Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 120(23): e2122053120, 2023 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-37252969

RESUMEN

The causes and consequences of abnormal biogenesis of extracellular vesicles (EVs) are not yet well understood in malignancies, including in breast cancers (BCs). Given the hormonal signaling dependence of estrogen receptor-positive (ER+) BC, we hypothesized that 17ß-estradiol (estrogen) might influence EV production and microRNA (miRNA) loading. We report that physiological doses of 17ß-estradiol promote EV secretion specifically from ER+ BC cells via inhibition of miR-149-5p, hindering its regulatory activity on SP1, a transcription factor that regulates the EV biogenesis factor nSMase2. Additionally, miR-149-5p downregulation promotes hnRNPA1 expression, responsible for the loading of let-7's miRNAs into EVs. In multiple patient cohorts, we observed increased levels of let-7a-5p and let-7d-5p in EVs derived from the blood of premenopausal ER+ BC patients, and elevated EV levels in patients with high BMI, both conditions associated with higher levels of 17ß-estradiol. In brief, we identified a unique estrogen-driven mechanism by which ER+ BC cells eliminate tumor suppressor miRNAs in EVs, with effects on modulating tumor-associated macrophages in the microenvironment.


Asunto(s)
Neoplasias de la Mama , Vesículas Extracelulares , MicroARNs , Humanos , Femenino , MicroARNs/genética , MicroARNs/metabolismo , Neoplasias de la Mama/patología , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Estradiol/farmacología , Estradiol/metabolismo , Estrógenos/metabolismo , Vesículas Extracelulares/genética , Vesículas Extracelulares/metabolismo , Microambiente Tumoral
2.
Pharmacol Res ; 192: 106757, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37023992

RESUMEN

The liver is a major organ that is involved in essential biological functions such as digestion, nutrient storage, and detoxification. Furthermore, it is one of the most metabolically active organs with active roles in regulating carbohydrate, protein, and lipid metabolism. Hepatocellular carcinoma is a cancer of the liver that is associated in settings of chronic inflammation such as viral hepatitis, repeated toxin exposure, and fatty liver disease. Furthermore, liver cancer is the most common cause of death associated with cirrhosis and is the 3rd leading cause of global cancer deaths. LKB1 signaling has been demonstrated to play a role in regulating cellular metabolism under normal and nutrient deficient conditions. Furthermore, LKB1 signaling has been found to be involved in many cancers with most reports identifying LKB1 to have a tumor suppressive role. In this review, we use the KMPlotter database to correlate RNA levels of LKB1 signaling genes and hepatocellular carcinoma patient survival outcomes with the hopes of identifying potential biomarkers clinical usage. Based on our results STRADß, CAB39L, AMPKα, MARK2, SIK1, SIK2, BRSK1, BRSK2, and SNRK expression has a statistically significant impact on patient survival.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Quinasas de la Proteína-Quinasa Activada por el AMP , Proteínas Quinasas Activadas por AMP/metabolismo , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo
3.
Mol Cancer ; 21(1): 138, 2022 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-35768871

RESUMEN

BACKGROUND: Triple-negative breast cancers (TNBCs) are clinically aggressive subtypes of breast cancer. TNBC is difficult to treat with targeted agents due to the lack of commonly targeted therapies within this subtype. Androgen receptor (AR) has been detected in 12-55% of TNBCs. AR stimulates breast tumor growth in the absence of estrogen receptor (ER), and it has become an emerging molecular target in TNBC treatment. METHODS: Ceritinib is a small molecule inhibitor of tyrosine kinase and it is used in the therapy of non-small lung cancer patients. Enzalutamide is a small molecule compound targeting the androgen receptor and it is used to treat prostate cancer. Combination therapy of these drugs were investigated using AR positive breast cancer mouse xenograft models. Also, combination treatment of ceritinib and paclitaxel investigated using AR- and AR low mouse xenograft and patient derived xenograft models. RESULTS: We screened 133 FDA approved drugs that have a therapeutic effect of AR+ TNBC cells. From the screen, we identified two drugs, ceritinib and crizotinib. Since ceritinib has a well- defined role in androgen independent AR signaling pathways, we further investigated the effect of ceritinib. Ceritinib treatment inhibited RTK/ACK/AR pathway and other downstream pathways in AR+ TNBC cells. The combination of ceritinib and enzalutamide showed a robust inhibitory effect on cell growth of AR+ TNBC cells in vitro and in vivo. Interestingly Ceritinib inhibits FAK-YB-1 signaling pathway that leads to paclitaxel resistance in all types of TNBC cells. The combination of paclitaxel and ceritinib showed drastic inhibition of tumor growth compared to a single drug alone. CONCLUSIONS: To improve the response of AR antagonist in AR positive TNBC, we designed a novel combinational strategy comprised of enzalutamide and ceritinib to treat AR+ TNBC tumors through the dual blockade of androgen-dependent and androgen-independent AR signaling pathways. Furthermore, we introduced a novel therapeutic combination of ceritinib and paclitaxel for AR negative or AR-low TNBCs and this combination inhibited tumor growth to a great extent. All agents used in our study are FDA-approved, and thus the proposed combination therapy will likely be useful in the clinic.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Andrógenos/uso terapéutico , Animales , Línea Celular Tumoral , Humanos , Ratones , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Pirimidinas , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Sulfonas , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo
4.
Int J Mol Sci ; 23(3)2022 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-35163298

RESUMEN

Nischarin (Nisch) is a cytosolic scaffolding protein that harbors tumor-suppressor-like characteristics. Previous studies have shown that Nisch functions as a scaffolding protein and regulates multiple biological activities. In the current study, we prepared a complete Nisch knockout model, for the first time, by deletion of exons 5 and 6. This knockout model was confirmed by Qrt-PCR and Western blotting with products from mouse embryonic fibroblast (MEF) cells. Embryos and adult mice of knockouts are significantly smaller than their wild-type counterparts. Deletion of Nisch enhanced cell migration, as demonstrated by wound type and transwell migration assays. Since the animals were small in size, we investigated Nisch's effect on metabolism by conducting several assays using the Seahorse analyzer system. These data indicate that Nisch null cells have lower oxygen consumption rates, lower ATP production, and lower levels of proton leak. We examined the expression of 15 genes involved in lipid and fat metabolism, as well as cell growth, and noted a significant increase in expression for many genes in Nischarin null animals. In summary, our results show that Nischarin plays an important physiological role in metabolic homeostasis.


Asunto(s)
Adenosina Trifosfato/metabolismo , Receptores de Imidazolina/metabolismo , Consumo de Oxígeno/genética , Adenosina Trifosfato/genética , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Respiración de la Célula , Fibroblastos , Expresión Génica/genética , Receptores de Imidazolina/genética , Péptidos y Proteínas de Señalización Intracelular , Metabolismo de los Lípidos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estrés Oxidativo , Consumo de Oxígeno/fisiología
5.
Int J Cancer ; 146(9): 2576-2587, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-31525254

RESUMEN

Previously, our lab discovered the protein Nischarin and uncovered its role in regulating cell migration and invasion via its interactions with several proteins. We subsequently described a role for Nischarin in breast cancer, in which it is frequently underexpressed. To characterize Nischarin's role in breast tumorigenesis and mammary gland development more completely, we deleted a critical region of the Nisch gene (exons 7-10) from the mouse genome and observed the effects. Mammary glands in mutant animals showed delayed terminal end bud formation but did not develop breast tumors spontaneously. Therefore, we interbred the animals with transgenic mice expressing the mouse mammary tumor virus-polyoma middle T-antigen (MMTV-PyMT) oncogene. The MMTV-PyMT mammary glands lacking Nischarin showed increased hyperplasia compared to wild-type animal tissues. Furthermore, we observed significantly increased tumor growth and metastasis in Nischarin mutant animals. Surprisingly, Nischarin deletion decreased activity of AMPK and subsequently its downstream effectors. Given this finding, we treated these animals with metformin, which enhances AMPK activity. Here, we show for the first time, metformin activates AMPK signaling and inhibits tumor growth of Nischarin lacking PyMT tumors suggesting a potential use for metformin as a cancer therapeutic, particularly in the case of Nischarin-deficient breast cancers.


Asunto(s)
Transformación Celular Neoplásica/patología , Receptores de Imidazolina/fisiología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Animales/tratamiento farmacológico , Neoplasias Mamarias Animales/patología , Metformina/farmacología , Animales , Antígenos Transformadores de Poliomavirus/genética , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/metabolismo , Femenino , Hipoglucemiantes/farmacología , Neoplasias Pulmonares/metabolismo , Neoplasias Mamarias Animales/metabolismo , Virus del Tumor Mamario del Ratón/genética , Ratones , Ratones Noqueados , Invasividad Neoplásica
6.
Anticancer Drugs ; 31(4): 359-367, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31917699

RESUMEN

Triple-negative breast cancers account for approximately 15-20% of breast cancer patients. Due to lack of expression of estrogen receptor, PR and human epidermal growth factor receptor 2 in triple-negative breast cancers, there are no targeted therapies available for these cancers. Therefore, a major research priority is to find potential therapeutic targets. Androgen receptor is present in 80-90% of all breast cancers, including 55% of estrogen receptor-α-negative cancers and 12%-35% of triple-negative breast cancers. Androgen receptor stimulates growth and survival in triple-negative breast cancer cells. Treatment with bicalutamide, an androgen receptor antagonist, has a good benefit for AR triple-negative breast cancer patients. AR triple-negative breast cancer cells were treated with curcumin or bicalutamide alone or in combination of both together. Cell growth, apoptosis and Wnt signaling pathways were examined. We found that curcumin dramatically suppressed Wnt signaling pathway in AR triple-negative breast cancer cells. Curcumin treatment inhibited androgen receptor protein expression in AR triple-negative breast cancer cells. Combination treatment of curcumin and bicalutamide has a robust increase in apoptosis. Furthermore, the combination treatment suppressed the growth of AR triple-negative breast cancer cells more effectively than with the single drug alone. Our data indicate that androgen receptor inhibition is a potential therapeutic approach for AR triple-negative breast cancers. In summary, our study for the first time shows that the combination treatment of curcumin and bicalutamide is effective in AR triple-negative breast cancer cells.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Animales , Apoptosis , Biomarcadores de Tumor , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Ciclo Celular , Movimiento Celular , Proliferación Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Pronóstico , Tasa de Supervivencia , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Mol Cancer ; 18(1): 75, 2019 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-30940145

RESUMEN

Tumor-derived exosomes (TDEs) participate in formation and progression of different cancer processes, including tumor microenvironment (TME) remodeling, angiogenesis, invasion, metastasis and drug-resistance. Exosomes initiate or suppress various signaling pathways in the recipient cells via transmitting heterogeneous cargoes. In this review we discuss exosome biogenesis, exosome mediated metastasis and chemoresistance. Furthermore, tumor derived exosomes role in tumor microenvironment remodeling, and angiogenesis is reviewed. Also, exosome induction of epithelial mesenchymal transition (EMT) is highlighted. More importantly, we discuss extensively how exosomes regulate drug resistance in several cancers. Thus, understanding exosome biogenesis, their contents and the molecular mechanisms and signaling pathways that are responsible for metastasis and drug-resistance mediated by TDEs may help to devise novel therapeutic approaches for cancer progression particularly to overcome therapy-resistance and preventing metastasis as major factors of cancer mortality.


Asunto(s)
Resistencia a Antineoplásicos , Exosomas/metabolismo , Neoplasias/metabolismo , Progresión de la Enfermedad , Humanos , Metástasis de la Neoplasia , Transducción de Señal , Microambiente Tumoral
8.
Int J Obes (Lond) ; 43(5): 1046-1057, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30546133

RESUMEN

BACKGROUND/OBJECTIVES: NISCH-STAB1 is a newly identified locus correlated to human waist-hip ratio (WHR), which is a risk indicator of developing obesity-associated diabetes. Our previous studies have shown that Nisch mutant male mice increased glucose tolerance in chow-fed conditions. Thus we hypothesized that Nisch mutant mice will have changes in insulin resistance, adipocytes, hepatic steatosis when mice are fed with high-fat diet (HFD). METHODS: Insulin resistance was assessed in Nisch mutant mice and WT mice fed with high-fat diet (60% by kCal) or chow diet. Whole-body energy metabolism was examined using an indirect calorimeter. Adipose depots including inguinal white adipose tissue (WAT), perigonadal WAT, retroperitoneal WAT, and mesenteric WAT were extracted. Area and eqdiameter of each adipocyte were determined, and insulin signaling was examined as well. Paired samples of subcutaneous and omental visceral adipose tissue were obtained from 400 individuals (267 women, 133 men), and examined the expression of Nischarin. RESULTS: We found that insulin signaling was impaired in major insulin-sensitive tissues of Nisch mutant female mice. When mice were fed with HFD for 15 weeks, the Nisch mutant female mice not only developed severe insulin resistance and decreased glucose tolerance compared with wild-type control mice, but also accumulated more white fat, had larger adipocytes and developed severe hepatic steatosis than wild-type control mice. To link our animal studies to human diseases, we further analyzed Nischarin expression in the paired human samples of visceral and subcutaneous adipose tissue from Caucasians. In humans, we found that Nischarin expression is attenuated in adipose tissue with obesity. More importantly, we found that Nischarin mRNA inversely correlated with parameters of obesity, fat distribution, lipid and glucose metabolism. CONCLUSIONS: Taken together, our data revealed sexual dimorphism of Nischarin in body fat distribution, insulin resistance, and glucose tolerance in mice.


Asunto(s)
Adipocitos/patología , Resistencia a la Insulina/fisiología , Grasa Intraabdominal/patología , Metabolismo de los Lípidos/fisiología , Mutación , Obesidad/fisiopatología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Distribución de la Grasa Corporal , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Femenino , Humanos , Receptores de Imidazolina/genética , Péptidos y Proteínas de Señalización Intracelular/fisiología , Masculino , Ratones , Persona de Mediana Edad , ARN Mensajero/fisiología , Caracteres Sexuales
9.
J Biol Chem ; 292(41): 16833-16846, 2017 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-28842496

RESUMEN

Nischarin (Nisch) is a key protein functioning as a molecular scaffold and thereby hosting interactions with several protein partners. To explore the physiological importance of Nisch, here we generated Nisch loss-of-function mutant mice and analyzed their metabolic phenotype. Nisch-mutant embryos exhibited delayed development, characterized by small size and attenuated weight gain. We uncovered the reason for this phenotype by showing that Nisch binds to and inhibits the activity of AMP-activated protein kinase (AMPK), which regulates energy homeostasis by suppressing anabolic and activating catabolic processes. The Nisch mutations enhanced AMPK activation and inhibited mechanistic target of rapamycin signaling in mouse embryonic fibroblasts as well as in muscle and liver tissues of mutant mice. Nisch-mutant mice also exhibited increased rates of glucose oxidation with increased energy expenditure, despite reduced overall food intake. Moreover, the Nisch-mutant mice had reduced expression of liver markers of gluconeogenesis associated with increased glucose tolerance. As a result, these mice displayed decreased growth and body weight. Taken together, our results indicate that Nisch is an important AMPK inhibitor and a critical regulator of energy homeostasis, including lipid and glucose metabolism.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Metabolismo Energético , Gluconeogénesis , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Metabolismo de los Lípidos , Hígado/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Animales , Línea Celular , Glucosa/genética , Glucosa/metabolismo , Humanos , Receptores de Imidazolina , Péptidos y Proteínas de Señalización Intracelular/genética , Hígado/patología , Ratones , Ratones Mutantes , Mutación , Oxidación-Reducción , Unión Proteica
10.
Mol Cancer ; 17(1): 100, 2018 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-30012170

RESUMEN

BACKGROUND: The disruption of normal gene regulation due to microRNA dysfunction is a common event in cancer pathogenesis. MicroRNA-27b is an example of an oncogenic miRNA, and it is frequently upregulated in breast cancer. MicroRNAs have been found to deregulate tumor metabolism, which typically manifests as heightened cellular glucose uptake in consort with increased flux through glycolysis, followed by the preferential conversion of glycolytic pyruvate into lactate (a phenomenon known as the Warburg Effect). Pyruvate Dehydrogenase, an enzyme complex linking glycolysis with downstream oxidative metabolism, represents a key location where regulation of metabolism occurs; PDHX is a key structural component of this complex and is essential for its function. METHODS: We sought to characterize the role of miR-27b in breast cancer by identifying novel transcripts under its control. We began by utilizing luciferase, RNA, and protein assays to establish PDHX as a novel target of miR-27b. We then tested whether miR-27b could alter metabolism using several metabolite assay kits and performed a seahorse analysis. We also examined how the altered metabolism might affect cell proliferation. Lastly, we confirmed the relevance of our findings in human breast tumor samples. RESULTS: Our data indicate that Pyruvate Dehydrogenase Protein X is a credible target of miR-27b in breast cancer. Mechanistically, by suppressing PDHX, miR-27b altered levels of pyruvate, lactate and citrate, as well as reducing mitochondrial oxidation and promoting extracellular acidification. These changes corresponded with an increased capacity for cell proliferation. In human breast tumor samples, PDHX expression was deficient, and low levels of PDHX were associated with reduced patient survival. CONCLUSIONS: MicroRNA-27b targets PDHX, resulting in an altered metabolic configuration that is better suited to fuel biosynthetic processes and cell proliferation, thereby promoting breast cancer progression.


Asunto(s)
Neoplasias de la Mama/genética , Regulación hacia Abajo , MicroARNs/genética , Complejo Piruvato Deshidrogenasa/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular , Ácido Cítrico/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Glucólisis , Humanos , Ácido Láctico/metabolismo , Células MCF-7 , Complejo Piruvato Deshidrogenasa/metabolismo , Ácido Pirúvico/metabolismo , Análisis de Supervivencia
11.
Mol Cancer ; 17(1): 21, 2018 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-29415725

RESUMEN

BACKGROUND: During metastasis, tumor cells move through the tracks of extracellular matrix (ECM). Focal adhesions (FAs) are the protein complexes that link the cell cytoskeleton to the ECM and their presence is necessary for cell attachment. The tumor suppressor Nischarin interacts with a number of signaling proteins such as Integrin α5, PAK1, LIMK1, LKB1, and Rac1 to prevent cancer cell migration. Although previous findings have shown that Nischarin exerts this migratory inhibition by interacting with other proteins, the effects of these interactions on the entire FA machinery are unknown. METHODS: RT-PCR, Western Blotting, invadopodia assays, and immunofluorescence were used to examine FA gene expression and determine whether Nischarin affects cell attachment, as well as the proteins that regulate it. RESULTS: Our data show that Nischarin prevents cell migration and invasion by altering the expression of key focal adhesion proteins. Furthermore, we have found that Nischarin-expressing cells have reduced ability to attach the ECM, which in turn leads to a decrease in invadopodia-mediated matrix degradation. CONCLUSIONS: These experiments demonstrate an important role of Nischarin in regulating cell attachment, which adds to our understanding of the early events of the metastatic process in breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Adhesión Celular/genética , Receptores de Imidazolina/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Podosomas/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Matriz Extracelular/metabolismo , Femenino , Expresión Génica , Regulación de la Expresión Génica , Humanos , Receptores de Imidazolina/metabolismo , Integrinas/genética , Integrinas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Modelos Biológicos , Podosomas/metabolismo
12.
EMBO J ; 32(5): 713-27, 2013 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-23386062

RESUMEN

The intracellular pathogenic bacterium Salmonella enterica serovar typhimurium (Salmonella) relies on acidification of the Salmonella-containing vacuole (SCV) for survival inside host cells. The transport and fusion of membrane-bound compartments in a cell is regulated by small GTPases, including Rac and members of the Rab GTPase family, and their effector proteins. However, the role of these components in survival of intracellular pathogens is not completely understood. Here, we identify Nischarin as a novel dual effector that can interact with members of Rac and Rab GTPase (Rab4, Rab14 and Rab9) families at different endosomal compartments. Nischarin interacts with GTP-bound Rab14 and PI(3)P to direct the maturation of early endosomes to Rab9/CD63-containing late endosomes. Nischarin is recruited to the SCV in a Rab14-dependent manner and enhances acidification of the SCV. Depletion of Nischarin or the Nischarin binding partners--Rac1, Rab14 and Rab9 GTPases--reduced the intracellular growth of Salmonella. Thus, interaction of Nischarin with GTPases may regulate maturation and subsequent acidification of vacuoles produced after phagocytosis of pathogens.


Asunto(s)
Endosomas/microbiología , Receptores de Imidazolina/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Salmonella typhimurium/crecimiento & desarrollo , Vacuolas/microbiología , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Transporte Biológico , Western Blotting , Endosomas/metabolismo , Guanosina Trifosfato/metabolismo , Células HeLa , Humanos , Receptores de Imidazolina/genética , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intracelular/genética , Lisosomas/metabolismo , Lisosomas/microbiología , Fosfatos de Fosfatidilinositol/metabolismo , Transporte de Proteínas , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Infecciones por Salmonella/microbiología , Técnicas del Sistema de Dos Híbridos , Vacuolas/metabolismo , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rac/genética
13.
Biomed Microdevices ; 19(4): 97, 2017 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-29103075

RESUMEN

Adherent cells produce cellular traction force (CTF) on a substrate to maintain their physical morphologies, sense external environment, and perform essential cellular functions. Precise characterization of the CTF can expand our knowledge of various cellular processes as well as lead to the development of novel mechanical biomarkers. However, current methods that measure CTF require special substrates and fluorescent microscopy, rendering them less suitable in a clinical setting. Here, we demonstrate a rapid and direct approach to measure the combined CTF of a large cell population using thin polydimethylsiloxane (PDMS) cantilevers. Cells attached to the top surface of the PDMS cantilever produce CTF, which causes the cantilever to bend. The side view of the cantilever was imaged with a low-cost camera to extract the CTF. We characterized the CTF of fibroblasts and breast cancer cells. In addition, we were able to directly measure the contractile force of a suspended cell sheet, which is similar to the CTF of the confluent cell layer before detachment. The demonstrated technique can provide rapid and real-time measurement of the CTF of a large cell population and can directly characterize its temporal dynamics. The developed thin film PDMS cantilever can be fabricated affordably and the CTF extraction technique does not require expensive equipment. Thus, we believe that the developed method can provide an easy-to-use and affordable platform for CTF characterization in clinical settings and laboratories.


Asunto(s)
Fenómenos Fisiológicos Celulares , Dimetilpolisiloxanos/química , Animales , Línea Celular Tumoral , Fibroblastos , Análisis de Elementos Finitos , Humanos , Procesamiento de Imagen Asistido por Computador , Ratones , Modelos Teóricos , Células 3T3 NIH
14.
Mol Cancer ; 15: 18, 2016 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-26905733

RESUMEN

The epithelial to mesenchymal transition (EMT) is a biological process in which a non-motile epithelial cell changes to a mesenchymal phenotype with invasive capacities. This phenomenon has been well documented in multiple biological processes including embryogenesis, fibrosis, tumor progression and metastasis. The hallmark of EMT is the loss of epithelial surface markers, most notably E-cadherin, and the acquisition of mesenchymal markers including vimentin and N-cadherin. The downregulation of E-cadherin during EMT can be mediated by its transcriptional repression through the binding of EMT transcription factors (EMT-TFs) such as SNAIL, SLUG and TWIST to E-boxes present in the E-cadherin promoter. Additionally, EMT-TFs can also cooperate with several enzymes to repress the expression of E-cadherin and regulate EMT at the epigenetic and post- translational level. In this review, we will focus on epigenetic and post- translational modifications that are important in EMT. In addition, we will provide an overview of the various therapeutic approaches currently being investigated to undermine EMT and hence, the metastatic progression of cancer as well.


Asunto(s)
Epigénesis Genética , Transición Epitelial-Mesenquimal/genética , Procesamiento Proteico-Postraduccional/genética , Biomarcadores de Tumor/genética , Cadherinas/metabolismo , Humanos , Modelos Biológicos
15.
J Biol Chem ; 288(22): 15495-509, 2013 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-23572524

RESUMEN

Biallelic inactivation of LKB1, a serine/threonine kinase, has been detected in 30% of lung adenocarcinomas, and inhibition of breast tumor growth has been demonstrated. We have identified the tumor suppressor, Nischarin, as a novel binding partner of LKB1. Our mapping analysis shows that the N terminus of Nischarin interacts with amino acids 44-436 of LKB1. Time lapse microscopy and Transwell migration data show that the absence of both Nischarin and LKB1 from an invasive breast cancer cell line (MDA-MB-231) enhances migration as measured by increased distance and speed of migrating cells. Our data suggest that this is a result of elevated PAK1 and LIMK1 phosphorylation. Moreover, the absence of Nischarin and LKB1 increased tumor growth in vivo. Consistent with this, the percentage of S phase cells was increased, as demonstrated by flow cytometry and enhanced cyclin D1. The absence of Nischarin and LKB1 also led to a dramatic increase in the formation of lung metastases. Our studies, for the first time, demonstrate functional interaction between LKB1 and Nischarin to inhibit cell migration and breast tumor progression. Mechanistically, we show that these two proteins together regulate PAK-LIMK-Cofilin and cyclin D1/CDK4 pathways.


Asunto(s)
Neoplasias de la Mama/metabolismo , Movimiento Celular , Células Epiteliales/metabolismo , Receptores de Imidazolina/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Glándulas Mamarias Humanas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Ciclina D1/genética , Ciclina D1/metabolismo , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 4 Dependiente de la Ciclina/metabolismo , Células Epiteliales/patología , Femenino , Humanos , Receptores de Imidazolina/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Quinasas Lim/genética , Quinasas Lim/metabolismo , Glándulas Mamarias Humanas/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Trasplante de Neoplasias , Proteínas Serina-Treonina Quinasas/genética , Trasplante Heterólogo , Proteínas Supresoras de Tumor/genética
16.
Oncogene ; 43(11): 763-775, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38310162

RESUMEN

Both breast cancer and obesity can regulate epigenetic changes or be regulated by epigenetic changes. Due to the well-established link between obesity and an increased risk of developing breast cancer, understanding how obesity-mediated epigenetic changes affect breast cancer pathogenesis is critical. Researchers have described how obesity and breast cancer modulate the epigenome individually and synergistically. In this review, the epigenetic alterations that occur in obesity, including DNA methylation, histone, and chromatin modification, accelerated epigenetic age, carcinogenesis, metastasis, and tumor microenvironment modulation, are discussed. Delineating the relationship between obesity and epigenetic regulation is vital to furthering our understanding of breast cancer pathogenesis.


Asunto(s)
Neoplasias de la Mama , Epigénesis Genética , Humanos , Femenino , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Metilación de ADN , Histonas/metabolismo , Obesidad/complicaciones , Obesidad/genética , Microambiente Tumoral/genética
17.
Mol Med ; 19: 253-62, 2013 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-23821363

RESUMEN

Although a relationship between PDZK1 expression and estrogen receptor (ER)-α stimulation has been suggested, the nature of such a connection and the function of PDZK1 in breast cancer remain unknown. Human tissue microarrays (cancer tissue: 262 cores; normal tissue: 87 cores) and breast cancer cell lines were used to conduct the study. We show that PDZK1 protein expression is tightly correlated with human breast malignancy, is negatively correlated with age and had no significant correlation with ER-α expression levels. PDZK1 exhibited an exclusive epithelial expression with mostly cytosolic subcellular localization. Additionally, 17ß-estradiol induced PDZK1 expression above its basal level more than 24 h after treatment in MCF-7 cells. PDZK1 expression was indirectly regulated by ER-α stimulation, requiring insulinlike growth factor 1 receptor (IGF-1R) expression and function. The molecular link between PDZK1 and IGF-1R was supported by a significant correlation between protein and mRNA levels (r = 0.591, p < 0.001, and r = 0.537, p < 0.001, respectively) of the two factors in two different cohorts of human breast cancer tissues. Interestingly, PDZK1 knockdown in MCF-7 cells blocked ER-dependent growth and reduced c-Myc expression, whereas ectopic expression of PDZK1 enhanced cell proliferation in the presence or absence of 17ß-estradiol potentially through an increase in c-Myc expression, suggesting that PDZK1 has oncogenic activity. PDKZ1 also appeared to interact with the Src/ER-α/epidermal growth factor receptor (EGFR) complex, but not with IGF-1R and enhanced EGFR-stimulated MEK/ERK1/2 signaling. Collectively, our results clarify the relationship between ER-α and PDZK1, propose a direct relationship between PDZK1 and IGF-1R, and identify a novel oncogenic activity for PDZK1 in breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas Portadoras/metabolismo , Receptor alfa de Estrógeno/metabolismo , Receptor IGF Tipo 1/metabolismo , Línea Celular Tumoral , Estrógenos/farmacología , Femenino , Humanos , Proteínas de la Membrana , Análisis de Matrices Tisulares
18.
Sci Rep ; 13(1): 11843, 2023 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-37481672

RESUMEN

Triple-negative breast cancers (TNBCs) are aggressive forms of breast cancer and tend to grow and spread more quickly than most other types of breast cancer. TNBCs can neither be targeted by hormonal therapies nor the antibody trastuzumab that targets the HER2 protein. There are urgent unmet medical needs to develop targeted drugs for TNBCs. We identified a small molecule NSC260594 from the NCI diversity set IV compound library. NSC260594 exhibited dramatic cytotoxicity in multiple TNBCs in a dose-and time-dependent manner. NSC260594 inhibited the Myeloid cell leukemia-1 (Mcl-1) expression through downregulation of Wnt signaling proteins. Consistent with this, NSC260594 treatment increased apoptosis, which was confirmed by using an Annexin-V/PI assay. Interestingly, NSC260594 treatment reduced the cancer stem cell (CSC) population in TNBCs. To make NSC260594 more clinically relevant, we treated NSC260594 with TNBC cell derived xenograft (CDX) mouse model, and with patient-derived xenograft (PDX) organoids. NSC260594 significantly suppressed MDA-MB-231 tumor growth in vivo, and furthermore, the combination treatment of NSC260594 and everolimus acted synergistically to decrease growth of TNBC PDX organoids. Together, we found that NSC260594 might serve as a lead compound for triple-negative breast cancer therapy through targeting Mcl-1.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama Triple Negativas , Animales , Humanos , Ratones , Anexina A5 , Anticuerpos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Modelos Animales de Enfermedad , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico
19.
Noncoding RNA ; 9(6)2023 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-38133210

RESUMEN

We are delighted to share with you our thirteenth Journal Club and highlight some of the most interesting papers published recently [...].

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA