Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Indian J Plast Surg ; 52(1): 73-80, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31456615

RESUMEN

Negative-pressure wound therapy (NPWT) has gained increasing popularity among clinicians since its introduction in 1997 as a potential aid to wound healing. Multiple benefits of NPWT have since been proven in studies, including increase in granulation tissue formation, decrease in bacterial load, and the improved survival of flaps. With our increasing use and greater understanding of the tissue and cellular changes that occur in a wound treated with NPWT, our lower-limb reconstructive practice has also evolved. Although controversial, the definite timing for lower-limb reconstruction has stretched from 72 hours to longer than 2 weeks as NPWT contains the wound within a sterile, closed system. It has also shown to decrease the rate of infection in open tibia fractures. Previously, a large number of critical defects of the lower limb would require free tissue transfer for definitive reconstruction. NPWT has reduced this rate by more than 50% and has allowed for less complicated resurfacing procedures to be performed instead.

2.
Autophagy ; 20(6): 1418-1441, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38261660

RESUMEN

RAS is one of the most commonly mutated oncogenes associated with multiple cancer hallmarks. Notably, RAS activation induces intracellular reactive oxygen species (ROS) generation, which we previously demonstrated as a trigger for autophagy-associated execution of mutant KRAS-expressing cancer cells. Here we report that drug (merodantoin; C1)-induced activation of mutant KRAS promotes phospho-AKT S473-dependent ROS-mediated S616 phosphorylation and mitochondrial localization of DNM1L/DRP1 (dynamin 1 like) and cleavage of the fusion-associated protein OPA1 (OPA1 mitochondrial dynamin like GTPase). Interestingly, accumulation of the outer mitochondrial membrane protein VDAC1 (voltage dependent anion channel 1) is observed in mutant KRAS-expressing cells upon exposure to C1. Conversely, silencing VDAC1 abolishes C1-induced mitophagy, and gene knockdown of either KRAS, AKT or DNM1L rescues ROS-dependent VDAC1 accumulation and stability, thus suggesting an axis of mutant active KRAS-phospho-AKT S473-ROS-DNM1L-VDAC1 in mitochondrial morphology change and cancer cell execution. Importantly, we identified MTOR (mechanistic target of rapamycin kinsase) complex 2 (MTORC2) as the upstream mediator of AKT phosphorylation at S473 in our model. Pharmacological or genetic inhibition of MTORC2 abrogated C1-induced phosphorylation of AKT S473, ROS generation and mitophagy induction, as well as rescued tumor colony forming ability and migratory capacity. Finally, increase in thermal stability of KRAS, AKT and DNM1L were observed upon exposure to C1 only in mutant KRAS-expressing cells. Taken together, our work has unraveled a novel mechanism of selective targeting of mutant KRAS-expressing cancers via MTORC2-mediated AKT activation and ROS-dependent mitofission, which could have potential therapeutic implications given the relative lack of direct RAS-targeting strategies in cancer.Abbreviations: ACTB/ß-actin: actin beta; AKT: AKT serine/threonine kinase; C1/merodantoin: 1,3-dibutyl-2-thiooxo-imidazoldine-4,5-dione; CAT: catalase; CETSA: cellular thermal shift assay; CHX: cycloheximide; DKO: double knockout; DNM1L/DRP1: dynamin 1 like; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; H2O2: hydrogen peroxide; HSPA1A/HSP70-1: heat shock protein family A (Hsp70) member 1A; HSP90AA1/HSP90: heat shock protein 90 alpha family class A member 1; KRAS: KRAS proto-oncogene, GTPase; MAP1LC3B/LC3B, microtubule associated protein 1 light chain 3 beta; LC3B-I: unlipidated form of LC3B; LC3B-II: phosphatidylethanolamine-conjugated form of LC3B; MAPKAP1/SIN1: MAPK associated protein 1; MAPK1/ERK2: mitogen-activated protein kinase 1; MAPK3/ERK1: mitogen-activated protein kinase 3; MFI: mean fluorescence intensity; MiNA: Mitochondrial Network Analysis; MTOR: mechanistic target of rapamycin kinase; MTORC1: mechanistic target of rapamycin kinase complex 1; MTORC2: mechanistic target of rapamycin kinase complex 2; O2.-: superoxide; OMA1: OMA1 zinc metallopeptidase; OPA1: OPA1 mitochondrial dynamin like GTPase; RICTOR: RPTOR independent companion of MTOR complex 2; ROS: reactive oxygen species; RPTOR/raptor: regulatory associated protein of MTOR complex 1; SOD1: superoxide dismutase 1; SOD2: superoxide dismutase 2; SQSTM1/p62: sequestosome 1; VDAC1: voltage dependent anion channel 1; VDAC2: voltage dependent anion channel 2.


Asunto(s)
Neoplasias Colorrectales , Diana Mecanicista del Complejo 2 de la Rapamicina , Mitocondrias , Mitofagia , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas p21(ras) , Especies Reactivas de Oxígeno , Humanos , Especies Reactivas de Oxígeno/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Mitofagia/efectos de los fármacos , Mitofagia/genética , Mitofagia/fisiología , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Mutación/genética , Transducción de Señal/efectos de los fármacos , Línea Celular Tumoral , Fosforilación/efectos de los fármacos
3.
Trends Microbiol ; 30(7): 679-692, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35063304

RESUMEN

Through oxidative phosphorylation, mitochondria play a central role in energy production and are an important production source of reactive oxygen species (ROS). Not surprisingly, viruses have evolved to exploit this organelle in order to support their infection cycle. Beyond its role in the cellular antiviral response, induction of oxidative stress has emerged as a common strategy employed by many viruses to promote their replication. Here, we review the key molecular mechanisms employed by viruses to interact with mitochondria and induce oxidative stress. Furthermore, we discuss how viruses benefit from increased ROS levels, how they control ROS production to maintain a favorable redox environment, and how they cope with ROS-mediated cell death.


Asunto(s)
Mitocondrias , Virosis , Estrés del Retículo Endoplásmico , Humanos , Oxidación-Reducción , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Virosis/metabolismo
4.
Int Rev Cell Mol Biol ; 352: 189-214, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32334816

RESUMEN

Aberrant cell death signaling and oxidative stress are implicated in myriad of human pathological states such as neurodegenerative, cardiovascular, metabolic and liver diseases, as well as drug-induced toxicities. While regulated cell death and mild oxidative stress are essential during normal tissue homeostasis, deregulated signaling can trigger massive depletion in a particular cell type and/or damage tissues and impair organ function with deleterious consequences that manifest as disease states. If regeneration cannot restore tissue homeostasis, the severity of the disease correlates with the extent of cell loss. Cell death can be executed via multiple modalities such as apoptosis, necrosis, pyroptosis, necroptosis and ferroptosis, depending on cell autonomous mechanisms (e.g., reactive oxygen species production, calcium overload and altered proteostasis) and/or non-cell autonomous processes (e.g., environmental stress, irradiation, chemotherapeutic agents, inflammation and pathogens). Accordingly, the inhibition of aberrant cell death and oxidative stress together with activation of autophagy, a regulated self-degradation process, are progressively emerging as relevant cytoprotective strategies to sustain homeostasis. In this review, we summarize the current literature on the crosstalk between cellular redox state and cell fate signaling, specifically from the standpoint of autophagy and its role in the maintenance of tissue/organ homeostasis via regulating oxidative stress and the potential implications for the design of novel therapeutic strategies.


Asunto(s)
Autofagia , Enfermedades Neurodegenerativas/metabolismo , Transducción de Señal , Animales , Humanos , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo
5.
Cancers (Basel) ; 12(3)2020 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-32131385

RESUMEN

Deregulated cellular apoptosis is a hallmark of cancer and chemotherapy resistance. The B-cell lymphoma 2 (BCL-2) protein family members are sentinel molecules that regulate the mitochondrial apoptosis machinery and arbitrate cell fate through a delicate balance between pro- and anti-apoptotic factors. The recognition of the anti-apoptotic BCL2 gene as an oncogenic driver in hematological malignancies has directed attention toward unraveling the biological significance of each of the BCL-2 superfamily members in cancer progression and garnered interest in the targeting of apoptosis in cancer therapy. Accordingly, the approval of venetoclax (ABT-199), a small molecule BCL-2 inhibitor, in patients with chronic lymphocytic leukemia and acute myeloid leukemia has become the proverbial torchbearer for novel candidate drug approaches selectively targeting the BCL-2 superfamily. Despite the inspiring advances in this field, much remains to be learned regarding the optimal therapeutic context for BCL-2 targeting. Functional assays, such as through BH3 profiling, may facilitate prediction of treatment response, development of drug resistance and shed light on rational combinations of BCL-2 inhibitors with other branches of cancer therapy. This review summarizes the pathological roles of the BCL-2 family members in cancer, discusses the current landscape of their targeting in clinical practice, and highlights the potential for future therapeutic inroads in this important area.

6.
Redox Biol ; 30: 101403, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31954371

RESUMEN

The death inhibitory proteins, cFLIP and Bcl-2, canonically act at different steps to regulate receptor-mediated apoptosis in cancer cells. Here we report that pharmacological or genetic means to effect an increase in intracellular superoxide result in cFLIP upregulation. Interestingly, Bcl-2 overexpression is associated with a concomitant increase in cFLIP, and reducing superoxide sensitizes Bcl-2 overexpressing cancer cells to receptor-mediated apoptosis via downregulation of cFLIP. Moreover, inhibiting glycolytic flux overcomes apoptosis resistance by superoxide-dependent downregulation of cFLIP. Superoxide-induced upregulation of cFLIP is a function of enhanced transcription, as evidenced by increases in cFLIP promoter activity and mRNA abundance. The positive effect of superoxide on cFLIP is mediated through its reaction with nitric oxide to generate peroxynitrite. Corroborating these findings in cell lines, subjecting primary cells derived from lymphoma patients to glucose deprivation ex vivo, as a means to decrease superoxide, not only reduced cFLIP expression but also significantly enhanced death receptor sensitivity. Based on this novel mechanistic insight into the redox regulation of cancer cell fate, modulation of intracellular superoxide could have potential therapeutic implications in cancers in which these two death inhibitory proteins present a therapeutic challenge.


Asunto(s)
Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Linfoma/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Superóxidos/metabolismo , Regulación hacia Arriba , Línea Celular Tumoral , Supervivencia Celular , Regulación Neoplásica de la Expresión Génica , Glucólisis , Humanos , Linfoma/genética , Óxido Nítrico/metabolismo , Regiones Promotoras Genéticas , Transducción de Señal
7.
Sci Rep ; 10(1): 18837, 2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-33139717

RESUMEN

Sepsis is a potentially fatal condition triggered by systemic inflammatory response to infection. Due to the heightened immune reactivity and multi-organ pathology, treatment options are limited and several clinical trials have not produced the desired outcome, hence the interest in the discovery of novel therapeutic strategies. The polyphenol resveratrol (RSV) has shown promise against several pathological states, including acute and chronic inflammation. In this study, we evaluated its therapeutic potential in a murine model of sepsis and in patients undergoing transrectal ultrasound biopsy. RSV was able to inhibit lipopolysaccharide (LPS) stimulated inflammatory responses through blocking Phospholipase D (PLD) and its downstream signaling molecules SphK1, ERK1/2 and NF-κB. In addition, RSV treatment resulted in the downregulation of MyD88, an adaptor molecule in the TLR4 signaling pathway, and this effect at least in part, involved RSV-induced autophagy. Notably, RSV protected mice against polymicrobial septic shock induced upon cecal ligation and puncture, and inhibited pro-inflammatory cytokine production by human monocytes from transrectal ultrasound (TRUS) biopsy patients. Together, these findings demonstrate the immune regulatory activity of RSV and highlight its therapeutic potential in the management of sepsis.


Asunto(s)
Inflamación/tratamiento farmacológico , Inflamación/etiología , Resveratrol/farmacología , Resveratrol/uso terapéutico , Sepsis/tratamiento farmacológico , Receptor Toll-Like 4 , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Masculino , Ratones Endogámicos C57BL , Monocitos/metabolismo , Sepsis/etiología , Sepsis/inmunología , Sepsis/prevención & control , Transducción de Señal
8.
Biochem Pharmacol ; 162: 79-88, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30689981

RESUMEN

The experimental validation of the existence of cancer stem cells (CSC) has had a significant impact on our understanding of the cellular mechanisms and signaling networks involved in the process of carcinogenesis and its progression. These findings provide insights into the critical role that tumor microenvironment and metabolism play in the acquisition of the drug resistance phenotype as well as provide potential targets for therapeutic exploitation. Here we briefly review the literature on the involvement of key signaling pathways such as Wnt/ß-catenin, Notch, Hedgehog and STAT3 in the appearance of cancer cells with stem cells-like characteristics. In addition, we also highlight some of the recent therapeutic strategies used to target these pathways as well as approaches aiming to specifically target CSCs through their distinctive metabolic features.


Asunto(s)
Antineoplásicos/uso terapéutico , Manejo de la Enfermedad , Neoplasias/tratamiento farmacológico , Células Madre Neoplásicas/fisiología , Fenotipo , Microambiente Tumoral/fisiología , Animales , Antineoplásicos/farmacología , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/fisiología
9.
Redox Biol ; 20: 307-320, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30390545

RESUMEN

Negative Pressure Wound Therapy (NPWT), a widely used modality in the management of surgical and trauma wounds, offers clear benefits over conventional wound healing strategies. Despite the wide-ranging effects ascribed to NPWT, the precise molecular mechanisms underlying the accelerated healing supported by NPWT remains poorly understood. Notably, cellular redox status-a product of the balance between cellular reactive oxygen species (ROS) production and anti-oxidant defense systems-plays an important role in wound healing and dysregulation of redox homeostasis has a profound effect on wound healing. Here we investigated potential links between the use of NPWT and the regulation of antioxidant mechanisms. Using patient samples and a rodent model of acute injury, we observed a significant accumulation of MnSOD protein as well as higher enzymatic activity in tissues upon NPWT. As a proof of concept and to outline the important role of SOD activity in wound healing, we replaced NPWT by the topical application of a MnSOD mimetic, Mn(III) meso-tetrakis(N-ethylpyridinium-2-yl)porphyrin (MnTE-2-PyP5+, MnE, BMX-010, AEOl10113) in the rodent model. We observed that MnE is a potent wound healing enhancer as it appears to facilitate the formation of new tissue within the wound bed and consequently advances wound closure by two days, compared to the non-treated animals. Taken together, these results show for the first time a link between NPWT and regulation of antioxidant mechanism through the maintenance of MnSOD activity. Additionally this discovery outlined the potential role of MnSOD mimetics as topical agents enhancing wound healing.


Asunto(s)
Terapia de Presión Negativa para Heridas , Superóxido Dismutasa/metabolismo , Cicatrización de Heridas , Administración Tópica , Animales , Antioxidantes/metabolismo , Mimetismo Biológico , Biomimética , Terapia Combinada , Manejo de la Enfermedad , Modelos Animales de Enfermedad , Activación Enzimática , Humanos , Metaloporfirinas/administración & dosificación , Ratas , Superóxido Dismutasa/administración & dosificación , Resultado del Tratamiento , Cicatrización de Heridas/efectos de los fármacos
10.
FEBS Lett ; 582(20): 3045-51, 2008 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-18687331

RESUMEN

Bax insertion into the mitochondrial outer membrane is essential for the implementation of apoptosis. However, little is known about the first stage of Bax integration into the mitochondrial outer membrane. We have recently shown that TOM22, a mitochondrial outer membrane receptor, is important for insertion, although other reports have suggested that only mitochondrial lipids are involved in this process. Here, we show that monomers, but not dimers, of Bax require the presence of TOM22 and TOM40 to integrate into mitochondria. In addition we show that once inserted into the membrane, Bax can act as a receptor for cytosolic Bax.


Asunto(s)
Apoptosis , Membranas Mitocondriales/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Animales , Dimerización , Proteínas de Transporte de Membrana/metabolismo , Mitocondrias Hepáticas/metabolismo , Proteínas de Transporte de Membrana Mitocondrial , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Proteínas Mitocondriales/antagonistas & inhibidores , Proteínas Mitocondriales/metabolismo , Ratas
11.
J Clin Invest ; 115(11): 3117-27, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16276417

RESUMEN

Panton-Valentine leukocidin (PVL) is a pore-forming toxin secreted by Staphylococcus aureus that has recently been associated with necrotizing pneumonia. In the present study, we report that in vitro, PVL induces polymorphonuclear cell death by necrosis or by apoptosis, depending on the PVL concentration. PVL-induced apoptosis was associated with a rapid disruption of mitochondrial homeostasis and activation of caspase-9 and caspase-3, suggesting that PVL-induced apoptosis is preferentially mediated by the mitochondrial pathway. Polymorphonuclear cell exposure to PVL leads to mitochondrial localization of the toxin, whereas Bax, 1 of the 2 essential proapoptotic members of the Bcl-2 family, was still localized in the cytosol. Addition of PVL to isolated mitochondria induced the release of the apoptogenic proteins cytochrome c and Smac/DIABLO. Therefore, we suggest that PVL, which belongs to the pore-forming toxin family, could act at the mitochondrion level by creating pores in the mitochondrial outer membrane. Furthermore, LukS-PV, 1 of the 2 components of PVL, was detected in lung sections of patients with necrotizing pneumonia together with DNA fragmentation, suggesting that PVL induces apoptosis in vivo and thereby is directly involved in the pathophysiology of necrotizing pneumonia.


Asunto(s)
Apoptosis/efectos de los fármacos , Leucocidinas/farmacología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Neutrófilos/citología , Neutrófilos/efectos de los fármacos , Proteína X Asociada a bcl-2/fisiología , Proteínas Reguladoras de la Apoptosis , Toxinas Bacterianas , Membrana Celular/fisiología , Células Cultivadas , Citocromos c/metabolismo , Exotoxinas , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Cinética , Pulmón/microbiología , Pulmón/patología , Mitocondrias/fisiología , Proteínas Mitocondriales/metabolismo , Necrosis , Neutrófilos/microbiología , Neumonía Estafilocócica/microbiología , Neumonía Estafilocócica/patología , Staphylococcus aureus/fisiología
12.
Antioxid Redox Signal ; 29(16): 1553-1588, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29186971

RESUMEN

SIGNIFICANCE: There is evidence to implicate reactive oxygen species (ROS) in tumorigenesis and its progression. This has been associated with the interplay between ROS and oncoproteins, resulting in enhanced cellular proliferation and survival. Recent Advances: To date, studies have investigated specific contributions of the crosstalk between ROS and signaling networks in cancer initiation and progression. These investigations have challenged the established dogma of ROS as agents of cell death by demonstrating a secondary function that fuels cell proliferation and survival. Studies have thus identified (onco)proteins (Bcl-2, STAT3/5, RAS, Rac1, and Myc) in manipulating ROS level as well as exploiting an altered redox environment to create a milieu conducive for cancer formation and progression. CRITICAL ISSUES: Despite these advances, drug resistance and its association with an altered redox metabolism continue to pose a challenge at the mechanistic and clinical levels. Therefore, identifying specific signatures, altered protein expressions, and modifications as well as protein-protein interplay/function could not only enhance our understanding of the redox networks during cancer initiation and progression but will also provide novel targets for designing specific therapeutic strategies. FUTURE DIRECTIONS: Not only a heightened realization is required to unravel various gene/protein networks associated with cancer formation and progression, particularly from the redox standpoint, but there is also a need for developing more sensitive tools for assessing cancer redox metabolism in clinical settings. This review attempts to summarize our current knowledge of the crosstalk between oncoproteins and ROS in promoting cancer cell survival and proliferation and treatment strategies employed against these oncoproteins. Antioxid. Redox Signal.


Asunto(s)
Neoplasias/metabolismo , Proteínas Oncogénicas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Animales , Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Proteínas Oncogénicas/antagonistas & inhibidores , Oxidación-Reducción , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos
13.
Antioxid Redox Signal ; 24(14): 781-94, 2016 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-26714745

RESUMEN

AIMS: We recently reported the death-inducing activity of a small-molecule compound, C1, which triggered reactive oxygen species (ROS)-dependent autophagy-associated apoptosis in a variety of human cancer cell lines. In this study, we examine the ability of the compound to specifically target cancer cells harboring mutant KRAS with minimal activity against wild-type (WT) RAS-expressing cells. RESULTS: HCT116 cells expressing mutated KRAS are susceptible, while the WT-expressing HT29 cells are resistant. Interestingly, C1 triggers activation of mutant RAS, which results in the downstream phosphorylation and activation of AKT/PKB. Gene knockdown of KRAS or AKT or their pharmacological inhibition resulted in the abrogation of C1-induced ROS production and rescued tumor colony-forming ability. We also made use of HCT116 mutant KRAS knockout (KO) cells, which express only a single WT KRAS allele. Exposure of KO cells to C1 failed to increase mitochondrial ROS and cell death, unlike the parental cells harboring mutant KRAS. Similarly, mutant KRAS-transformed prostate epithelial cells (RWPE-1-RAS) were more sensitive to the ROS-producing and death-inducing effects of C1 than the vector only expressing RWPE-1 cells. An in vivo model of xenograft tumors generated with HCT116 KRAS(WT/MUT) or KRAS(WT/-) cells showed the efficacy of C1 treatment and its ability to affect the relative mitotic index in tumors harboring KRAS mutant. INNOVATION AND CONCLUSION: These data indicate a synthetic lethal effect against cells carrying mutant KRAS, which could have therapeutic implications given the paucity of KRAS-specific chemotherapeutic strategies. Antioxid. Redox Signal. 24, 781-794.


Asunto(s)
Antineoplásicos/farmacología , Etilenotiourea/análogos & derivados , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Supervivencia Celular/efectos de los fármacos , Activación Enzimática , Etilenotiourea/farmacología , Expresión Génica , Células HCT116 , Células HT29 , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Mutación Missense , Proteínas Proto-Oncogénicas p21(ras)/genética , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Cell Signal ; 26(9): 1928-34, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24794530

RESUMEN

The proteins Bax and Bak are central in the execution phase of apoptosis; however, little is known about the partners involved in the control of this complex process. Here, we show that mitochondrial Bak is incorporated into a VDAC2/Mtx1/Mtx2 multi-protein complex in both resting and dying cells. VDAC2 is a porin that has previously been described as a partner of Bak while Mtx1 and Mtx2 are two proteins of the mitochondrial sorting and assembly machinery (SAM) that have been implicated in TNF-induced apoptosis. We show that, after the induction of apoptosis, Bak switches from its association with Mtx2 and VDAC2 to interact with Mtx1.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Línea Celular Tumoral , Cicloheximida/toxicidad , Células HeLa , Humanos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Proteínas de Transporte de Membrana Mitocondrial , Proteínas Mitocondriales/antagonistas & inhibidores , Proteínas Mitocondriales/genética , Unión Proteica , Transporte de Proteínas , Proteínas/antagonistas & inhibidores , Proteínas/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos , Canal Aniónico 2 Dependiente del Voltaje/antagonistas & inhibidores , Canal Aniónico 2 Dependiente del Voltaje/genética , Canal Aniónico 2 Dependiente del Voltaje/metabolismo , Proteína X Asociada a bcl-2/antagonistas & inhibidores , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
15.
Mitochondrion ; 13(3): 155-62, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-22750269

RESUMEN

Recent advances have highlighted the complex web of biological mechanisms and pathways involved in oncogenic transformation and maintenance of the cancer phenotype. To that end, a number of key factors have been identified and thoroughly investigated over the past couple of decades, such as redox regulation of cell fate decisions, cellular metabolism and bioenergetics, autophagy induction as a survival signal, and how these pathways interplay with oncogene-induced transformation. This has been particularly well documented for oncoprotein Ras-induced carcinogenesis, and recent reports provide ample evidence to indicate a well-coordinated crosstalk between these diverse cellular pathways in the process of cancer initiation and progression. Here we provide a brief summary of the recent advances in the field to illustrate the dual role of autophagy as a tumor suppressor and as a survival mechanism required for cancer maintenance as well as its implication in the complex relationship between Ras-mediated carcinogenesis, mitochondrial metabolism, cellular redox status and bioenergetics.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Mitocondrias/metabolismo , Neoplasias/metabolismo , Proteína Oncogénica p21(ras)/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Autofagia/genética , Supervivencia Celular/genética , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Metabolismo Energético/genética , Humanos , Mitocondrias/genética , Neoplasias/genética , Neoplasias/patología , Proteína Oncogénica p21(ras)/genética , Oxidación-Reducción , Transducción de Señal/genética
16.
J Vis Exp ; (81): e50465, 2013 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-24300406

RESUMEN

Glycogen is the main energetic polymer of glucose in vertebrate animals and plays a crucial role in whole body metabolism as well as in cellular metabolism. Many methods to detect glycogen already exist but only a few are quantitative. We describe here a method using the Abcam Glycogen assay kit, which is based on specific degradation of glycogen to glucose by glucoamylase. Glucose is then specifically oxidized to a product that reacts with the OxiRed probe to produce fluorescence. Titration is accurate, sensitive and can be achieved on cell extracts or tissue sections. However, in contrast to other techniques, it does not give information about the distribution of glycogen in the cell. As an example of this technique, we describe here the titration of glycogen in two cell lines, Chinese hamster lung fibroblast CCL39 and human colon carcinoma LS174, incubated in normoxia (21% O2) versus hypoxia (1% O2). We hypothesized that hypoxia is a signal that prepares cells to synthesize and store glycogen in order to survive(1).


Asunto(s)
Glucógeno/análisis , Animales , Hipoxia de la Célula/fisiología , Línea Celular , Línea Celular Tumoral , Neoplasias del Colon/química , Neoplasias del Colon/metabolismo , Cricetinae , Cricetulus , Fibroblastos/química , Fibroblastos/metabolismo , Fluorescencia , Glucosa/análisis , Glucosa/química , Glucosa/metabolismo , Glucógeno/química , Glucógeno/metabolismo , Humanos , Hidrólisis , Oxidación-Reducción , Juego de Reactivos para Diagnóstico , Volumetría/métodos
17.
J Cancer Res Clin Oncol ; 138(3): 463-82, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22186935

RESUMEN

PURPOSE: Since primary tumor cells from patients have been used as a model for assessment of drug response for individual patients, this study aims to evaluate the reliability of such a model in colorectal cancer (CRC) in predicting the response of tumor tissues through comparison of their expression profiles. METHODS: Establishment of primary cultures from tissues obtained surgically from CRC patients allowed us to study the gene expression differences between normal and tumor tissues as well as primary cultures derived from the tumor mass. The tissues comparison highlights the molecular characteristics of tumors, while the comparison between primary tumor cells versus normal and tumor tissues allowed us to identify alterations associated with the establishment of culture. Genes-drug association analyses allowed us to fine-tune our expectations while using primary culture as a model for drug assessment. RESULTS: Comparison between tumor cultures and original tissues through functional analyses showed the deregulations caused by culture establishment. Investigating the impact of such changes in genes-drug associations to identify the potential alterations in drug response, we found that primary cultures may have increased susceptibility toward paclitaxel, but reduced susceptibility toward analogues of fluorouracil compared with original tumors. CONCLUSIONS: Response of primary tumor cells toward different drugs is not linearly associated to tumor tissues. Our results highlight the importance to account for the discrepancy in responses between the primary tumor cells and original counterparts in order to provide clinicians with important insights to improve selection of drugs for individual patients based on in vitro assays.


Asunto(s)
Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Antineoplásicos/farmacología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células Tumorales Cultivadas/efectos de los fármacos , Adenocarcinoma/tratamiento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Camptotecina/análogos & derivados , Camptotecina/farmacología , Capecitabina , Cisplatino/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Regulación hacia Abajo/efectos de los fármacos , Doxorrubicina/farmacología , Etopósido/farmacología , Floxuridina/farmacología , Técnica del Anticuerpo Fluorescente , Fluorouracilo/análogos & derivados , Fluorouracilo/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Inestabilidad Genómica/efectos de los fármacos , Humanos , Irinotecán , Metafase , Paclitaxel/farmacología , Reacción en Cadena de la Polimerasa , Valor Predictivo de las Pruebas , Análisis por Matrices de Proteínas , Reproducibilidad de los Resultados , Tegafur/farmacología , Análisis de Matrices Tisulares , Regulación hacia Arriba/efectos de los fármacos , Uracilo/farmacología
18.
Front Oncol ; 2: 18, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22649778

RESUMEN

The hypoxia-inducible factor 1 (HIF-1), in addition to genetic and epigenetic changes, is largely responsible for alterations in cell metabolism in hypoxic tumor cells. This transcription factor not only favors cell proliferation through the metabolic shift from oxidative phosphorylation to glycolysis and lactic acid production but also stimulates nutrient supply by mediating adaptive survival mechanisms. In this study we showed that glycogen synthesis is enhanced in non-cancer and cancer cells when exposed to hypoxia, resulting in a large increase in glycogen stores. Furthermore, we demonstrated that the mRNA and protein levels of the first enzyme of glycogenesis, phosphoglucomutase1 (PGM1), were increased in hypoxia. We showed that induction of glycogen storage as well as PGM1 expression were dependent on HIF-1 and HIF-2. We established that hypoxia-induced glycogen stores are rapidly mobilized in cells that are starved of glucose. Glycogenolysis allows these "hypoxia-preconditioned" cells to confront and survive glucose deprivation. In contrast normoxic control cells exhibit a high rate of cell death following glucose removal. These findings point to the important role of hypoxia and HIF in inducing mechanisms of rapid adaptation and survival in response to a decrease in oxygen tension. We propose that a decrease in pO(2) acts as an "alarm" that prepares the cells to face subsequent nutrient depletion and to survive.

19.
Curr Opin Genet Dev ; 21(1): 67-72, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21074987

RESUMEN

The hypoxia-inducible factor (HIF-1), in addition to genetic and epigenetic changes, is largely responsible for alterations in cell metabolism in hypoxic tumour cells. This transcription factor not only favours cell proliferation through the metabolic shift from oxidative phosphorylation to glycolysis and lactic acid production but also stimulates nutrient supply by mediating adaptive survival mechanisms. These include epithelial-mesenchymal transition, angiogenesis, autophagy, and synthesis and storage of lipid and glycogen. HIF-1 also ensures survival by correcting tumour acidosis via increased expression of the carbonic anhydrase CA IX and the lactate/H+ symporter MCT4. The targeting of key HIF-1-mediated steps, responsible for exacerbated glycolysis and pHi-control, and of the 'guardian of cellular energy' AMP-kinase should offer novel therapeutic opportunities to fight cancer.


Asunto(s)
Metabolismo Energético , Neoplasias/metabolismo , Hipoxia de la Célula , Supervivencia Celular , Humanos , Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias/patología , Neoplasias/terapia , Estrés Fisiológico
20.
Mol Cell Biol ; 29(10): 2570-81, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19273585

RESUMEN

While hypoxia-inducible factor (HIF) is a major actor in the cell survival response to hypoxia, HIF also is associated with cell death. Several studies implicate the HIF-induced putative BH3-only proapoptotic genes bnip3 and bnip3l in hypoxia-mediated cell death. We, like others, do not support this assertion. Here, we clearly demonstrate that the hypoxic microenvironment contributes to survival rather than cell death by inducing autophagy. The ablation of Beclin1, a major actor of autophagy, enhances cell death under hypoxic conditions. In addition, the ablation of BNIP3 and/or BNIP3L triggers cell death, and BNIP3 and BNIP3L are crucial for hypoxia-induced autophagy. First, while the small interfering RNA-mediated ablation of either BNIP3 or BNIP3L has little effect on autophagy, the combined silencing of these two HIF targets suppresses hypoxia-mediated autophagy. Second, the ectopic expression of both BNIP3 and BNIP3L in normoxia activates autophagy. Third, 20-mer BH3 peptides of BNIP3 or BNIP3L are sufficient in initiating autophagy in normoxia. Herein, we propose a model in which the atypical BH3 domains of hypoxia-induced BNIP3/BNIP3L have been designed to induce autophagy by disrupting the Bcl-2-Beclin1 complex without inducing cell death. Hypoxia-induced autophagy via BNIP3 and BNIP3L is clearly a survival mechanism that promotes tumor progression.


Asunto(s)
Autofagia/fisiología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Hipoxia de la Célula , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Proteínas de la Membrana/metabolismo , Fragmentos de Péptidos/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Beclina-1 , Línea Celular Tumoral , Fibroblastos/citología , Fibroblastos/fisiología , Regulación de la Expresión Génica , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Fragmentos de Péptidos/genética , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Alineación de Secuencia , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA