Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Sci Rep ; 13(1): 7584, 2023 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-37165028

RESUMEN

The 90 kDa heat shock protein, Hsp90, functions as a cancer chaperone contributing to tumor proliferation. We have encountered the mitochondrial homolog of Hsp90, the TRAP-1, regulating mitochondrial dynamics, metabolism, and tumor metastasis. Although Hsp90 is associated with a broad network of proteins regulating various cellular processes, TRAP-1-mediated cellular networks are unclear. Therefore, using TRAP-1 knockdown (KD) and overexpression (OE) systems, we compared their quantitative transcriptome (RNA Sequencing) and proteomic (LC-MS/MS) patterns to obtain molecular signatures that are altered in response to TRAP-1 KD or OE. We report TRAP-1 modulating vital metabolic pathways such as the tricarboxylic acid cycle, oxidative phosphorylation, electron transport chain, glycolysis, and gluconeogenesis. In addition, TRAP-1 facilitated the pentose phosphate pathway to shunt carbons back to glycolysis or gluconeogenesis, a much-solicited tumor response. Subsequently, we examined the TRAP-1 interactome using the tandem affinity purification system and identified 255 unique proteins. These diverse proteins appear to regulate several cellular processes, including energy metabolism, suggesting that TRAP-1, in addition to metabolic rewiring, maintains mitochondrial integrity. Our study exposes the unknown functions of TRAP-1 in cancer cells. Systematic evaluation of TRAP-1 interactors may uncover novel regulatory mechanisms in disease aggression. Since metabolic inhibitors are emerging as potential anticancer agents, our study gains importance.


Asunto(s)
Neoplasias , Proteómica , Humanos , Cromatografía Liquida , Espectrometría de Masas en Tándem , Chaperonas Moleculares/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Neoplasias/genética
2.
Mitochondrion ; 69: 159-170, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36828164

RESUMEN

Understanding cancer cell metabolism always provides information on hidden dimensions of tumor adaptations. Warburg's theory that cancer cells opt for aerobic glycolysis over the mitochondrial oxidative phosphorylation (OXPHOS) system is widely accepted. However, the hypothesis does not explain the mitochondrion's role in these cells. Here, we demonstrate that intact mitochondria are used for anaplerotic functions and ATP production by utilizing glutamine with the help of mitochondrial chaperone TRAP-1 (Tumor Necrosis Factor Receptor-associated Protein 1). TRAP-1 otherwise promotes aerobic glycolysis by lowering the mitochondrial OXPHOS in the presence of glucose. Here, we show that TRAP-1 maintains mitochondrial integrity and augments glutamine metabolism upon glucose deprivation to meet the cellular energy demand. The enhanced PER and ECAR correlating with increased ATP production suggest that glutamine fuels mitochondria in the presence of TRAP-1. We also found that TRAP1-dependent glutamine utilization involves the HIF2α-SLC1A5-GLS axis and is independent of hypoxia. Subsequently, we show that the metastatic potential of tumor cells is linked with glucose utilization, whereas the proliferative potential is linked with both glucose and glutamine utilization. Our findings establish that TRAP-1 contributes to enhanced glutamine utilization through the HIF2α-SLC1A5-GLS axis. Our results endow that TRAP-1 inhibitors can be potential drug candidates to combat tumor metabolism. Therefore, their use, either alone or in combination with existing chemotherapeutic agents, may target tumor metabolism and improve anticancer treatment response.


Asunto(s)
Glucólisis , Neoplasias , Humanos , Glutamina/metabolismo , Metabolismo Energético , Chaperonas Moleculares/metabolismo , Neoplasias/metabolismo , Mitocondrias/metabolismo , Adenosina Trifosfato/metabolismo , Glucosa/metabolismo , Antígenos de Histocompatibilidad Menor/metabolismo , Sistema de Transporte de Aminoácidos ASC/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo
3.
Biochem Biophys Res Commun ; 600: 44-50, 2022 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-35182974

RESUMEN

The heat shock transcription factor HSF1 regulates the inducible Hsp gene transcription, whereas HSF2 is involved in the constitutive transcription. HSFs can work for the non-heat shock genes transcription in a case-specific manner to facilitate normal cellular functions. Here, we demonstrate that HSF2 acts as an upstream regulator of heat shock-induced autophagy response in a rat histiocytoma. The heat-induced HSF2 transactivates the B-cell translocation gene-2 (BTG2) transcription, and the latter acts as a transcriptional coactivator for superoxide dismutase (SOD2). The altered HSF2 promoter occupancy on the BTG2 promoter enhances BTG2 transcription. Since SOD2 regulation is linked to mitochondrial redox sensing, HSF2 appears to act as a redox sensor in deciding the cell fate. The HSF2 shRNA or NFE2L2/BTG2 siRNA treatments have interfered with the autophagy response. We demonstrate that HSF2 is an upstream activator of autophagy response, and the HSF2-BTG2-SOD2 axis acts as a switch between the non-selective (micro/macro) and selective (chaperone-mediated) autophagy.


Asunto(s)
Proteínas de Unión al ADN , Proteínas Inmediatas-Precoces , Animales , Autofagia , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción del Choque Térmico/genética , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , ARN Interferente Pequeño , Ratas , Superóxido Dismutasa/genética , Factores de Transcripción/genética , Proteínas Supresoras de Tumor
4.
Mitochondrion ; 60: 101-111, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34365052

RESUMEN

Mitochondrial adaptations to various environmental cues contribute to cellular and organismal adaptations across multiple model organisms. Due to increased complexity, a direct connection between mitochondrial integrity and oxygen fluctuations, and survival fitness was not demonstrated. Here, using C. elegans as a model system, we studied the role of HIF-1, Hsp90, and TRAP-1 in mitochondrial adaptations during chemical hypoxia. We show that Hsp90mt (Hsp90 mutant) but not HIF-1mt (HIF-1 mutant) affects hypoxia adaptation in nematodes. TRAP-1KD (TRAP-1 knockdown) interfered with the survival and fecundity of worms. Compared to Hsp90mt, TRAP-1KD has induced a significant decrease in mitochondrial integrity and oxygen consumption rate. The complex I inhibitor rotenone did not affect ATP levels in Hsp90mt worms. However, ATP levels were decreased in TRAP-1KD worms under similar conditions. The glucose restriction has reduced, and glucose supplementation has increased the survival rate in Hsp90mt worms. Neither glucose restriction nor glucose supplementation has significantly affected the survival of TRAP-1KD worms in response to hypoxia. However, TRAP-1 inhibition using a nanocarrier drug has dramatically reduced the survival rate in response to hypoxia. Our results suggest that Hsp90 and TRAP-1 independently regulate hypoxia adaptations and metabolic plasticity in C. elegans. Considering the emerging roles of TRAP-1 in altered energy metabolism and cellular adaptations, our findings gain importance.


Asunto(s)
Adaptación Fisiológica , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Oxígeno/metabolismo , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Fertilidad , Proteínas HSP90 de Choque Térmico/genética , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Oxígeno/farmacología , Consumo de Oxígeno , Interferencia de ARN , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
6.
Inflammopharmacology ; 29(2): 343-366, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33723711

RESUMEN

Inflammation is an intrinsic defence mechanism triggered by the immune system against infection or injury. Chronic inflammation allows the host to recover or adapt through cellular and humoral responses, whereas acute inflammation leads to cytokine storms resulting in tissue damage. In this review, we present the overlapping outcomes of cancer inflammation with virus-induced inflammation. The study emphasises how anti-inflammatory drugs that work against cancer inflammation may work against the inflammation caused by the viral infection. It is established that the cytokine storm induced in response to SARS-CoV-2 infection contributes to disease-associated mortality. While cancer remains the second among the diseases associated with mortality worldwide, cancer patients' mortality rates are often observed upon extended periods after illness, usually ranging from months to years. However, the mortality rates associated with COVID-19 disease are robust. The cytokine storm induced by SARS-CoV-2 infection appeared to be responsible for the multi-organ failure and increased mortality rates. Since both cancer and COVID-19 disease share overlapping inflammatory mechanisms, repurposing some anticancer and anti-inflammatory drugs for COVID-19 may lower mortality rates. Here, we review some of these inflammatory mechanisms and propose some potential chemotherapeutic agents to intervene in them. We also discuss the repercussions of anti-inflammatory drugs such as glucocorticoids and hydroxychloroquine with zinc or antiviral drugs such as ivermectin and remdesivir against SARS-CoV-2 induced cytokine storm. In this review, we emphasise on various possibilities to reduce SARS-CoV-2 induced cytokine storm.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , COVID-19/patología , Inflamación/tratamiento farmacológico , Neoplasias/tratamiento farmacológico , Neoplasias/patología , SARS-CoV-2/efectos de los fármacos , Antiinflamatorios/uso terapéutico , Antivirales/uso terapéutico , COVID-19/virología , Humanos , Inflamación/patología , Inflamación/virología
7.
Eur J Cell Biol ; 100(1): 151148, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33388604

RESUMEN

The 90 kDa heat shock protein, Hsp90, is involved in the conformational stabilization and functional maturation of diverse cancer-promoting proteins. To date, more than 300 Hsp90 clients have identified, suggesting that Hsp90 plays a central role in deciding cancer cell fate. In this study, we present the nuclear functions of Hsp90 in regulating the E2F-dependent gene transcription. We show that the conformation specific Hsp90 inhibitor, 17AAG decreases the total cellular E2F levels more selectively in cancer cells than transformed cells. With the help of coimmunoprecipitation experiments, we show that Hsp90 interacts with E2F1 and E2F2 in cancer cells, whereas in transformed cells, only E2F1 interacts with Hsp90. Retention of E2F2 in the nucleus of cancer cells upon MG132 combination with 17AAG has suggested that Hsp90 is required for E2F2 stability and function. The HDAC6 inhibitor tubacin treatment did not interfere with E2F1/2 stability and nuclear accumulation. However, the HDAC3 inhibitor, RGFP966 treatment, decreased nuclear E2F1/2 and its target gene expression. The nuclear accumulation of E2F1 and E2F2 upon cell cycle inhibition correlated with decreased acetylated Hsp90. We expose the nuclear functions of Hsp90 in facilitating the cell cycle progression through stabilizing E2F1/2.


Asunto(s)
Neoplasias de la Mama/genética , Factor de Transcripción E2F1/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética/genética , Femenino , Humanos
8.
Biochim Biophys Acta Mol Cell Res ; 1868(3): 118943, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33359710

RESUMEN

Cancer emergence is associated with cellular adaptations to altered signal transduction mechanisms arbitrated by mutated kinases. Since conventional kinase inhibitors can exhibit certain limitations to such kinase adaptations, overcoming kinase adaptation for cancer treatment gains importance. The cancer chaperone, Hsp90, is implicated in the conformational maturation and functional stabilization of mutated gene products. However, its role in kinase adaptations is not explored in detail. Therefore, the present study aims to understand the mechanisms of Hsp90-dependent kinase adaptation and develop a novel antitumor strategy. We chose malignant human lung cancer cells to demonstrate Hsp90-dependent RAF oncogene adaptation. We show that RAF oncogene adaptations were predominant over wild type RAF and are facilitated by conformation-specific Hsp90. Consequently, the conformation-specific Hsp90 inhibitor, 17AAG, interfered with oncogenic RAF stability and function and inhibited cell proliferation. The enforced cytostasis further triggered premature cellular senescence and acted as an efficient and irreversible tumor suppressor mechanism. Our results also display that oncogenic RAF interactions with Hsp90 require the middle-charged region of the chaperone. Our mice xenografts revealed that 17AAG pretreated tumor cells lost their ability to proliferate and metastasize in vivo. In summary, we demonstrated Hsp90-dependent kinase adaptation in tumor cells and the effect of Hsp90 inhibition in triggering premature senescence to interfere with the tumor progression. Our findings are of both biological relevance and clinical importance.


Asunto(s)
Antineoplásicos/administración & dosificación , Benzoquinonas/administración & dosificación , Proteínas HSP90 de Choque Térmico/química , Proteínas HSP90 de Choque Térmico/metabolismo , Lactamas Macrocíclicas/administración & dosificación , Neoplasias Pulmonares/tratamiento farmacológico , Quinasas raf/genética , Células A549 , Animales , Antineoplásicos/farmacología , Benzoquinonas/farmacología , Proliferación Celular/efectos de los fármacos , Senescencia Celular , Reparación del ADN/efectos de los fármacos , Células HEK293 , Humanos , Lactamas Macrocíclicas/farmacología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Masculino , Ratones , Mutación , Conformación Proteica , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Cell Signal ; 76: 109801, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33017618

RESUMEN

Deregulated DNA methylation and post-translational histone modifications are majorly associated with cancer progression. Histone modification regulates the gene expression patterns that contribute to the emergence of sporadic cancers. Histone deacetylases (HDACs) act as erasers of acetylation marks, and their functions are often deregulated in cancer. Since non-histone proteins can also act as substrates for HDACs, identifying their involvement in vital regulatory molecules contributing to cancer progression is essential. Hsp90 is a cancer chaperone that contributes to kinase evolution and, thus, cellular adaptations. Acetylated Hsp90 loses its chaperoning functions and client protein interactions. Robust cell proliferation is one of the hallmarks of cancer. However, Hsp90 involvement in cancer promoting gene transcription is less understood. Using human breast cancer cells, we demonstrate that nuclear Hsp90 functions are regulated by HDAC3, while Hsp90 regulates HDAC3 nuclear translocation. Pharmacological inhibition of Hsp90 decreased the HDAC3 nuclear translocation and increased the gene expression relevant to epithelial to mesenchymal transition. Further, inhibition of HDAC3 resulted in the nuclear accumulation of acetylated Hsp90. Additionally, Hsp90 inhibition affected the global histone acetylation and methylation patterns, whereas HDAC3 inhibition exhibited less impact. Our results display a novel regulatory mechanism mediated by Hsp90 and HDAC3 in tumor cells. Considering that Hsp90 and histone deacetylase inhibitors are emerging as novel anticancer agents, our findings may have clinical relevance.


Asunto(s)
Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Neoplasias , Acetilación/efectos de los fármacos , Humanos , Células MCF-7 , Metilación/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo
10.
Mitochondrion ; 54: 92-101, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32784002

RESUMEN

Mitochondria play a central role in regulating cellular energy metabolism. However, the present understanding of mitochondria has changed from its unipotent functions to pluripotent and insists on understanding the role of mitochondria not only in regulating the life and death of cells, but in pathological conditions such as cancer. Unlike other cellular organelles, subtle alterations in mitochondrial organization may significantly influence the balance between metabolic networks and cellular behavior. Therefore, the delicate balance between the fusion and fission dynamics of mitochondrion can indicate cell fate. Here, we present mitochondrial chaperone TRAP1 influence on mitochondrial architecture and its correlation with tumor growth and metastasis. We show that TRAP1 overexpression (TRAP1 OE) promotes mitochondrial fission, whereas, TRAP1 knockdown (TRAP1 KD) promotes mitochondrial fusion. Interestingly, TRAP1 OE or KD had a negligible effect on mitochondrial integrity. However, TRAP1 OE cells exhibited enhanced proliferative potential, while TRAP1 KD cells showing increased doubling time. Further, TRAP1 dependent mitochondrial dynamic alterations appeared to be unique since mitochondrial localization of TRAP1 is a mandate for dynamic changes. The expression patterns of fusion and fission genes have failed to correlate with TRAP1 expression, indicating a possibility that the dynamic changes can be independent of these genes. In agreement with enhanced proliferative potential, TRAP1 OE cells also exhibited enhanced migration in vitro and tumor metastasis in vivo. Further, TRAP1 OE cells showed altered homing properties, which may challenge site-specific anticancer treatments. Our findings unravel the TRAP1 role in tumor metastasis, which is in addition to altered energy metabolism.


Asunto(s)
Proteínas HSP90 de Choque Térmico/genética , Proteínas HSP90 de Choque Térmico/metabolismo , Mitocondrias/metabolismo , Neuroblastoma/patología , Animales , Línea Celular Tumoral , Metabolismo Energético , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones , Dinámicas Mitocondriales , Metástasis de la Neoplasia , Trasplante de Neoplasias , Neuroblastoma/genética , Neuroblastoma/metabolismo
11.
Biochim Biophys Acta Mol Cell Res ; 1867(8): 118728, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32343987

RESUMEN

Acquired multidrug resistance of cancer cells challenges the chemotherapeutic interventions. To understand the role of molecular chaperone, Hsp90 in drug adapted tumor cells, we have used in vitro drug adapted epidermoid tumor cells as a model system. We found that chemotherapeutic drug adaptation of tumor cells is mediated by induced activities of both Hsp90 and P-glycoprotein (P-gp). Although the high-affinity conformation of Hsp90 has correlated with the enhanced drug efflux activity, we did not observe a direct interaction between P-gp and Hsp90. The enrichment of P-gp and Hsp90 at the cholesterol-rich membrane microdomains is found obligatory for enhanced drug efflux activity. Since inhibition of cholesterol biosynthesis is not interfering with the drug efflux activity, it is presumed that the net cholesterol redistribution mediated by Hsp90 regulates the enhanced drug efflux activity. Our in vitro cholesterol and Hsp90 interaction studies have furthered our presumption that Hsp90 facilitates cholesterol redistribution. The drug adapted cells though exhibited anti-proliferative and anti-tumor effects in response to 17AAG treatment, drug treatment has also enhanced the drug efflux activity. Our findings suggest that drug efflux activity and metastatic potential of tumor cells are independently regulated by Hsp90 by distinct mechanisms. We expose the limitations imposed by Hsp90 inhibitors against multidrug resistant tumor cells.


Asunto(s)
Antineoplásicos/farmacología , Colesterol/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Proteínas HSP90 de Choque Térmico/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP , Inductores de la Angiogénesis , Animales , Apoptosis/efectos de los fármacos , Benzoquinonas/farmacología , Línea Celular Tumoral , Progresión de la Enfermedad , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Proteínas HSP90 de Choque Térmico/efectos de los fármacos , Proteínas HSP90 de Choque Térmico/genética , Humanos , Lactamas Macrocíclicas/farmacología , Masculino , Ratones Desnudos , Transcriptoma , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Toxicol In Vitro ; 65: 104828, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32184171

RESUMEN

The altered molecular pathways in response to chemotherapeutic interventions impose limitations on breast cancer treatments. Therefore, understanding the outcome of these alternative pathways may help in improving the chemotherapy. In this study, using hormone responsive and hormone independent breast cancer cells, MCF-7 and MDAMB-231 respectively, we studied some of the molecular pathways that contribute to cancer progression. Since the cancer chaperone, Hsp90 inhibitors have entered the clinical trials, we used Hsp90 inhibitor, 17AAG to examine the outcome of altered molecular pathways. The observed differential sensitivity in MCF7 and MDAMB-231 cells to 17AAG treatment is then attributed to both tumor microenvironment mediated by hypoxia and acquired alterations in the endogenous stem cell pool. Interestingly, tumor cells are able to retain epithelial characteristics in addition to gaining mesenchymal characteristics in response to 17AAG treatment. We observed MCF-7 cells exhibiting induced cellular differentiation, whereas MDAMB-231 cells exhibiting reduced cellular differentiation in response to 17AAG treatment. These changes are subsequently found to be the sporadic outcome of altered epigenetic landscape. The mice tumor xenograft studies have revealed that decreased metastatic potential of MCF-7 and increased metastatic potential with altered homing properties of MDAMB-231 are the outcome of altered molecular pathways. Our findings expose the interference of altered molecular pathways influencing the therapeutic outcome.


Asunto(s)
Antineoplásicos/farmacología , Benzoquinonas/farmacología , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Lactamas Macrocíclicas/farmacología , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Animales , Antineoplásicos/uso terapéutico , Benzoquinonas/uso terapéutico , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Epigénesis Genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Lactamas Macrocíclicas/uso terapéutico , Neoplasias Mamarias Experimentales/genética , Ratones Desnudos , Resultado del Tratamiento , Microambiente Tumoral/efectos de los fármacos
13.
ACS Appl Bio Mater ; 3(7): 4188-4197, 2020 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-35025420

RESUMEN

Among human diseases, cancer has been in the frontlines of drug discovery and development. Despite having several decades of research efforts, therapeutic targeting of cancer is still challenging, which is due to the ability of cancer cells to adapt to the tumor microenvironment, exhibiting resistance to therapeutic drugs, and facilitated altered cancer metabolism. The small molecule inhibitors aimed at targeting a selective pathway are becoming void since cancer cells can activate alternate mechanisms. Despite broad acceptance of the Warburg effect, cellular energy metabolism, which determines the cell fate, is often overlooked for cancer treatment. We reported earlier that mitochondrial chaperone, TRAP-1 acts as a switch for activating the alternate cellular metabolism. Hence, we hypothesized that interfering with TRAP-1 inhibition can target the activation of alternative energy metabolism and sensitize tumor cells to existing chemotherapeutic drugs. We developed a nanocarrier where the iron oxide nanoparticles (IONs) were conjugated to Hsp90 inhibitor, geldanamycin (GA), and the mitochondria localization signal (MLS) peptide. We examined its effect against mitochondrial dynamics and metabolic status of human tumor cells. The synthesized nanocarrier exhibited both stability and target-specific activity and did not show nanoparticle-associated cytotoxicity. However, the nanocarrier treated cancer cells exhibited altered mitochondrial morphology and decreased cellular ATP levels suggesting that selective TRAP-1 targeting interferes with the altered energy metabolism. We present a nanoparticle-based TRAP-1 inhibitor to target tumor metabolism.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA