Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
J Biol Chem ; 295(20): 6946-6957, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32265301

RESUMEN

The incidence of pancreatic cancer increases with age, suggesting that chronological aging is a significant risk factor for this disease. Fibroblasts are the major nonmalignant cell type in the stroma of human pancreatic ductal adenocarcinoma (PDAC). In this study, we investigated whether the chronological aging of normal human fibroblasts (NHFs), a previously underappreciated area in pancreatic cancer research, influences the progression and therapeutic outcomes of PDAC. Results from experiments with murine xenografts and 2D and 3D co-cultures of NHFs and PDAC cells revealed that older NHFs stimulate proliferation of and confer resistance to radiation therapy of PDAC. MS-based metabolite analysis indicated that older NHFs have significantly increased arachidonic acid 12-lipoxygenase (ALOX12) expression and elevated levels of its mitogenic metabolite, 12-(S)-hydroxy-5,8,10,14-eicosatetraenoic acid (12-(S)-HETE) compared with their younger counterparts. In co-cultures with older rather than with younger NHFs, PDAC cells exhibited increases in mitogen-activated protein kinase signaling and cellular metabolism, as well as a lower oxidation state that correlated with their enhanced proliferation and resistance to radiation therapy. Expression of ALOX12 was found to be significantly lower in PDAC cell lines and tumor biopsies, suggesting that PDAC cells rely on a stromal supply of mitogens for their proliferative needs. Pharmacological (hydroxytyrosol) and molecular (siRNA) interventions of ALOX12 in older NHFs suppressed their ability to stimulate proliferation of PDAC cells. We conclude that chronological aging of NHFs contributes to PDAC progression and that ALOX12 and 12-(S)-HETE may be potential stromal targets for interventions that seek to halt progression and improve therapy outcomes.


Asunto(s)
Araquidonato 12-Lipooxigenasa/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Senescencia Celular , Ácidos Hidroxieicosatetraenoicos/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Pancreáticas/metabolismo , Animales , Araquidonato 12-Lipooxigenasa/genética , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Ratones , Proteínas de Neoplasias/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Células del Estroma/metabolismo , Células del Estroma/patología
2.
Int J Mol Sci ; 22(19)2021 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-34639220

RESUMEN

Interest in the use of pharmacological ascorbate as a treatment for cancer has increased considerably since it was introduced by Cameron and Pauling in the 1970s. Recently, pharmacological ascorbate has been used in preclinical and early-phase clinical trials as a selective radiation sensitizer in cancer. The results of these studies are promising. This review summarizes data on pharmacological ascorbate (1) as a safe and efficacious adjuvant to cancer therapy; (2) as a selective radiosensitizer of cancer via a mechanism involving hydrogen peroxide; and (3) as a radioprotector in normal tissues. Additionally, we present new data demonstrating the ability of pharmacological ascorbate to enhance radiation-induced DNA damage in glioblastoma cells, facilitating cancer cell death. We propose that pharmacological ascorbate may be a general radiosensitizer in cancer therapy and simultaneously a radioprotector of normal tissue.


Asunto(s)
Ácido Ascórbico/farmacología , Peróxido de Hidrógeno/farmacología , Neoplasias/radioterapia , Estrés Oxidativo/efectos de los fármacos , Tolerancia a Radiación/efectos de los fármacos , Fármacos Sensibilizantes a Radiaciones/farmacología , Animales , Antioxidantes/farmacología , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Oxidantes/farmacología , Especies Reactivas de Oxígeno/metabolismo
3.
Int J Mol Sci ; 20(1)2019 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-30626124

RESUMEN

Combination radiation and chemotherapy are commonly used to treat locoregionally advanced head and neck squamous cell carcinoma (HNSCC). Aggressive dosing of these therapies is significantly hampered by side effects due to normal tissue toxicity. Selenium represents an adjuvant that selectively sensitizes cancer cells to these treatments modalities, potentially by inducing lipid peroxidation (LPO). This study investigated whether one such selenium compound, methylseleninic acid (MSA), induces LPO and radiation sensitivity in HNSCC cells. Results from 4,4-difluoro-4-bora-3a,4a-diaza-S-indacene (BODIPY) C11 oxidation and ferric thiocyanate assays revealed that MSA induced LPO in cells rapidly and persistently. Propidium iodide (PI) exclusion assay found that MSA was more toxic to cancer cells than other related selenium compounds; this toxicity was abrogated by treatment with α-tocopherol, an LPO inhibitor. MSA exhibited no toxicity to normal fibroblasts at similar doses. MSA also sensitized HNSCC cells to radiation as determined by clonogenic assay. Intracellular glutathione in cancer cells was depleted following MSA treatment, and supplementation of the intracellular glutathione pool with N-acetylcysteine sensitized cells to MSA. The addition of MSA to a cell-free solution of glutathione resulted in an increase in oxygen consumption, which was abrogated by catalase, suggesting the formation of H2O2. Results from this study identify MSA as an inducer of LPO, and reveal its capability to sensitize HNSCC to radiation. MSA may represent a potent adjuvant to radiation therapy in HNSCC.


Asunto(s)
Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Peroxidación de Lípido/efectos de los fármacos , Compuestos de Organoselenio/farmacología , Tolerancia a Radiación/efectos de los fármacos , Acetilcisteína/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Línea Celular Tumoral , Rayos gamma , Glutatión/metabolismo , Humanos , Peroxidación de Lípido/efectos de la radiación , Consumo de Oxígeno/efectos de los fármacos , Tolerancia a Radiación/efectos de la radiación , Factores de Tiempo
4.
Mol Biol Rep ; 43(1): 31-9, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26671656

RESUMEN

The rebuilding of the connective tissue during wound healing requires the recruitment of fibroblasts to the wound area as well as reentry of quiescent fibroblasts to the proliferative cycle. Whether this process can be modulated by a small molecular weight thiol antioxidant N-acetyl-L-cysteine (NAC) was tested in normal human skin fibroblasts (NHFs) using a uni-directional wound healing assay. NAC treated cells demonstrated a decreased migration rate but increased number of proliferating cells recruited into the wound area post wounding. Fifteen day quiescent control and NAC treated NHFs were re-plated at a lower density and cell numbers counted at different days post-plating. Interestingly, NAC treated cells exhibited increased cellular proliferation indicated by both decreased cell population doubling time and increased S phase cells. NAC treated cells demonstrated decreased steady state levels of reactive oxygen species as well as increased protein and activity levels of manganese superoxide dismutase (MnSOD). NAC treatment failed to induce proliferation in quiescent cells lacking MnSOD expression. These results demonstrate that NAC enhanced the recruitment of quiescent NHFs into proliferation cycle during wound healing. Our results also suggest that the wound healing properties of NAC might be due to its ability to induce and enhance MnSOD expression and activity. Altogether, these findings suggest NAC might be potentially developed as a dietary intervention to improve tissue injury in animals and humans.


Asunto(s)
Acetilcisteína/farmacología , Fibroblastos/efectos de los fármacos , Superóxido Dismutasa/metabolismo , Cicatrización de Heridas/efectos de los fármacos , Animales , Antioxidantes/farmacología , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Ratones , Ratones Noqueados , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo
5.
Radiat Res ; 200(5): 444-455, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37758045

RESUMEN

Pharmacological ascorbate (P-AscH-, high dose, intravenous vitamin C) preferentially sensitizes human pancreas ductal adenocarcinoma (PDAC) cells to radiation-induced toxicity compared to non-tumorigenic epithelial cells. Radiation-induced G2-checkpoint activation contributes to the resistance of cancer cells to DNA damage induced toxicity. We hypothesized that P-AscH- induced radio-sensitization of PDAC cells is mediated by perturbations in the radiation induced activation of the G2-checkpoint pathway. Both non-tumorigenic pancreatic ductal epithelial and PDAC cells display decreased clonogenic survival and increased doubling times after radiation treatment. In contrast, the addition of P-AscH- to radiation increases clonogenic survival and decreases the doubling time of non-tumorigenic epithelial cells but decreasing clonogenic survival and increasing the doubling time of PDAC cells. Results from the mitotic index and propidium iodide assays showed that while the P-AscH- treatments did not affect radiation-induced G2-checkpoint activation, it enhanced G2-accumulation. The addition of catalase reverses the increases in G2-accumulation, indicating a peroxide-mediated mechanism. In addition, P-AscH- treatment of PDAC cells suppresses radiation-induced accumulation of cyclin B1 protein levels. Both translational and post-translational pathways appear to regulate cyclin B1 protein levels after the combination treatment of PDAC cells with P-AscH- and radiation. The protein changes seen are reversed by the addition of catalase suggesting that hydrogen peroxide mediates P-AscH- induced radiation sensitization of PDAC cells by enhancing G2-accumulation and reducing cyclin B1 protein levels.


Asunto(s)
Antineoplásicos , Neoplasias Pancreáticas , Humanos , Catalasa/metabolismo , Catalasa/uso terapéutico , Peróxido de Hidrógeno/farmacología , Peróxido de Hidrógeno/metabolismo , Peróxido de Hidrógeno/uso terapéutico , Ciclina B1 , Línea Celular Tumoral , Neoplasias Pancreáticas/tratamiento farmacológico , Antineoplásicos/farmacología , Páncreas/metabolismo , Páncreas/patología , Neoplasias Pancreáticas
6.
Int J Radiat Oncol Biol Phys ; 115(4): 933-944, 2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36228747

RESUMEN

PURPOSE: Ataxia telangiectasia mutated kinase (ATM) inhibitors are potent radiosensitizers that regulate DNA damage responses and redox metabolism, but they have not been translated clinically because of the potential for excess normal tissue toxicity. Pharmacologic ascorbate (P-AscH-; intravenous administration achieving mM plasma concentrations) selectively enhances H2O2-induced oxidative stress and radiosensitization in tumors while acting as an antioxidant and mitigating radiation damage in normal tissues including the bowel. We hypothesized that P-AscH- could enhance the therapeutic index of ATM inhibitor-based chemoradiation by simultaneously enhancing the intended effects of ATM inhibitors in tumors and mitigating off-target effects in adjacent normal tissues. METHODS AND MATERIALS: Clonogenic survival was assessed in human (human colon tumor [HCT]116, SW480, HT29) and murine (CT26, MC38) colorectal tumor lines and normal cells (human umbilical vein endothelial cell, FHs74) after radiation ± DNA repair inhibitors ± P-AscH-. Tumor growth delay was assessed in mice with HCT116 or MC38 tumors after fractionated radiation (5 Gy × 3) ± the ATM inhibitor KU60019 ± P-AscH-. Intestinal injury, oxidative damage, and transforming growth factor ß immunoreactivity were quantified using immunohistochemistry after whole abdominal radiation (10 Gy) ± KU60019 ± P-AscH-. Cell cycle distribution and ATM subcellular localization were assessed using flow cytometry and immunohistochemistry. The role of intracellular H2O2 fluxes was assessed using a stably expressed doxycycline-inducible catalase transgene. RESULTS: KU60019 with P-AscH- enhanced radiosensitization in colorectal cancer models in vitro and in vivo by H2O2-dependent oxidative damage to proteins and enhanced DNA damage, abrogation of the postradiation G2 cell cycle checkpoint, and inhibition of ATM nuclear localization. In contrast, concurrent P-AscH- markedly reduced intestinal toxicity and oxidative damage with KU60019. CONCLUSIONS: We provide evidence that redox modulating drugs, such as P-AscH-, may facilitate the clinical translation of ATM inhibitors by enhancing tumor radiosensitization while simultaneously protecting normal tissues.


Asunto(s)
Ataxia Telangiectasia , Neoplasias Pancreáticas , Humanos , Animales , Ratones , Ácido Ascórbico/farmacología , Ácido Ascórbico/uso terapéutico , Peróxido de Hidrógeno , Línea Celular Tumoral , Neoplasias Pancreáticas/patología , Oxidación-Reducción , Índice Terapéutico , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Daño del ADN , Proteínas de Ciclo Celular/metabolismo
7.
Free Radic Biol Med ; 165: 421-434, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33561488

RESUMEN

Dihydroartemisinin (DHA) is an FDA-approved antimalarial drug that has been repurposed for cancer therapy because of its preferential antiproliferative effects on cancer versus normal cells. Mitochondria represent an attractive target for cancer therapy based on their regulatory role in proliferation and cell death. This study investigates whether DHA conjugated to innately fluorescent N-alkyl triphenylvinylpyridinium (TPVP) perturbs mitochondrial functions resulting in a differential toxicity of cancer versus normal cells. TPVP-DHA treatments resulted in a dose-dependent toxicity of human melanoma and pancreatic cancer cells, whereas normal human fibroblasts were resistant to this treatment. TPVP-DHA treatments resulted in a G1-delay of the cancer cell cycle, which was also associated with a significant inhibition of the mTOR-metabolic and ERK1/2-proliferative signaling pathways. TPVP-DHA treatments perturbed mitochondrial functions, which correlated with increases in mitochondrial fission. In summary, TPVP mediated mitochondrial targeting of DHA enhanced cancer cell toxicity by perturbing mitochondrial functions and morphology.


Asunto(s)
Antimaláricos , Artemisininas , Neoplasias , Antimaláricos/toxicidad , Apoptosis , Artemisininas/farmacología , Línea Celular Tumoral , Proliferación Celular , Humanos , Mitocondrias
8.
Radiat Res ; 195(3): 230-234, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33347596

RESUMEN

MR-linac technology enhances the precision of therapeutic radiation by clarifying the tumor-normal tissue interface and provides the potential for adaptive treatment planning. Accurate delineation of tumors on diagnostic magnetic resonance imaging (MRI) frequently requires gadolinium-based contrast agents (GBCAs). Despite generally being considered safe, previous literature suggests that GBCAs are capable of contrast-induced acute kidney injury (AKI). It is unclear if the risk for AKI is enhanced when GBCAs are administered concurrently with ionizing radiotherapy. During irradiation, gadolinium may be liberated from its chelator which may induce AKI. The goal of this work was to determine if radiation combined with GBCAs increased the incidence of AKI. Using a preclinical MRI-guided irradiation system, where MRI acquisitions and radiation delivery are performed in rapid succession, tumor-bearing mice with normal kidney function were injected with GBCA and treated with 2, 8 or 18 Gy irradiation. Renal function was assessed on days three and seven postirradiation to assess for AKI. No clinically relevant changes in blood urea nitrogen and creatinine were observed in any combination of GBCA and radiation dose. From these data, we conclude that GBCA in combination with radiation does not increase the risk for AKI in mice. Additional investigation of multiple doses of GBCA administered concurrently with irradiation is warranted to evaluate the risk of chronic kidney injury.


Asunto(s)
Lesión Renal Aguda/diagnóstico por imagen , Medios de Contraste/farmacología , Compuestos Organometálicos/farmacología , Radiación Ionizante , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/fisiopatología , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/efectos de los fármacos , Encéfalo/patología , Encéfalo/efectos de la radiación , Medios de Contraste/efectos adversos , Modelos Animales de Enfermedad , Gadolinio/efectos adversos , Gadolinio/farmacología , Humanos , Riñón/diagnóstico por imagen , Riñón/efectos de los fármacos , Riñón/patología , Riñón/efectos de la radiación , Imagen por Resonancia Magnética , Ratones , Compuestos Organometálicos/efectos adversos , Radioterapia Guiada por Imagen/efectos adversos , Radioterapia Guiada por Imagen/métodos
9.
Int J Radiat Oncol Biol Phys ; 108(1): 81-92, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32036006

RESUMEN

PURPOSE: The majority of colorectal cancers are resistant to cancer immune checkpoint inhibitors. Ionizing radiation (IR) and several radiosensitizers, including PARP inhibitors, can enhance responsiveness to immune checkpoint inhibitors by potentially complementary mechanisms of action. We assessed the ability of radiation and PARP inhibition to induce proimmunogenic changes in tumor cells and enhance their in vivo responsiveness to anti-PD-1 antibodies. METHODS AND MATERIALS: We performed a candidate drug screen and used flow cytometry to assess effects of the PARP inhibitor veliparib on IR-mediated changes in MHC-1 antigen presentation and surface localization of immune-modulating proteins including PD-L1 and calreticulin in colorectal cancer tumor models. Reverse transcription polymerase chain reaction was used to assess the effects of veliparib and radiation on the expression of proinflammatory and immunosuppressive cytokines. The ability of concurrent PARP inhibition and subablative doses of radiation therapy to enhance in vivo responsiveness to anti-PD-1 antibodies was assessed using unilateral flank-tumor models with or without T-cell depletion. RESULTS: Veliparib was a potent radiosensitizer in both cell lines. Radiation increased surface localization of MHC-1 and PD-L1 in a dose-dependent manner, and veliparib pretreatment significantly enhanced these effects with high (8 Gy) but not with lower radiation doses. Enhancement of MHC-1 and PD-L1 surface localization by IR and IR+ veliparib remained significant 1, 3, and 7 days after treatment. IR significantly increased delayed tumoral expression of proinflammatory cytokines interferon-Ƴ and CXCL10 but had no significant effect on the expression of IL-6 or TGF-ß. Concurrent administration of veliparib and subablative radiation therapy (8 Gy × 2) significantly prolonged anti-PD-1-mediated in vivo tumor growth delay and survival in both tumor models. Moreover, these effects were more pronounced in the microsatellite instability-mutated MC38 tumor model. Enhancement of anti-PD-1 mediated tumor growth delay with veliparib and IR was attenuated by CD8+ T-cell depletion. CONCLUSIONS: We provide preclinical evidence for a novel therapeutic strategy to enhance responsiveness of colorectal tumors to immune checkpoint inhibitors.


Asunto(s)
Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/radioterapia , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Animales , Presentación de Antígeno/efectos de los fármacos , Bencimidazoles/farmacología , Bencimidazoles/uso terapéutico , Transformación Celular Neoplásica , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Terapia Combinada , Femenino , Humanos , Masculino , Ratones , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Fármacos Sensibilizantes a Radiaciones/farmacología , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Resultado del Tratamiento
10.
Antioxidants (Basel) ; 9(2)2020 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-31991904

RESUMEN

This study used a nitroaliphatic chemistry approach to synthesize a novel artemisinin-derived carba-dimer (AG-1) and determined its anti-proliferative effects in human normal and cancer cells. AG-1 treatments selectively inhibit proliferation of cancer cells compared to normal human fibroblasts. Compared to artemisinin, AG-1 is more toxic to human breast, prostate, head-neck, pancreas and skin cancer cells; 50% inhibition (IC50) 123 µM in AG-1 vs. 290 µM in artemisinin-treated breast cancer cells. AG-1 treatment decreased (~ 5 folds) cyclin D1 protein expression that correlated with an increase in the percentage of cells in the G1-phase, suggesting a G1 delay. AG-1-induced toxicity was independent of the DNA damage at 72 h post-treatment, as measured by micronuclei frequency and H2AX protein levels. Results from electron paramagnetic resonance spectroscopy showed Fe-catalyzed formation of AG-1 carbon-centered radicals in a cell-free system. Flow cytometry analysis of H2DCF-DA oxidation showed a significant increase in the steady-state levels of reactive oxygen species (ROS) in AG-1-treated cells. Pre-treatment with N-acetyl-l-cysteine and antioxidant enzymes (superoxide dismutase and catalase) significantly suppressed AG-1-induced toxicity, suggesting that superoxide and hydrogen peroxide contribute to AG-1-induced toxicity in human cancer cells. AG-1 represents a novel class of anti-cancer drug that is more potent than its parent compound, artemisinin.

11.
Cancer Res ; 80(7): 1401-1413, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32041838

RESUMEN

Pharmacologic ascorbate treatment (P-AscH-, high-dose, intravenous vitamin C) results in a transient short-term increase in the flux of hydrogen peroxide that is preferentially cytotoxic to cancer cells versus normal cells. This study examines whether an increase in hydrogen peroxide is sustained posttreatment and potential mechanisms involved in this process. Cellular bioenergetic profiling following treatment with P-AscH- was examined in tumorigenic and nontumorigenic cells. P-AscH- resulted in sustained increases in the rate of cellular oxygen consumption (OCR) and reactive oxygen species (ROS) in tumor cells, with no changes in nontumorigenic cells. Sources for this increase in ROS and OCR were DUOX 1 and 2, which are silenced in pancreatic ductal adenocarcinoma, but upregulated with P-AscH- treatment. An inducible catalase system, to test causality for the role of hydrogen peroxide, reversed the P-AscH--induced increases in DUOX, whereas DUOX inhibition partially rescued P-AscH--induced toxicity. In addition, DUOX was significantly downregulated in pancreatic cancer specimens compared with normal pancreas tissues. Together, these results suggest that P-AscH--induced toxicity may be enhanced by late metabolic shifts in tumor cells, resulting in a feed-forward mechanism for generation of hydrogen peroxide and induction of metabolic stress through enhanced DUOX expression and rate of oxygen consumption. SIGNIFICANCE: A high dose of vitamin C, in addition to delivering an acute exposure of H2O2 to tumor cells, activates DUOX in pancreatic cancer cells, which provide sustained production of H2O2.


Asunto(s)
Ácido Ascórbico/farmacología , Carcinoma Ductal Pancreático/terapia , Oxidasas Duales/metabolismo , Peróxido de Hidrógeno/metabolismo , Neoplasias Pancreáticas/terapia , Administración Intravenosa , Animales , Ácido Ascórbico/uso terapéutico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Quimioterapia Adyuvante/métodos , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/genética , Oxidasas Duales/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Estrés Oxidativo/efectos de los fármacos , Oxígeno/metabolismo , Consumo de Oxígeno/efectos de los fármacos , Páncreas/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Pancreaticoduodenectomía , Especies Reactivas de Oxígeno/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Cancer Res ; 67(13): 6392-9, 2007 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-17616699

RESUMEN

Thiol antioxidants, including N-acetyl-L-cysteine (NAC), are widely used as modulators of the intracellular redox state. We investigated the hypothesis that NAC-induced reactive oxygen species (ROS) signaling perturbs cellular proliferation by regulating the cell cycle regulatory protein cyclin D1 and the ROS scavenging enzyme Mn-superoxide dismutase (MnSOD). When cultured in media containing NAC, mouse fibroblasts showed G(1) arrest with decreased cyclin D1 protein levels. The absence of a NAC-induced G(1) arrest in fibroblasts overexpressing cyclin D1 (or a nondegradable mutant of cyclin D1-T286A) indicates that cyclin D1 regulates this G(1) arrest. A delayed response to NAC exposure was an increase in both MnSOD protein and activity. NAC-induced G(1) arrest is exacerbated in MnSOD heterozygous fibroblasts. Results from electron spin resonance spectroscopy and flow cytometry measurements of dihydroethidine fluorescence showed an approximately 2-fold to 3-fold increase in the steady-state levels of superoxide (O(2)(*-)) in NAC-treated cells compared with control. Scavenging of O(2)(*-) with Tiron reversed the NAC-induced G(1) arrest. These results show that an O(2)(*-) signaling pathway regulates NAC-induced G(1) arrest by decreasing cyclin D1 protein levels and increasing MnSOD activity.


Asunto(s)
Acetilcisteína/farmacología , Ciclina D1/metabolismo , Superóxido Dismutasa/metabolismo , Superóxidos/metabolismo , Animales , Dicarbetoxidihidrocolidina/análogos & derivados , Dicarbetoxidihidrocolidina/farmacología , Espectroscopía de Resonancia por Spin del Electrón , Fibroblastos/metabolismo , Fase G1 , Humanos , Ratones , Células 3T3 NIH , Oxidación-Reducción , Especies Reactivas de Oxígeno , Transducción de Señal
13.
Free Radic Biol Med ; 45(11): 1501-9, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18790046

RESUMEN

This study investigates the hypothesis that CuZn superoxide dismutase (SOD1) overexpression confers radioresistance to human glioma cells by regulating the late accumulation of reactive oxygen species (ROS) and the G(2)/M-checkpoint pathway. U118-9 human glioma cells (wild type, neo vector control, and stably overexpressing SOD1) were irradiated (0-10 Gy) and assayed for cell survival, cellular ROS levels, cell-cycle-phase distributions, and cyclin B1 expression. SOD1-overexpressing cells were radioresistant compared to wild-type (wt) and neo vector control (neo) cells. Irradiated wt and neo cells showed a significant increase (approximately twofold) in DHE fluorescence beginning at 2 days postirradiation, which remained elevated at 8 days postirradiation. Interestingly, the late accumulation of ROS was suppressed in irradiated SOD1-overexpressing cells. The increase in ROS levels was followed by a decrease in cell growth and viability and an increase in the percentage of cells with sub-G(1) DNA content. SOD1 overexpression enhanced radiation-induced G(2) accumulation within 24 h postirradiation, which was accompanied by a decrease in cyclin B1 mRNA and protein levels. These results support the hypothesis that long after radiation exposure a "metabolic redox response" regulates radiosensitivity of human glioma cells.


Asunto(s)
Glioma/radioterapia , Neuroglía/efectos de la radiación , Tolerancia a Radiación , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo , Western Blotting , Supervivencia Celular/efectos de la radiación , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Ciclina B/genética , Ciclina B/metabolismo , Ciclina B1 , Electroforesis en Gel de Poliacrilamida , Citometría de Flujo , Fase G2/efectos de la radiación , Glioma/metabolismo , Humanos , Neuroglía/citología , Neuroglía/metabolismo , Oxidación-Reducción , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/efectos de la radiación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Superóxido Dismutasa-1
14.
Free Radic Biol Med ; 45(8): 1094-102, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18691649

RESUMEN

Polychlorinated biphenyls (PCBs) are environmental chemical contaminants believed to adversely affect cellular processes. We investigated the hypothesis that PCB-induced changes in the levels of cellular reactive oxygen species (ROS) induce DNA damage resulting in cytotoxicity. Exponentially growing cultures of human nonmalignant breast epithelial cells (MCF10A) were incubated with PCBs for 3 days and assayed for cell number, ROS levels, DNA damage, and cytotoxicity. Exposure to 2,2',4,4',5,5'-hexachlorobiphenyl (PCB153) or 2-(4-chlorophenyl)benzo-1,4-quinone (4-Cl-BQ), a metabolite of 4-chlorobiphenyl (PCB3), significantly decreased cell number and MTS reduction and increased the percentage of cells with sub-G1 DNA content. Results from electron paramagnetic resonance (EPR) spectroscopy showed a 4-fold increase in the steady-state levels of ROS, which was suppressed in cells pretreated with catalase. EPR measurements in cells treated with 4-Cl-BQ detected the presence of a semiquinone radical, suggesting that the increased levels of ROS could be due to the redox cycling of 4-Cl-BQ. A dose-dependent increase in micronuclei frequency was observed in PCB-treated cells, consistent with an increase in histone 2AX phosphorylation. Treatment of cells with catalase blunted the PCB-induced increase in micronuclei frequency and H2AX phosphorylation that was consistent with an increase in cell survival. Our results demonstrate a PCB-induced increase in cellular levels of ROS causing DNA damage, resulting in cell killing.


Asunto(s)
Mama/efectos de los fármacos , Mama/enzimología , Catalasa/metabolismo , Células Epiteliales/efectos de los fármacos , Bifenilos Policlorados/toxicidad , Mama/patología , Células Cultivadas , Espectroscopía de Resonancia por Spin del Electrón , Células Epiteliales/enzimología , Células Epiteliales/patología , Femenino , Histonas/efectos de los fármacos , Histonas/metabolismo , Humanos , Immunoblotting , Micronúcleos con Defecto Cromosómico/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo
15.
Cancer Res ; 66(15): 7615-20, 2006 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-16885361

RESUMEN

Mutations in genes coding for succinate dehydrogenase (SDH) subunits are believed to contribute to cancer and aging, but the mechanism for this is unclear. Hamster fibroblasts expressing a mutation in SDH subunit C (SDHC; B9) showed 3-fold increases in dihydroethidine and dichlorodihydrofluorescein (CDCFH(2)) oxidation indicative of increased steady-state levels of O2(.-) and H2O2, increases in glutathione/glutathione disulfide (indicative of oxidative stress), as well as increases in superoxide dismutase activity, relative to parental B1 cells. B9 cells also showed characteristics associated with cancer cells, including aneuploidy, increases in glucose consumption, and sensitivity to glucose deprivation-induced cytotoxicity. Expression of wild-type (WT) human SDHC in B9 cells caused prooxidant production, glucose consumption, sensitivity to glucose deprivation-induced cytotoxicity, and aneuploidy to revert to the WT phenotype. These data show that SDHC mutations cause increased O2(.-) production, metabolic oxidative stress, and genomic instability and that mutations in genes coding for mitochondrial electron transport chain proteins can contribute to phenotypic changes associated with cancer cells. These results also allow for the speculation that DNA damage to genes coding for electron transport chain proteins could result in a "mutator phenotype" by increasing steady-state levels of O2(.-) and H2O2.


Asunto(s)
Inestabilidad Genómica/fisiología , Proteínas de la Membrana/genética , Superóxidos/metabolismo , Animales , Secuencia de Bases , Línea Celular , Cricetinae , Cricetulus , Fibroblastos/enzimología , Fibroblastos/metabolismo , Humanos , Pulmón/citología , Proteínas de la Membrana/metabolismo , Datos de Secuencia Molecular , Estrés Oxidativo , Transfección
16.
Antioxidants (Basel) ; 6(4)2017 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-29149089

RESUMEN

Communication between the nucleus and mitochondrion could coordinate many cellular processes. While the mechanisms regulating this communication are not completely understood, we hypothesize that cell cycle checkpoint proteins coordinate the cross-talk between nuclear and mitochondrial functions following oxidative stress. Human normal skin fibroblasts, representative of the G2-phase, were irradiated with 6 Gy of ionizing radiation and assayed for cyclin B1 translocation, mitochondrial function, reactive oxygen species (ROS) levels, and cytotoxicity. In un-irradiated controls, cyclin B1 was found primarily in the nucleus of G2-cells. However, following irradiation, cyclin B1 was excluded from the nucleus and translocated to the cytoplasm and mitochondria. These observations were confirmed further by performing transmission electron microscopy and cell fractionation assays. Cyclin B1 was absent in mitochondria isolated from un-irradiated G2-cells and present in irradiated G2-cells. Radiation-induced translocation of cyclin B1 from the nucleus to the mitochondrion preceded changes in the activities of mitochondrial proteins, that included decreases in the activities of aconitase and the mitochondrial antioxidant enzyme, manganese superoxide dismutase (MnSOD), and increases in complex II activity. Changes in the activities of mito-proteins were followed by an increase in dihydroethidium (DHE) oxidation (indicative of increased superoxide levels) and loss of the mitochondrial membrane potential, events that preceded the restart of the stalled cell cycle and subsequently the loss in cell viability. Comparable results were also observed in un-irradiated control cells overexpressing mitochondria-targeted cyclin B1. These results indicate that MnSOD and cyclin B1 coordinate a cross-talk between nuclear and mitochondrial functions, to regulate a mito-checkpoint during the cell cycle response to oxidative stress.

17.
Aging Cell ; 16(5): 1136-1145, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28758339

RESUMEN

Replicative and chronological lifespan are two different modes of cellular aging. Chronological lifespan is defined as the duration during which quiescent normal cells retain their capacity to re-enter the proliferative cycle. This study investigated whether changes in metabolism occur during aging of quiescent normal human fibroblasts (NHFs) and the mechanisms that regulate these changes. Bioenergetics measurements were taken in quiescent NHFs from younger (newborn, 3-day, 5-month, and 1-year) and older (58-, 61-, 63-, 68-, and 70-year) healthy donors as well as NHFs from the same individual at different ages (29, 36, and 46 years). Results show significant changes in cellular metabolism during aging of quiescent NHFs: Old NHFs exhibit a significant decrease in glycolytic flux and lactate levels, and increase in oxygen consumption rate (OCR) and ATP levels compared to young NHFs. Results from the Seahorse XF Cell Mito Stress Test show that old NHFs with a lower Bioenergetic Health Index (BHI) are more prone to oxidative stress compared to young NHFs with a higher BHI. The increase in OCR in old NHFs is associated with a shift in mitochondrial dynamics more toward fusion. Genetic knockdown of mitofusin 1 (MFN1) and optic atrophy 1 (OPA1) in old NHFs decreased OCR and shifted metabolism more toward glycolysis. Downregulation of MFN1 and OPA1 also suppressed the radiation-induced increase in doubling time of NHFs. In summary, results show that a metabolic shift from glycolysis in young to mitochondrial respiration in old NHFs occurs during chronological lifespan, and MFN1 and OPA1 regulate this process.


Asunto(s)
Envejecimiento/genética , GTP Fosfohidrolasas/genética , Glucólisis/genética , Mitocondrias/genética , Proteínas de Transporte de Membrana Mitocondrial/genética , Fosforilación Oxidativa , Adulto , Anciano , Envejecimiento/metabolismo , División Celular , Respiración de la Célula/genética , Células Cultivadas , Fibroblastos/citología , Fibroblastos/metabolismo , GTP Fosfohidrolasas/antagonistas & inhibidores , GTP Fosfohidrolasas/metabolismo , Regulación de la Expresión Génica , Humanos , Lactante , Recién Nacido , Persona de Mediana Edad , Mitocondrias/metabolismo , Dinámicas Mitocondriales , Proteínas de Transporte de Membrana Mitocondrial/antagonistas & inhibidores , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Consumo de Oxígeno , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal
18.
Antioxid Redox Signal ; 8(7-8): 1273-81, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16910775

RESUMEN

This study investigates the hypothesis that Mn-superoxide dismutase (MnSOD) influences cancer cell radiosensitivity by regulating the G(2)-checkpoint pathway. Human oral squamous carcinoma cells (SCC25) stably overexpressing MnSOD were irradiated (6 Gy) and assayed for cell survival, cell-cycle phase distributions, and bromodeoxyuridine (BrdU) pulse-chase flow-cytometric measurements of cell-cycle phase transits. Electron paramagnetic resonance (EPR) spectroscopy was used to measure steady-state levels of oxygen-centered free radicals. Glutathione and glutathione disulfide levels were used as indicators of changes in the intracellular redox state. MnSOD overexpression increased radioresistance threefold to fourfold; this increase was associated with twofold to threefold increases in radiation-induced G(2) accumulation. BrdU pulse-chase and flow-cytometric measurements of the percentage of G(1) and relative movement showed no significant changes in G(1) and S transits; however, the percentage of G(2) cells and BrdU-positive cells showed delayed G(2)+M transits in MnSOD-overexpressing irradiated cells. The steady-state levels of oxygen-centered free radicals were not significantly different in vector compared with MnSOD-overexpressing cells, suggesting that the free radical generation is essentially similar. MnSOD overexpression did prevent radiation-induced decreases in total glutathione content, which correlated with radioresistance and enhanced G(2) accumulation. These results support the hypothesis that a "metabolic redox-response" to IR exposure regulates radiosensitivity by altering radiation-induced G(2) accumulation.


Asunto(s)
Carcinoma de Células Escamosas , Fase G2 , Neoplasias de la Boca , Tolerancia a Radiación , Superóxido Dismutasa/biosíntesis , Bromodesoxiuridina/metabolismo , Carcinoma de Células Escamosas/enzimología , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/radioterapia , Ciclo Celular , Supervivencia Celular/efectos de la radiación , Espectroscopía de Resonancia por Spin del Electrón , Citometría de Flujo , Radicales Libres/análisis , Radicales Libres/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de la radiación , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Glutatión/análisis , Glutatión/metabolismo , Disulfuro de Glutatión/análisis , Disulfuro de Glutatión/metabolismo , Humanos , Neoplasias de la Boca/enzimología , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , Neoplasias de la Boca/radioterapia , Oxidación-Reducción , Superóxido Dismutasa/genética , Factores de Tiempo
19.
Cancer Res ; 75(16): 3314-26, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26081808

RESUMEN

The toxicity of pharmacologic ascorbate is mediated by the generation of H2O2 via the oxidation of ascorbate. Because pancreatic cancer cells are sensitive to H2O2 generated by ascorbate, they would also be expected to become sensitized to agents that increase oxidative damage such as ionizing radiation. The current study demonstrates that pharmacologic ascorbate enhances the cytotoxic effects of ionizing radiation as seen by decreased cell viability and clonogenic survival in all pancreatic cancer cell lines examined, but not in nontumorigenic pancreatic ductal epithelial cells. Ascorbate radiosensitization was associated with an increase in oxidative stress-induced DNA damage, which was reversed by catalase. In mice with established heterotopic and orthotopic pancreatic tumor xenografts, pharmacologic ascorbate combined with ionizing radiation decreased tumor growth and increased survival, without damaging the gastrointestinal tract or increasing systemic changes in parameters indicative of oxidative stress. Our results demonstrate the potential clinical utility of pharmacologic ascorbate as a radiosensitizer in the treatment of pancreatic cancer.


Asunto(s)
Ácido Ascórbico/farmacología , Neoplasias Pancreáticas/terapia , Fármacos Sensibilizantes a Radiaciones/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Antioxidantes/farmacología , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Supervivencia Celular/efectos de la radiación , Quimioradioterapia , Daño del ADN , Relación Dosis-Respuesta en la Radiación , Glutatión/metabolismo , Disulfuro de Glutatión/metabolismo , Humanos , Peróxido de Hidrógeno/metabolismo , Estimación de Kaplan-Meier , Modelos Lineales , Ratones Desnudos , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/efectos de la radiación , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Radiación Ionizante , Carga Tumoral/efectos de los fármacos , Carga Tumoral/genética , Carga Tumoral/efectos de la radiación
20.
Antioxid Redox Signal ; 20(10): 1618-27, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23590434

RESUMEN

SIGNIFICANCE: Manganese superoxide dismutase (MnSOD) is a nuclear-encoded and mitochondria-matrix-localized oxidation-reduction (redox) enzyme that regulates cellular redox homeostasis. Cellular redox processes are known to regulate proliferative and quiescent growth states. Therefore, MnSOD and mitochondria-generated reactive oxygen species (ROS) are believed to be critical regulators of quiescent cells' entry into the cell cycle and exit from the proliferative cycle back to the quiescent state. RECENT ADVANCES/CRITICAL ISSUES: Recent evidence suggests that the intracellular redox environment fluctuates during the cell cycle, shifting toward a more oxidized status during mitosis. MnSOD activity is higher in G0/G1 cells compared with S, G2 and M phases. After cell division, MnSOD activity increases in the G1 phase of the daughter generation. The periodic fluctuation in MnSOD activity during the cell cycle inversely correlates with cellular superoxide levels as well as glucose and oxygen consumption. Based on an inverse correlation between MnSOD activity and glucose consumption during the cell cycle, it is proposed that MnSOD is a central molecular player for the "Warburg effect." FUTURE DIRECTIONS: In general, loss of MnSOD activity results in aberrant proliferation. A better understanding of the MnSOD and mitochondrial ROS-dependent cell cycle processes may lead to novel approaches to overcome aberrant proliferation. Since ROS have both deleterious (pathological) and beneficial (physiological) effects, it is proposed that "eustress" should be used when discussing ROS processes that regulate normal physiological functions, while "oxidative stress" should be used to discuss the deleterious effects of ROS.


Asunto(s)
Ciclo Celular , Superóxido Dismutasa/fisiología , Animales , Antioxidantes/metabolismo , Proteínas de Ciclo Celular/metabolismo , Metabolismo Energético , Humanos , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA