed fibrosis and chronic inflammation. One of the 3 controls had persistent stricture on retrograde urethrograms and cystoscopy. CONCLUSIONS: This proof of concept study demonstrated the feasibility of using liquid buccal mucosal grafts for endoscopic urethral stricture repair. Such a method may allow for wide application of this novel concept of using liquid buccal mucosal grafts to augment direct vision internal urethrotomy.


Asunto(s)
Cistoscopía/métodos , Mucosa Bucal/trasplante , Estrechez Uretral/cirugía , Procedimientos Quirúrgicos Urológicos Masculinos/métodos , Animales , Modelos Animales de Enfermedad , Masculino , Conejos , Trasplante Autólogo , Uretra/cirugía
15.
Cell Oncol (Dordr) ; 39(4): 353-63, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27042827

RESUMEN

PURPOSE: Tumor progression is associated with cell migration, invasion and metastasis. These processes are accompanied by the activation of specific proteases that are either linked to cellular membranes or are secreted into extracellular spaces. TNF-α is known to play an important role in various aspects of tumor progression. The aim of this work was to assess the effect of TNF-α on the migration of breast cancer cells and, in addition, to assess its association with the location of membrane-associated proteases in lipid rafts. METHODS: Wound scratch healing and Transwell migration assays were used to study the effect of TNF-α on the migration of both hormone-dependent and hormone-independent breast cancer-derived cells, i.e., MCF7 and MDA-MB-231, respectively. The expression and secretion of three matrix metalloproteases, MMP9, MMP2 and MT1-MMP, and two dipeptidyl peptidases, CD26 and FAP-α, was investigated using RT-PCR, Western blotting and gelatin zymography. In addition, activation of the MAPK/ERK signaling pathway was investigated by Western blotting. RESULTS: We found that a TNF-α-induced enhancement of breast cancer cell migration was accompanied by an increased secretion of MMP9, but not MMP2, into the culture media. We also found that TNF-α upregulated the expression of the dipeptidyl peptidases CD26 and FAP-α in a dose-dependent manner and, in addition, enhanced the concentration of all five proteases in lipid rafts in the breast cancer-derived cells tested, regardless of cell type. Furthermore, we found that TNF-α activated the MAPK/ERK signaling pathway by increasing the ERK1/2 phosphorylation level. Application of the MEK/ERK1/2 inhibitor U-0126 resulted in down-regulation of TNF-α-induced MMP9 secretion and abrogation of the enhanced concentration of proteases in the lipid rafts. CONCLUSIONS: From our results we conclude that TNF-α-induced activation of the MAPK/ERK signaling pathway may promote breast cancer cell migration via both upregulation of MMP9, CD26 and FAP-α and concentration of these proteases, as also MT1-MMP and MMP2, in the lipid rafts. TNF-α may serve as a potential therapeutic target in breast cancers susceptible to TNF-α stimulation.


Asunto(s)
Neoplasias de la Mama/patología , Movimiento Celular , Microdominios de Membrana/enzimología , Péptido Hidrolasas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Western Blotting , Neoplasias de la Mama/enzimología , Línea Celular Tumoral , Femenino , Humanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/fisiología
16.
Cancer Lett ; 358(1): 1-7, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25528630

RESUMEN

A hallmark of most cancer cells is an altered metabolism involving a shift to aerobic glycolysis with lactate production coupled with a higher uptake of glucose as the main source of energy. Lactate dehydrogenase 5 (LDH-5) catalyzes the reduction of pyruvate by NADH to form lactate, thus determining the availability of NAD(+) to maintain the continuity of glycolysis. It is therefore an important control point in the system of cellular energy release. Its upregulation is common in many malignant tumors. Inhibiting LDH-5 activity has an anti-proliferative effect on cancer cells. It may reverse their resistance to conventional chemo- and radiotherapy. Recent research has renewed interest in LDH-5 as an anticancer drug target. This review summarizes recent studies exploring the role of LDH-5 in cancer growth, its utility as a tumor marker, and developments made in identifying and designing anti-LDH-5 therapeutic agents.


Asunto(s)
Carcinogénesis/genética , Glucólisis/genética , L-Lactato Deshidrogenasa/metabolismo , Neoplasias/enzimología , Ciclo del Ácido Cítrico/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Isoenzimas/biosíntesis , Isoenzimas/genética , Isoenzimas/metabolismo , L-Lactato Deshidrogenasa/biosíntesis , L-Lactato Deshidrogenasa/genética , Lactato Deshidrogenasa 5 , Terapia Molecular Dirigida , Neoplasias/patología , Neoplasias/terapia
17.
Oncol Rep ; 31(6): 2820-6, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24789592

RESUMEN

To better understand the role of membrane-associated proteolytic systems in the development of esophageal cancer, we studied the expression of two serine proteases, fibroblast activation protein-α (FAP-α) and dipeptidyl peptidase IV (DPPIV) and three metalloproteinases, matrix metalloproteinase (MMP)-2, MMP-9 and MT1-MMP in 24 primary esophageal squamous cell carcinoma (ESCC) tissues and paired non-cancer tissues. Using reverse-transcription PCR, western blotting and zymography, we showed that both serine proteases and all three metalloproteinases were highly altered in ESCC. A positive correlation between the expression of FAP-α and DPPIV and the activity of both gelatinases was found. This may indicate that these proteolytic systems are tightly linked to each other and collectively are involved in the process of ECM degradation that facilitates cancer cell invasion and metastasis.


Asunto(s)
Carcinoma de Células Escamosas/genética , Dipeptidil Peptidasa 4/biosíntesis , Neoplasias Esofágicas/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 13/biosíntesis , Activación Transcripcional/genética , Anciano , Carcinoma de Células Escamosas/patología , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Metaloproteinasa 14 de la Matriz/biosíntesis , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/biosíntesis , Persona de Mediana Edad
18.
Cell Mol Biol Lett ; 19(1): 158-79, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24569979

RESUMEN

The spectrin-based membrane skeleton is crucial for the mechanical stability and resilience of erythrocytes. It mainly contributes to membrane integrity, protein organization and trafficking. Two transmembrane protein macro-complexes that are linked together by spectrin tetramers play a crucial role in attaching the membrane skeleton to the cell membrane, but they are not exclusive. Considerable experimental data have shown that direct interactions between spectrin and membrane lipids are important for cell membrane cohesion. Spectrin is a multidomain, multifunctional protein with several distinctive structural regions, including lipid-binding sites within CH tandem domains, a PH domain, and triple helical segments, which are excellent examples of ligand specificity hidden in a regular repetitive structure, as recently shown for the ankyrin-sensitive lipid-binding domain of beta spectrin. In this review, we summarize the state of knowledge about interactions between spectrin and membrane lipids.


Asunto(s)
Membrana Eritrocítica/metabolismo , Lípidos de la Membrana/metabolismo , Fosfolípidos/metabolismo , Espectrina/metabolismo , Citoesqueleto de Actina/metabolismo , Ancirinas , Sitios de Unión , Membrana Eritrocítica/genética , Humanos , Fosfolípidos/genética , Unión Proteica , Estructura Terciaria de Proteína , Espectrina/genética
19.
Biochim Biophys Acta ; 1845(2): 155-65, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24480320

RESUMEN

Membrane rafts are distinct plasma membrane microdomains that are enriched in sphingolipids and cholesterol. They organize receptors and their downstream molecules and regulate a number of intracellular signaling pathways. This review presents information on the dependence of several growth factor receptor signaling pathways on membrane rafts. It also discusses the involvement of rafts in the regulation of differentiation, apoptosis and cell migration connected with invasiveness and metastasis. Examples of known synthetic and naturally occurring substances that are known to affect lateral membrane organization in tumor cell growth are discussed as potential or actual therapeutics.


Asunto(s)
Microdominios de Membrana/genética , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Apoptosis/genética , Diferenciación Celular/genética , Movimiento Celular/genética , Humanos , Microdominios de Membrana/metabolismo , Neoplasias/patología , Transducción de Señal
20.
Biochim Biophys Acta ; 1838(2): 620-34, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23673272

RESUMEN

This review focuses on structure and functions of spectrin as a major component of the membrane skeleton. Recent advances on spectrin function as an interface for signal transduction mediation and a number of data concerning interaction of spectrin with membrane channels, adhesion molecules, receptors and transporters draw a picture of multifaceted protein. Here, we attempted to show the current depiction of multitask role of spectrin in cell physiology. This article is part of a Special Issue entitled: Reciprocal influences between cell cytoskeleton and membrane channels, receptors and transporters. Guest Editor: Jean Claude Hervé.


Asunto(s)
Canales Iónicos/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Receptores de Superficie Celular/metabolismo , Espectrina/metabolismo , Animales , Humanos , Canales Iónicos/química , Proteínas de Transporte de Membrana/química , Receptores de Superficie Celular/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...