Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
J Biol Chem ; 299(11): 105311, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37797694

RESUMEN

While the role of endocytosis in focal adhesion turnover-coupled cell migration has been established in addition to its conventional role in cellular functions, the molecular regulators and precise molecular mechanisms that underlie this process remain largely unknown. In this study, we report that proto-oncoprotein hematopoietic PBX-interacting protein (HPIP) localizes to focal adhesions as well as endosomal compartments along with RUN FYVE domain-containing protein 3 (RUFY3) and Rab5, an early endosomal protein. HPIP contains two coiled-coil domains (CC1 and CC2) that are necessary for its association with Rab5 and RUFY3 as CC domain double mutant, that is, mtHPIPΔCC1-2 failed to support it. Furthermore, we show that HPIP and RUFY3 activate Rab5 by serving as noncanonical guanine nucleotide exchange factors of Rab5. In support of this, either deletion of coiled-coil domains or silencing of HPIP or RUFY3 impairs Rab5 activation and Rab5-dependent cell migration. Mechanistic studies further revealed that loss of HPIP or RUFY3 expression severely impairs Rab5-mediated focal adhesion disassembly, FAK activation, fibronectin-associated-ß1 integrin trafficking, and thus cell migration. Together, this study underscores the importance of HPIP and RUFY3 as noncanonical guanine nucleotide exchange factors of Rab5 and in integrin trafficking and focal adhesion turnover, which implicates in cell migration.


Asunto(s)
Adhesiones Focales , Factores de Intercambio de Guanina Nucleótido , Movimiento Celular , Endocitosis , Adhesiones Focales/genética , Adhesiones Focales/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas de Unión al GTP rab5/genética , Proteínas de Unión al GTP rab5/metabolismo , Humanos , Línea Celular , Línea Celular Tumoral
2.
Cytokine ; 160: 156024, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36182709

RESUMEN

Cytokines are multifunctional glycoproteins that play a vital role in the tumor microenvironment and progression of breast cancer. Genetic polymorphisms may influence the immune responses restrained by pro- and anti-inflammatory cytokine expression in tumors. Hence, the present study evaluated the contribution of Interleukin (IL) 6 (rs1800797, rs1800796, and rs1800795) and IL18 (rs1946518, rs187238, and rs549908) genotypes and their haplotypes to the risk, progression of breast cancer in South Indian population. The polymorphisms of IL6 -597G > A, -572C > G & -174G > C, and IL18 -607C > A, -137G > C, 105A > T were genotyped through PCR-RFLP and As-PCR assays in the blood DNA of 600 subjects. We have performed haplotype, LD, univariate, multivariate logistic regression, and Kaplan-Meier analyses for the obtained data. The frequency of AA genotype & A-allele of IL6 -597G > A, and CC genotype & C-allele of IL6 -174G > C polymorphism was higher in breast cancer patients and was found to be significantly associated with late (advanced) stage, metastasis, etc. Further, IL18 -607C > A, -137G > C, and 105A > T polymorphisms were found to be associated with lobular carcinoma subtype, PgR -ve, and HER2 +ve breast cancer patients. In survival analysis, we have observed that the C-allele of IL6 -174G > C polymorphism to be significantly associated with 5 years of overall survival in breast cancer subjects. All SNPs of the IL6 and IL18 genes showed perfect LD; the G-C-C, A-G-G, and A-C-C haplotype combinations of IL6 gene conferred 2.09, 2.25, and 4.72 folds risk for breast cancer respectively. Hence, our results suggest the importance of genotypic and haplotype analysis of IL6 and IL18 gene variants in the progression and risk prediction of breast cancer.


Asunto(s)
Neoplasias de la Mama , Interleucina-18/genética , Interleucina-6/genética , Neoplasias de la Mama/genética , Citocinas/genética , ADN , Femenino , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Genotipo , Haplotipos/genética , Humanos , Polimorfismo de Nucleótido Simple/genética , Microambiente Tumoral
3.
Biochim Biophys Acta Rev Cancer ; 1877(5): 188785, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36031088

RESUMEN

Metabolic reprogramming is a unique but complex biochemical adaptation that allows solid tumors to tolerate various stresses that challenge cancer cells for survival. Under conditions of metabolic stress, mammalian cells employ adenosine monophosphate (AMP)-activated protein kinase (AMPK) to regulate energy homeostasis by controlling cellular metabolism. AMPK has been described as a cellular energy sensor that communicates with various metabolic pathways and networks to maintain energy balance. Earlier studies characterized AMPK as a tumor suppressor in the context of cancer. Later, a paradigm shift occurred in support of the oncogenic nature of AMPK, considering it a contextual oncogene. In support of this, various cellular and mouse models of tumorigenesis and clinicopathological studies demonstrated increased AMPK activity in various cancers. This review will describe AMPK's pro-tumorigenic activity in various malignancies and explain the rationale and context for using AMPK inhibitors in combination with anti-metabolite drugs to treat AMPK-driven cancers.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Neoplasias , Proteínas Quinasas Activadas por AMP/metabolismo , Adenosina Monofosfato/metabolismo , Adenosina Monofosfato/uso terapéutico , Animales , Metabolismo Energético/fisiología , Humanos , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Oncogenes/genética , Fosforilación
4.
FEBS J ; 289(6): 1575-1590, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34668648

RESUMEN

Hematopoietic PBX-interacting protein (HPIP, also known as PBXIP1) is an estrogen receptor (ER) interacting protein that regulates estrogen-mediated breast cancer cell proliferation and tumorigenesis. However, its functional significance in the context of mammary gland development is unexplored. Here, we report that HPIP is required for prolactin (PRL)-induced lactogenic differentiation in vitro. Molecular analysis of HPIP expression in mice revealed its induced expression at pregnancy and lactation stages of mammary gland. Moreover, PRL is a lactogenic hormone that controls pregnancy as well as lactation and induces Hpip/Pbxip1 expression in a signal transducer and activator of transcription 5a-dependent manner. Using mammary epithelial and lactogenic-competent cell lines, we further show that HPIP plays a regulatory role in PRL-mediated mammary epithelial cell differentiation, which is measured by acini formation, ß-casein synthesis, and lipid droplet formation. Further mechanistic studies using pharmacological inhibitors revealed that HPIP modulates PRL-induced ß-casein synthesis via phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) activation. This study also identified HPIP as a critical regulator of autocrine PRL signaling as treatment with the PRL receptor antagonist Δ1-9-G129R-hPRL restrained HPIP-mediated PRL synthesis, AKT activation, and ß-casein synthesis in cultured HC11 cells. Interestingly, we also uncovered that microRNA-148a (miR-148a) antagonizes HPIP-mediated mammary epithelial cell differentiation. Together, our study identified HPIP as a critical regulator of PRL signaling and revealed a novel molecular circuitry involving PRL, HPIP, PI3K/AKT, and miR-148a that controls mammary epithelial cell differentiation in vitro.


Asunto(s)
MicroARNs , Proteínas Proto-Oncogénicas c-akt , Animales , Caseínas/genética , Caseínas/metabolismo , Diferenciación Celular , Proteínas Co-Represoras , Células Epiteliales/metabolismo , Femenino , Glándulas Mamarias Animales , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Embarazo , Prolactina/genética , Prolactina/farmacología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo
5.
Cancer Lett ; 526: 12-28, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34767928

RESUMEN

While phenotypic plasticity is a critical factor contributing to tumor heterogeneity, molecular mechanisms underlying this process are largely unknown. Here we report that breast cancer cells display phenotypic diversity in response to hypoxia or normoxia microenvironments by operating a reciprocal positive feedback regulation of HPIP and HIF-1α. We show that under hypoxia, HIF-1α induces HPIP expression that establishes cell survival, and also promotes cell migration/invasion, EMT and metastatic phenotypes in breast cancer cells. Mechanistic studies revealed that HPIP interacts with SRP14, a component of signal recognition particle, and stimulates MMP9 synthesis under hypoxic stress. Whereas, in normoxia, HPIP stabilizes HIF-1α, causing the Warburg effect to support cell growth. Concurrently, mathematical modelling corroborates this reciprocal feedback loop in enabling cell-state transitions in cancer cells. Clinical data indicate that elevated levels of HPIP and HIF-1α correlate with unfavorable prognosis and shorter survival rates in breast cancer subjects. Together, this data shows a reciprocal positive feedback loop between HPIP and HIF-1α that was unknown hitherto. It unveils how the tumor microenvironment influences phenotypic plasticity that has an impact on tumor growth and metastasis and, further signifies considering this pathway as a potential therapeutic target in breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Humanos , Ratones , Fenotipo
6.
Cancer Lett ; 518: 243-255, 2021 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-34302919

RESUMEN

While cancer cells rewire metabolic pathways to sustain growth and survival under metabolic stress in solid tumors, the molecular mechanisms underlying these processes remain largely unknown. In this study, cancer cells switched from survival to death during the early to late phases of metabolic stress by employing a novel signaling switch from AMP activated protein kinase (AMPK)-Forkhead box O3 (FOXO3a)-hematopoietic PBX1-interacting protein (HPIP) to the ring finger protein 2 (RNF2)-HPIP-ubiquitin (Ub) pathway. Acute metabolic stress induced proto-oncogene HPIP expression in an AMPK-FOXO3a-dependent manner in breast cancer (BC) cells. HPIP depletion reduced cell survival and tumor formation in mouse xenografts, which was accompanied by diminished intracellular ATP levels and increased apoptosis in BC cells in response to metabolic (glucose) stress. Glutamine flux (13C-labeled) analysis further suggested that HPIP rewired glutamine metabolism by controlling the expression of the solute carrier family 1 member 5 (SLC1A5) and glutaminase (GLS) genes by acting as a coactivator of MYC to ensure cell survival upon glucose deprivation. However, in response to chronic glucose stress, HPIP was ubiquitinated by the E3-Ub ligase, RNF2, and was concomitantly degraded by the proteasome-mediated pathway, ensuring apoptosis. In support of these data, clinical analyses further indicated that elevated levels of HPIP correlated with AMPK activation in BC. Taken together, these data suggest that HPIP is a signal coordinator during metabolic stress and thus serves as a potential therapeutic target in BC.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Neoplasias de la Mama/metabolismo , Glucosa/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Complejo Represivo Polycomb 1/metabolismo , Adaptación Fisiológica/fisiología , Animales , Apoptosis/fisiología , Diferenciación Celular/fisiología , Línea Celular , Línea Celular Tumoral , Femenino , Glutaminasa/metabolismo , Glutamina/metabolismo , Células HEK293 , Humanos , Células MCF-7 , Ratones , Ratones Desnudos , Antígenos de Histocompatibilidad Menor , Estrés Fisiológico/fisiología , Ubiquitina-Proteína Ligasas/metabolismo
7.
Biochim Biophys Acta Rev Cancer ; 1876(1): 188576, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34090932

RESUMEN

Hematopoietic PBX interacting protein (HPIP or pre-B-cell leukemia transcription factor interacting protein (PBXIP1) was discovered two decades ago as a corepressor of pre-B-cell leukemia homeobox (PBX) 1 with a vital functional role in hematopoiesis. Later it emerged as a potential biomarker of poor prognosis and tumorigenesis for more than a dozen different cancers. It regulates aggressive cancer phenotypes, cell proliferation, metastasis, EMT, etc. The anomaly in the regulation of HPIP is linked with physiological disorders like renal fibrosis, chronic kidney disease and osteoarthritis. Scientists have unraveled more than twenty interacting proteins of HPIP and its functional role in various physiological and cellular processes that involves normal neuronal development, embryogenesis, endometrium decidualization, and germ cell proliferation. Over the past 20 years, we have witnessed the emerging role of HPIP and its association with a myriad of cellular activities ranging from germ cell proliferation to cancer aggressiveness, modulating multitude of signaling cascades like TGF-ß1, PI3K/AKT, Wnt, mTOR, and Sonic hedgehog signaling pathways. This review will give the current understanding of HPIP, in terms of its diverse functions, theoretical ideas, and further explore cellular links and promising areas that need to be investigated. We also provide a comprehensive overview of the transcript variants of HPIP and distinct sets of transcription factors regulating their expression, which may help to understand the role of HPIP in various cellular or physiological conditions.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Proteínas Co-Represoras/metabolismo , Células Germinativas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Sitios de Unión , Movimiento Celular , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Proteínas Co-Represoras/genética , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Invasividad Neoplásica , Neoplasias/genética , Neoplasias/patología , Regiones Promotoras Genéticas , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/genética , Transducción de Señal
8.
Cell Signal ; 85: 110046, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34004332

RESUMEN

Acting as a bridge between the cytoskeleton of the cell and the extra cellular matrix (ECM), the cell-ECM adhesions with integrins at their core, play a major role in cell signalling to direct mechanotransduction, cell migration, cell cycle progression, proliferation, differentiation, growth and repair. Biochemically, these adhesions are composed of diverse, yet an organised group of structural proteins, receptors, adaptors, various enzymes including protein kinases, phosphatases, GTPases, proteases, etc. as well as scaffolding molecules. The major integrin adhesion complexes (IACs) characterised are focal adhesions (FAs), invadosomes (podosomes and invadopodia), hemidesmosomes (HDs) and reticular adhesions (RAs). The varied composition and regulation of the IACs and their signalling, apart from being an integral part of normal cell survival, has been shown to be of paramount importance in various developmental and pathological processes. This review per-illustrates the recent advancements in the research of IACs, their crucial roles in normal as well as diseased states. We have also touched on few of the various methods that have been developed over the years to visualise IACs, measure the forces they exert and study their signalling and molecular composition. Having such pertinent roles in the context of various pathologies, these IACs need to be understood and studied to develop therapeutical targets. We have given an update to the studies done in recent years and described various techniques which have been applied to study these structures, thereby, providing context in furthering research with respect to IAC targeted therapeutics.


Asunto(s)
Adhesiones Focales , Mecanotransducción Celular , Adhesión Celular , Matriz Extracelular/metabolismo , Adhesiones Focales/metabolismo , Integrinas/metabolismo
9.
Biomed Mater ; 15(5): 055025, 2020 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-32464611

RESUMEN

Zirconia-containing wollastonite (CaSiO3) ceramics with partial substitution of zirconia (1, 3 and 5 mol%) were prepared using eggshells and rice husk ash as source materials for calcium oxide and silica, respectively, through a sol-gel technique. The effect of incorporation of zirconia on in vitro bioactivity, mechanical properties, degradability and cytocompatibility of wollastonite was studied. Bioactivity was evaluated by in vitro assay using simulated body fluid while degradability was tested in Tris-HCl buffer solution for different time periods (1, 3, 7, 14 and 21 d) according to the ISO 10 993-14 standard. Human osteosarcoma (MG-63) cells were used to assess cytocompatibility with the MTT assay. X-ray diffractometry, Fourier transform infrared spectroscopy and scanning electron microscopy-energy dispersive spectroscopy were used to characterize the ceramics before and after in vitro studies. The results obtained showed that increasing the zirconia content in the wollastonite phase increases microhardness, compressive strength, bending strength and the elasticity modulus, while slightly decreasing the rate of formation of the hydroxyapatite layer. Moreover, the samples doped with zirconia had a lower degradation rate and it was noticed that cell viability is unaffected by the incorporation of zirconia.


Asunto(s)
Materiales Biocompatibles/química , Compuestos de Calcio/química , Silicatos/química , Ingeniería de Tejidos/métodos , Circonio/química , Tampones (Química) , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cerámica , Durapatita/química , Humanos , Técnicas In Vitro , Ensayo de Materiales , Microscopía Electrónica de Rastreo , Osteosarcoma/tratamiento farmacológico , Espectroscopía Infrarroja por Transformada de Fourier , Sales de Tetrazolio/farmacología , Termogravimetría , Tiazoles/farmacología , Difracción de Rayos X
10.
RSC Adv ; 10(40): 24095-24107, 2020 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-35517325

RESUMEN

Among the breast cancers, triple negative breast cancer (TNBC) has relatively poor outcomes with a lower survival rate and personalised chemotherapy is the only option available for treatment. Currently in the biomedical domain, nanomaterials with porous morphology have revealed their tremendous possibilities to be used as a nanocarrier in treating cancer by offering void space to encapsulate/entrap biological agents. However, the development of nanocarrier-based targeted therapy with high therapeutic efficacy and fewer side effects to normal cells is always a challenge. Here, we have developed nanocargos based on biodegradable mesoporous PCL (polycaprolactone) of approx. diameter of 75 nm by template removal synthesis techniques. Succeeding the comparative analysis of the nanocarriers, the efficiencies of core shell PCL-mZnO (PZ) and mesoporous PCL (HPZ) to deliver paclitaxel (Taxol/T) into breast cancer cells, is investigated. We found that HPZ nanocapsules have less cytotoxicity and drug loading efficiency of about 600 µg mg-1. The Taxol-loaded nanoparticles (T-HPZ) have exhibited more cytotoxicity than Taxol alone treated cancer cells. Furthermore, T-HPZ treated MDA-MB231 cells are accumulated at G2/M phase of the cell cycle and eventually undergo apoptosis. In support of this, anchorage independent growth of MDA-MB231 cells are significantly inhibited by T-HPZ treatment. Together, our findings suggest that T-HPZ-based paclitaxel (Taxol/T) loaded nanoparticles provide a novel therapeutic option in the treatment of TNBC.

11.
J Biol Chem ; 294(26): 10236-10252, 2019 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-31101654

RESUMEN

Proper cell division relies on the coordinated regulation between a structural component, the mitotic spindle, and a regulatory component, anaphase-promoting complex/cyclosome (APC/C). Hematopoietic PBX-interacting protein (HPIP) is a microtubule-associated protein that plays a pivotal role in cell proliferation, cell migration, and tumor metastasis. Here, using HEK293T and HeLa cells, along with immunoprecipitation and immunoblotting, live-cell imaging, and protein-stability assays, we report that HPIP expression oscillates throughout the cell cycle and that its depletion delays cell division. We noted that by utilizing its D box and IR domain, HPIP plays a dual role both as a substrate and inhibitor, respectively, of the APC/C complex. We observed that HPIP enhances the G2/M transition of the cell cycle by transiently stabilizing cyclin B1 by preventing APC/C-Cdc20-mediated degradation, thereby ensuring timely mitotic entry. We also uncovered that HPIP associates with the mitotic spindle and that its depletion leads to the formation of multiple mitotic spindles and chromosomal abnormalities, results in defects in cytokinesis, and delays mitotic exit. Our findings uncover HPIP as both a substrate and an inhibitor of APC/C-Cdc20 that maintains the temporal stability of cyclin B1 during the G2/M transition and thereby controls mitosis and cell division.


Asunto(s)
Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Proteínas Cdc20/metabolismo , Ciclo Celular , Ciclina B1/química , Regulación de la Expresión Génica/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/farmacología , Mitosis , Ciclosoma-Complejo Promotor de la Anafase/antagonistas & inhibidores , Ciclosoma-Complejo Promotor de la Anafase/genética , Proteínas Cdc20/antagonistas & inhibidores , Proteínas Cdc20/genética , Células HEK293 , Células HeLa , Humanos , Huso Acromático , Especificidad por Sustrato
12.
Oncogene ; 38(4): 581-595, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30166591

RESUMEN

Autophagy is a homeostatic process that recycles damaged organelles and long-lived proteins by delivering them in double-membrane vesicles to lysosomes for degradation. Autophagy has a prominent role in survival, proliferation, and resistance of tumors in metabolic and chemotherapeutic stress conditions. Clinical trials with chloroquine-a known autophagy inhibitor-were unable to achieve complete autophagy inhibition in vivo, warranting the search for more potent autophagy inhibitors. In a process of exploring the mechanism of action of previously identified cytotoxic s-triazine analogs, we discovered that both IITZ-01 and IITZ-02 act as potent autophagy inhibitors. Treatment with these compounds resulted in the vacuolated appearance of cells due to their specific accumulation in lysosomes. In addition, these basic compounds also deacidify lysosomes as evidenced by the decrease in lysotracker red staining and inhibit maturation of lysosomal enzymes leading to lysosomal dysfunction. IITZ-01 and IITZ-02 enhance autophagosome accumulation but inhibit autophagosomal degradation by impairing lysosomal function, finally resulting in the inhibition of autophagy. Interestingly, compound IITZ-01 exhibited more than 10-fold potent autophagy inhibition along with 12- to 20-fold better cytotoxic action than CQ. IITZ-01 and IITZ-02 also abolished mitochondrial membrane potential and triggered apoptosis through the mitochondria-mediated pathway. Furthermore, IITZ-01 and IITZ-02 displayed potent antitumor action in vivo through autophagy inhibition and apoptosis induction in MDA-MB-231 breast cancer xenograft model with IITZ-01 exhibiting superior anticancer efficacy. Overall, these data demonstrate that IITZ-01 is potent autophagy inhibitor with single-agent anticancer activity and awaits further preclinical development as potential anticancer therapeutic.


Asunto(s)
Antineoplásicos/uso terapéutico , Autofagia/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Autofagosomas/efectos de los fármacos , Autofagosomas/ultraestructura , Línea Celular Tumoral , Femenino , Humanos , Concentración de Iones de Hidrógeno , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Ratones Endogámicos BALB C , Ratones Desnudos , Mitocondrias/efectos de los fármacos , Estructura Molecular , Distribución Aleatoria , Método Simple Ciego , Neoplasias de la Mama Triple Negativas/patología , Ensayo de Tumor de Célula Madre , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Cell Death Dis ; 9(11): 1087, 2018 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-30352996

RESUMEN

Of late, nimbolide, a limonoid from the neem tree (Azadirachta indica) has gained increasing research attention owing to its potent antiproliferative and apoptosis-inducing effects. The present study was designed to investigate the effect of nimbolide on autophagy and the time point at which the phosphorylation status of GSK-3ß and PI3K dictate the choice between autophagy and apoptosis in SCC131 and SCC4 oral cancer cells. Additionally, we analysed changes in the expression of proteins involved in autophagy and apoptosis after therapeutic intervention with nimbolide in a hamster model of oral oncogenesis. Furthermore, we also demonstrate changes in the expression of key genes involved in apoptosis and autophagy during the stepwise evolution of hamster and human OSCCs. Nimbolide-induced stereotypical changes in oral cancer cells characteristic of both apoptosis and autophagy. Time-course experiments revealed that nimbolide induces autophagy as an early event and then switches over to apoptosis. Nimbolide negatively regulates PI3K/Akt signalling with consequent increase in p-GSK-3ßTyr216, the active form of GSK-3ß that inhibits autophagy. Downregulation of HOTAIR, a competing endogenous RNA that sponges miR-126 may be a major contributor to the inactivation of PI3K/Akt/GSK3 signalling by nimbolide. Analysis of key markers of apoptosis and autophagy as well as p-AktSer473 during sequential progression of hamster and human OSCC revealed a gradual evolution to a pro-autophagic and antiapoptotic phenotype that could confer a survival advantage to tumors. In summary, the results of the present study provide insights into the molecular mechanisms by which nimbolide augments apoptosis by overcoming the shielding effects of cytoprotective autophagy through modulation of the phosphorylation status of Akt and GSK-3ß as well as the ncRNAs miR-126 and HOTAIR. Development of phytochemicals such as nimbolide that target the complex interaction between proteins and ncRNAs that regulate the autophagy/apoptosis flux is of paramount importance in cancer prevention and therapeutics.


Asunto(s)
Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Carcinoma de Células Escamosas/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Limoninas/farmacología , Neoplasias de la Boca/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Extractos Vegetales/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Antracenos/farmacología , Azadirachta/química , Carcinoma de Células Escamosas/inducido químicamente , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cricetinae , Modelos Animales de Enfermedad , Humanos , Limoninas/uso terapéutico , Masculino , Mesocricetus , MicroARNs/metabolismo , Neoplasias de la Boca/inducido químicamente , Neoplasias de la Boca/tratamiento farmacológico , Neoplasias de la Boca/patología , Fosforilación/efectos de los fármacos , Piperidinas/farmacología , Extractos Vegetales/uso terapéutico , ARN Largo no Codificante/antagonistas & inhibidores , ARN Largo no Codificante/metabolismo , Transducción de Señal/efectos de los fármacos
14.
PLoS Biol ; 16(4): e2005317, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29621237

RESUMEN

Activation of the amino acid starvation response (AAR) increases lifespan and acute stress resistance as well as regulates inflammation. However, the underlying mechanisms remain unclear. Here, we show that activation of AAR pharmacologically by Halofuginone (HF) significantly inhibits production of the proinflammatory cytokine interleukin 1ß (IL-1ß) and provides protection from intestinal inflammation in mice. HF inhibits IL-1ß through general control nonderepressible 2 kinase (GCN2)-dependent activation of the cytoprotective integrated stress response (ISR) pathway, resulting in rerouting of IL-1ß mRNA from translationally active polysomes to inactive ribocluster complexes-such as stress granules (SGs)-via recruitment of RNA-binding proteins (RBPs) T cell-restricted intracellular antigen-1(TIA-1)/TIA-1-related (TIAR), which are further cleared through induction of autophagy. GCN2 ablation resulted in reduced autophagy and SG formation, which is inversely correlated with IL-1ß production. Furthermore, HF diminishes inflammasome activation through suppression of reactive oxygen species (ROS) production. Our study unveils a novel mechanism by which IL-1ß is regulated by AAR and further suggests that administration of HF might offer an effective therapeutic intervention against inflammatory diseases.


Asunto(s)
Aminoácidos/deficiencia , Autofagia/inmunología , Colitis/inmunología , Interleucina-1beta/inmunología , Biosíntesis de Proteínas , Proteínas Serina-Treonina Quinasas/genética , Adaptación Fisiológica , Animales , Autofagia/efectos de los fármacos , Células Cultivadas , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Colitis/genética , Regulación de la Expresión Génica , Inflamasomas/genética , Inflamasomas/inmunología , Interleucina-1beta/genética , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Piperidinas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/inmunología , Inhibidores de la Síntesis de la Proteína/farmacología , Quinazolinonas/farmacología , ARN Mensajero/genética , ARN Mensajero/inmunología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/inmunología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/inmunología , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo , Dodecil Sulfato de Sodio/administración & dosificación , Inanición/genética , Inanición/inmunología , Estrés Fisiológico , Antígeno Intracelular 1 de las Células T/genética , Antígeno Intracelular 1 de las Células T/inmunología
15.
Biomed Mater ; 13(1): 015013, 2017 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-29216013

RESUMEN

Self-assembly is an important auto-organization process used in designing structural biomaterials which have the potential capability to heal tissues after traumatic injury. Although various materials having the ability to heal after injury are available, there is still a substantial need to develop new improved materials. To address this issue, we have developed hierarchical three-dimensional (3D) self-assembled zinc phosphate (Zn3(PO4)2) in the presence of cowpea mosaic virus (CPMV). Zn3(PO4)2 nanoparticles are self-assembled into nanosheets with a high degree of isotropy and then self-organized into a 3D structure that can enhance surface interactions with biological entities. The self-assembled structure is formed through the auto-organization of nanoparticles of size ∼50 nm under the influence of CPMV. The cellular response of self-assembled Zn3(PO4)2 and cell-particle adhesion behavior have been investigated through in vitro studies using modeled osteoblast-like MG63 cells. Self-assembled Zn3(PO4)2 resulted in proliferation of MG63 cells of up to 310% within 7 days of incubation. A 15% higher proliferation was obtained than with commercially available hydroxyapatite (HAp). Immunofluorescent analysis of MG63 cells after co-culturing with self-assembled Zn3(PO4)2 confirmed the healthy cytoskeletal organization and dense proliferation of MG63 cells. Further, Zn3(PO4)2 exhibited ∼28% cell-cycle progression in S phase, which is higher than obtained with commercially available HAp. Overall, these results demonstrate the multiple functions of hierarchical self-assembled Zn3(PO4)2 in the regeneration of bone tissue without defects and increasing the formation of cellular networks, and suggest its use in bone tissue engineering.


Asunto(s)
Comovirus , Durapatita/química , Nanopartículas del Metal/química , Fosfatos/química , Regeneración , Ingeniería de Tejidos/métodos , Compuestos de Zinc/química , Materiales Biocompatibles/química , Huesos , Adhesión Celular , Técnicas de Cultivo de Célula , Ciclo Celular , Proliferación Celular , Supervivencia Celular , Técnicas de Cocultivo , Humanos , Nanoestructuras , Osteoblastos/metabolismo , Propiedades de Superficie
16.
Biomed Pharmacother ; 95: 956-964, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28915537

RESUMEN

Remarkably, majority of the cancer deaths are due to metastasis, not because of primary tumors. Metastasis is one of the important hallmarks of cancer. During metastasis invasion of primary tumor cells from the site of origin to a new organ occurs. Metastasis associated proteins (MTAs) are a small family of transcriptional coregulators that are closely associated with tumor metastasis. These proteins are integral components of nuclear remodeling and deacetylation complex (NuRD). By virtue of being integral components of NuRD, these proteins regulate the gene expression by altering the epigenetic changes such as acetylation and methylation on the target gene chromatin. Among the MTA proteins, MTA1 expression is very closely correlated with the aggressiveness of several cancers that includes breast, liver, colon, pancreas, prostate, blood, esophageal, gastro-intestinal etc. Considering its close association with aggressiveness in human cancers, MTA1 may be considered as a potential therapeutic target for cancer treatment. The recent developments in its crystal structure further strengthened the idea of developing small molecule inhibitors for MTA1. In this review, we discuss the recent trends on the diverse functions of MTA1 and its role in various cancers, with the focus to consider MTA1 as a 'druggable' target in the control of human cancers.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Histona Desacetilasas/genética , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Proteínas Represoras/genética , Animales , Histona Desacetilasas/química , Histona Desacetilasas/metabolismo , Humanos , Modelos Biológicos , Terapia Molecular Dirigida , Proteínas Represoras/química , Proteínas Represoras/metabolismo , Transactivadores
17.
Mater Sci Eng C Mater Biol Appl ; 77: 772-779, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28532092

RESUMEN

The present study aims to elucidate the applications of Titania (TiO2) doped calcium borosilicate glass as a biocompatible material in regenerative orthopedic applications. In this context, we have examined the bioactivity of various concentrations of TiO2 doped glasses with the help of simulated body fluid (SBF). Cytocompatibility, cell proliferation, and protein expression studies revealed the potential candidature of TiO2 doped glasses on osteoblast cell lines (MG-63). We hypothesized that TiO2 doped calcium borosilicate glasses are most cytocompatible material for bone implants. Glasses with composition 31B2O3-20SiO2-24.5Na2O-(24.5-x) CaO- x TiO2 (x=0,0.5,1,2) have been prepared by the conventional melt-quenching technique. After immersion of glasses in the SBF, formation of hydroxyapatite layer on the surface was confirmed by X-ray Diffractometer (XRD), Fourier Transform Infrared Spectroscopy (FT-IR) and Scanning Electron Microscopy-Energy Dispersive Spectroscopy (SEM-EDS) analysis. Significant change in the pH of the body fluid was observed with the addition of titania. Degradation test was performed as per the ISO 10993. The results showed that partial substitution of TiO2 with CaO negatively influenced bioactivity; it decreased with increase in concentration of TiO2. Vickers hardness tester was used to measure the microhardness values of the prepared glasses. With the increasing of TiO2 content, the microhardness of the glass samples was increased from 545Hv to 576Hv. Cytocompatibility has been evaluated with MG-63 cells by using MTT assay. Further, we observed that there was no change in expressions of cyclin levels even after the incorporation of titania. The antibacterial properties were examined against E. coli and S. aureus. Strong antibacterial efficacy was observed for 2% TiO2 in the system. Hence it can be concluded that titania-doped borosilicate glasses may be used as potential materials in bone tissue engineering.


Asunto(s)
Vidrio , Materiales Biocompatibles , Calcio , Escherichia coli , Humanos , Ensayo de Materiales , Espectroscopía Infrarroja por Transformada de Fourier , Staphylococcus aureus , Titanio , Difracción de Rayos X
18.
Cell Oncol (Dordr) ; 40(2): 133-144, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28039608

RESUMEN

PURPOSE: Hematopoietic PBX interacting protein (HPIP), a scaffold protein, is known to regulate the proliferation, migration and invasion in different cancer cell types. The aim of this study was to assess the role of HPIP in ovarian cancer cell migration, invasion and epithelial-mesenchymal transition (EMT), and to unravel the mechanism by which it regulates these processes. METHODS: HPIP expression was assessed by immunohistochemistry of tissue microarrays containing primary ovarian tumor samples of different grades. OAW42, an ovarian carcinoma-derived cell line exhibiting a high HPIP expression, was used to study the role of HPIP in cell migration, invasion and EMT. HPIP knockdown in these cells was achieved using a small hairpin RNA (shRNA) approach. Cell migration and invasion were assessed using scratch wound and transwell invasion assays, respectively. The extent of EMT was assessed by determining the expression levels of Snail, Vimentin and E-cadherin using Western blotting. The effect of HPIP expression on AKT and MAPK activation was also investigated by Western blotting. Cell viabilities in response to cisplatin treatment were assessed using a MTT assay, whereas apoptosis was assessed by determining caspase-3 and PARP cleavage in ovarian carcinoma-derived SKOV3 cells. RESULTS: We found that HPIP is highly expressed in high-grade primary ovarian tumors. In addition, we found that HPIP promotes the migration, invasion and EMT in OAW42 cells and induces EMT in these cells via activation of the PI3K/AKT pathway. The latter was found to lead to stabilization of the Snail protein and to repression of E-cadherin expression through inactivation of GSK-3ß. We also found that HPIP expression confers cisplatin resistance to SKOV3 cells after prolonged exposure and that its subsequent knockdown decreases the viability of these cells and increases caspase-3 activation and PARP proteolysis in these cells following cisplatin treatment. CONCLUSIONS: From these results we conclude that HPIP expression is associated with high-grade ovarian tumors and may promote their migration, invasion and EMT, a process that is associated with metastasis. In addition, we conclude that HPIP may serve as a potential therapeutic target for cisplatin resistant ovarian tumors.


Asunto(s)
Cisplatino/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias Ováricas/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Técnicas de Silenciamiento del Gen , Silenciador del Gen/efectos de los fármacos , Humanos , Clasificación del Tumor , Invasividad Neoplásica , Neoplasias Ováricas/enzimología , Estabilidad Proteica/efectos de los fármacos , Factores de Transcripción de la Familia Snail/metabolismo
19.
Biosci Rep ; 36(6)2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27884978

RESUMEN

Endocrine resistance, which occurs either by de novo or acquired route, is posing a major challenge in treating hormone-dependent breast cancers by endocrine therapies. The loss of oestrogen receptor α (ERα) expression is the vital cause of establishing endocrine resistance in this subtype. Understanding the mechanisms that determine the causes of this phenomenon are therefore essential to reduce the disease efficacy. But how we negate oestrogen receptor (ER) negativity and endocrine resistance in breast cancer is questionable. To answer that, two important approaches are considered: (1) understanding the cellular origin of heterogeneity and ER negativity in breast cancers and (2) characterization of molecular regulators of endocrine resistance. Breast tumours are heterogeneous in nature, having distinct molecular, cellular, histological and clinical behaviour. Recent advancements in perception of the heterogeneity of breast cancer revealed that the origin of a particular mammary tumour phenotype depends on the interactions between the cell of origin and driver genetic hits. On the other hand, histone deacetylases (HDACs), DNA methyltransferases (DNMTs), miRNAs and ubiquitin ligases emerged as vital molecular regulators of ER negativity in breast cancers. Restoring response to endocrine therapy through re-expression of ERα by modulating the expression of these molecular regulators is therefore considered as a relevant concept that can be implemented in treating ER-negative breast cancers. In this review, we will thoroughly discuss the underlying mechanisms for the loss of ERα expression and provide the future prospects for implementing the strategies to negate ER negativity in breast cancers.


Asunto(s)
Antineoplásicos Hormonales/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Resistencia a Antineoplásicos , Antagonistas de Estrógenos/uso terapéutico , Receptor alfa de Estrógeno/genética , Animales , Neoplasias de la Mama/clasificación , Metilación de ADN , Receptor alfa de Estrógeno/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Heterogeneidad Genética , Histona Desacetilasas/metabolismo , Humanos , Ratones , MicroARNs/metabolismo , Neoplasias Hormono-Dependientes/tratamiento farmacológico , Neoplasias Hormono-Dependientes/genética , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Ubiquitinación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA