Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
ChemMedChem ; : e202400081, 2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-38976686

RESUMEN

A series of 7-substituted coumarin derivatives have been characterized as pan-aldo-keto reductase family 1C (AKR1C) inhibitors. The AKR1C family of enzymes are overexpressed in numerous cancers where they are involved in drug resistance development. 7-hydroxy coumarin ethyl esters and their corresponding amides have high potency for AKR1C3 and AKR1C2 inhibition. Coumarin amide 3a possessed IC50 values of 50 nM and 90 nM for AKR1C3 and AKR1C2, respectively, and exhibits 'drug-like' metabolic stability and half-life in human and mouse liver microsomes and plasma. Compound 3a was employed as a chemical tool to determine pan-AKR1C2/3 inhibition effects both as a radiation sensitizer and as a potentiator of chemotherapy cytotoxicity. In contrast to previously reported pan-AKR1C inhibitors, 3a demonstrated no radiation sensitization effect in a radiation-resistant prostate cancer cell line model. Pan-AKR1C inhibition also did not potentiate the in vitro cytotoxicity of ABT-737, daunorubicin or dexamethasone, in two patient-derived T-cell ALL and pre-B-cell ALL cell lines.  In contrast, a highly selective AKR1C3 inhibitor, compound K90, enhanced the cytotoxicity of both ABT-737 and daunorubicin in the T-cell ALL cell line model. Thus, the inhibitory profile of the AKR1C family inhibitor required to effect enhancement of chemotherapeutic cytotoxicity may be chemotherapeutic agent-specific in leukemia.

2.
Redox Biol ; 73: 103219, 2024 07.
Artículo en Inglés | MEDLINE | ID: mdl-38851001

RESUMEN

Radiation causes damage to normal tissues that leads to increased oxidative stress, inflammation, and fibrosis, highlighting the need for the selective radioprotection of healthy tissues without hindering radiotherapy effectiveness in cancer. This study shows that adiponectin, an adipokine secreted by adipocytes, protects normal tissues from radiation damage invitro and invivo. Specifically, adiponectin (APN) reduces chronic oxidative stress and fibrosis in irradiated mice. Importantly, APN also conferred no protection from radiation to prostate cancer cells. Adipose tissue is the primary source of circulating endogenous adiponectin. However, this study shows that adipose tissue is sensitive to radiation exposure exhibiting morphological changes and persistent oxidative damage. In addition, radiation results in a significant and chronic reduction in blood APN levels from adipose tissue in mice and human prostate cancer patients exposed to pelvic irradiation. APN levels negatively correlated with bowel toxicity and overall toxicities associated with radiotherapy in prostate cancer patients. Thus, protecting, or modulating APN signaling may improve outcomes for prostate cancer patients undergoing radiotherapy.


Asunto(s)
Adiponectina , Fibrosis , Estrés Oxidativo , Neoplasias de la Próstata , Masculino , Animales , Neoplasias de la Próstata/radioterapia , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Humanos , Ratones , Estrés Oxidativo/efectos de la radiación , Adiponectina/metabolismo , Adiponectina/sangre , Traumatismos por Radiación/metabolismo , Traumatismos por Radiación/patología , Tejido Adiposo/metabolismo , Tejido Adiposo/efectos de la radiación , Protectores contra Radiación/farmacología , Protectores contra Radiación/uso terapéutico
3.
Radiat Res ; 199(1): 89-111, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36368026

RESUMEN

Increasing utilization of nuclear power enhances the risks associated with industrial accidents, occupational hazards, and the threat of nuclear terrorism. Exposure to ionizing radiation interferes with genomic stability and gene expression resulting in the disruption of normal metabolic processes in cells and organs by inducing complex biological responses. Exposure to high-dose radiation causes acute radiation syndrome, which leads to hematopoietic, gastrointestinal, cerebrovascular, and many other organ-specific injuries. Altered genomic variations, gene expression, metabolite concentrations, and microbiota profiles in blood plasma or tissue samples reflect the whole-body radiation injuries. Hence, multi-omic profiles obtained from high-resolution omics platforms offer a holistic approach for identifying reliable biomarkers to predict the radiation injury of organs and tissues resulting from radiation exposures. In this review, we performed a literature search to systematically catalog the radiation-induced alterations from multi-omic studies and radiation countermeasures. We covered radiation-induced changes in the genomic, transcriptomic, proteomic, metabolomic, lipidomic, and microbiome profiles. Furthermore, we have covered promising multi-omic biomarkers, FDA-approved countermeasure drugs, and other radiation countermeasures that include radioprotectors and radiomitigators. This review presents an overview of radiation-induced alterations of multi-omics profiles and biomarkers, and associated radiation countermeasures.


Asunto(s)
Síndrome de Radiación Aguda , Protectores contra Radiación , Humanos , Protectores contra Radiación/farmacología , Multiómica , Proteómica , Síndrome de Radiación Aguda/diagnóstico , Síndrome de Radiación Aguda/etiología , Biomarcadores
4.
PLoS One ; 17(11): e0277239, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36331948

RESUMEN

Prostate cancer is one of the most diagnosed cancers in men in the United States. In mouse models, orthotopic tumors are favored for their biological relevance and simulation of growth in a microenvironment akin to that found in humans. However, to monitor the disease course, animal models require consistent and noninvasive surveillance. In vivo bioluminescent imaging has become a mainstay imaging modality due to its flexibility and ease of use. However, with some orthotopic prostate tumor models, bioluminescence fails to describe disease progression due to optical scattering and signal attenuation. CT scanning, in addition to its utility in human cancer diagnosis and surveillance, can be applied to mouse models with improved results. However, CT imaging has poor definition when imaging soft tissues and is not routinely used in prostate cancer models. Using an orthotopic prostate cancer model, our results demonstrate that, when compared to bioluminescent imaging, CT imaging correlates more closely to orthotopic prostate tumor growth in mice. Based on the data from this study, we conclude that CT imaging can be used as an alternative to the more commonly used bioluminescent imaging for measuring orthotopic prostate cancer growth over time.


Asunto(s)
Imagen Óptica , Neoplasias de la Próstata , Tomografía Computarizada por Rayos X , Animales , Humanos , Masculino , Ratones , Línea Celular Tumoral , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Próstata/diagnóstico por imagen , Próstata/patología , Neoplasias de la Próstata/diagnóstico por imagen , Neoplasias de la Próstata/patología , Tomografía Computarizada por Rayos X/métodos , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto , Imagen Óptica/métodos , Mediciones Luminiscentes/métodos
5.
Cell Mol Bioeng ; 15(5): 391-407, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36444349

RESUMEN

Introduction: Oxidative stress due to excess reactive oxygen species (ROS) is related to many chronic illnesses including degenerative disc disease and osteoarthritis. MnTnBuOE-2-PyP5+ (BuOE), a manganese porphyrin analog, is a synthetic superoxide dismutase mimetic that scavenges ROS and has established good treatment efficacy at preventing radiation-induced oxidative damage in healthy cells. BuOE has not been studied in degenerative disc disease applications and only few studies have loaded BuOE into drug delivery systems. The goal of this work is to engineer BuOE microparticles (MPs) as an injectable therapeutic for long-term ROS scavenging. Methods: Methacrylated chondroitin sulfate-A MPs (vehicle) and BuOE MPs were synthesized via water-in-oil polymerization and the size, surface morphology, encapsulation efficiency and release profile were characterized. To assess long term ROS scavenging of BuOE MPs, superoxide scavenging activity was evaluated over an 84-day time course. In vitro cytocompatibility and cellular uptake were assessed on human intervertebral disc cells. Results: BuOE MPs were successfully encapsulated in MACS-A MPs and exhibited a slow-release profile over 84 days. BuOE maintained high potency in superoxide scavenging after encapsulation and after 84 days of incubation at 37 °C as compared to naked BuOE. Vehicle and BuOE MPs (100 µg/mL) were non-cytotoxic on nucleus pulposus cells and MPs up to 23 µm were endocytosed. Conclusions: BuOE MPs can be successfully fabricated and maintain potent superoxide scavenging capabilities up to 84-days. In vitro assessment reveals the vehicle and BuOE MPs are not cytotoxic and can be taken up by cells. Supplementary Information: The online version contains supplementary material available at 10.1007/s12195-022-00744-w.

6.
Front Physiol ; 13: 989793, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36213236

RESUMEN

Metabolic alterations are a common survival mechanism for prostate cancer progression and therapy resistance. Oxidative stress in the cellular and tumor microenvironment dictates metabolic switching in the cancer cells to adopt, prosper and escape therapeutic stress. Therefore, regulation of oxidative stress in tumor cells and in the tumor-microenvironment may enhance the action of conventional anticancer therapies. NRF2 is the master regulator for oxidative stress management. However, the overall oxidative stress varies with PCa clinical stage, metabolic state and therapy used for the cancer. In agreement, the blanket use of NRF2 inducers or inhibitors along with anticancer therapies cause adverse effects in some preclinical cancer models. In this review, we have summarized the levels of oxidative stress, metabolic preferences and NRF2 activity in the different stages of prostate cancer. We also propose condition specific ways to use NRF2 inducers or inhibitors along with conventional prostate cancer therapies. The significance of this review is not only to provide a detailed understanding of the mechanism of action of NRF2 to regulate oxidative stress-mediated metabolic switching by prostate cancer cells to escape the radiation, chemo, or hormonal therapies, and to grow aggressively, but also to provide a potential therapeutic method to control aggressive prostate cancer growth by stage specific proper use of NRF2 regulators.

7.
Free Radic Biol Med ; 193(Pt 1): 274-291, 2022 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-36265795

RESUMEN

The castration-resistant (CR) prostate cancer (PCa) is lethal and is the second leading cause of cancer-related deaths in U.S. males. To develop effective treatments toward CR PCa, we investigated reactive oxygen species (ROS) signaling pathway for its role involving in CR PCa progression. ROS can regulate both cell growth and apoptosis: a moderate increase of ROS promotes proliferation; its substantial rise results in cell death. p66Shc protein can increase oxidant species production and its elevated level is associated with the androgen-independent (AI) phenotype of CR PCa cells; while heme oxygenase-1 (HO-1) is an antioxidant enzyme and elevated in a sub-group of metastatic PCa cells. In this study, our data revealed that HO-1 and p66Shc protein levels are co-elevated in various AI PCa cell lines as well as p66Shc cDNA-transfected cells. Knockdown and/or inhibition of either p66Shc or HO-1 protein leads to reduced tumorigenicity as well as a reduction of counterpart protein. Knockdown of HO-1 alone results in increased ROS levels, nucleotide and protein oxidation and induction of cell death. Together, our data indicate that elevated HO-1 protein levels protect PCa cells from otherwise apoptotic conditions induced by aberrant p66Shc/ROS production, which thereby promotes PCa progression to the CR phenotype. p66Shc and HO-1 can serve as functional targets for treating CR PCa.


Asunto(s)
Hemo-Oxigenasa 1 , Neoplasias de la Próstata , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Humanos , Masculino , Antioxidantes/metabolismo , Hemo-Oxigenasa 1/metabolismo , Neoplasias de la Próstata/patología , Especies Reactivas de Oxígeno/metabolismo , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/genética , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/metabolismo
8.
Breast Cancer Res ; 24(1): 48, 2022 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-35836253

RESUMEN

BACKGROUND: A pro-oxidant enzyme, NADPH oxidase 4 (Nox4) has been reported to be a critical downstream effector of TGFß-induced myofibroblast transformation during fibrosis. While there are a small number of studies suggesting an oncogenic role of Nox4 derived from activated fibroblasts, direct evidence linking this pro-oxidant to the tumor-supporting CAF phenotype and the mechanisms involved are lacking, particularly in breast cancer. METHODS: We targeted Nox4 in breast patient-derived CAFs via siRNA-mediated knockdown or administration of a pharmaceutical inhibitor (GKT137831). We also determine primary tumor growth and metastasis of implanted tumor cells using a stable Nox4-/- syngeneic mouse model. Autophagic flux of CAFs was assessed using a tandem fluorescent-tagged ptfl-LC3 plasmid via confocal microscopy analysis and determination of the expression level of autophagy markers (beclin-1 and LC3B). Nox4 overexpressing CAFs depend on the Nrf2 (nuclear factor-erythroid factor 2-related factor 2) pathway for survival. We then determined the dependency of Nox4-overexpressing CAFs on the Nrf2-mediated adaptive stress response pathway for survival. Furthermore, we investigated the involvement of Birc5 on CAF phenotype (viability and collagen contraction activity) as well as the expression level of CAF markers, FAP and αSMA. CONCLUSIONS: We found that deletion of stroma Nox4 and pharmaceutically targeting its activity with GKT137831 significantly inhibited orthotopic tumor growth and metastasis of implanted E0771 and 4T1 murine mammary carcinoma cell lines in mice. More importantly, we found a significant upregulation of Nox4 expression in CAFs isolated from human breast tumors versus normal mammary fibroblasts (RMFs). Our in situ RNA hybridization analysis for Nox4 transcription on a human breast tumor microarray further support a role of this pro-oxidant in the stroma of breast carcinomas. In addition, we found that Nox4 promotes autophagy in CAFs. Moreover, we found that Nox4 promoted survival of CAFs via activation of Nrf2, a master regulator of oxidative stress response. We have further shown Birc5 is involved as a downstream modulator of Nrf2-mediated pro-survival phenotype. Together these studies indicate a role of redox signaling via the Nox4-Nrf2 pathway in tumorigenesis and metastasis of breast cancer cells by promoting autophagy and survival of CAFs.


Asunto(s)
Neoplasias de la Mama , Fibroblastos Asociados al Cáncer , Animales , Neoplasias de la Mama/patología , Fibroblastos Asociados al Cáncer/patología , Carcinogénesis/metabolismo , Línea Celular Tumoral , Femenino , Fibroblastos/metabolismo , Humanos , Ratones , NADPH Oxidasa 4/genética , NADPH Oxidasa 4/metabolismo , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Survivin/metabolismo , Regulación hacia Arriba
9.
Clin Exp Metastasis ; 39(4): 641-659, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35604506

RESUMEN

Bone metastatic prostate cancer (BM-PCa) remains one of the most difficult cancers to treat due to the complex interactions of cancer and stromal cells. We previously showed that bone marrow neutrophils elicit an anti-tumor immune response against BM-PCa. Further, we demonstrated that BM-PCa induces neutrophil oxidative burst, which has previously been identified to promote primary tumor growth of other cancers, and a goal of this study was to define the importance of neutrophil oxidative burst in BM-PCa. To do this, we first examined the impact of depletion of reactive oxygen species (ROS), via systemic deletion of the main source of ROS in phagocytes, NADPH oxidase (Nox)2, which we found to suppress prostate tumor growth in bone. Further, using pharmacologic ROS inhibitors and Nox2-null neutrophils, we found that ROS depletion specifically suppresses growth of androgen-insensitive prostate cancer cells. Upon closer examination using bulk RNA sequencing analysis, we identified that metastatic prostate cancer induces neutrophil transcriptomic changes that activates pathways associated with response to oxidative stress. In tandem, prostate cancer cells resist neutrophil anti-tumor response via extracellular (i.e., regulation of neutrophils) and intracellular alterations of glutathione synthesis, the most potent cellular antioxidant. These findings demonstrate that BM-PCa thrive under oxidative stress conditions and such that regulation of ROS and glutathione programming could be leveraged for targeting of BM-PCa progression.


Asunto(s)
Neoplasias Óseas , Neoplasias de la Próstata , Neoplasias Óseas/secundario , Glutatión/metabolismo , Humanos , Masculino , Neutrófilos/patología , Estrés Oxidativo , Neoplasias de la Próstata/patología , Especies Reactivas de Oxígeno/metabolismo
10.
Redox Biol ; 52: 102301, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35358851

RESUMEN

Radiation is a common anticancer therapy for prostate cancer, which transforms tumor-associated normal fibroblasts to myofibroblasts, resulting in fibrosis. Oxidative stress caused by radiation-mediated mitochondrial damage is one of the major contributors to fibrosis. As diabetics are oxidatively stressed, radiation-mediated reactive oxygen species cause severe treatment failure, treatment-related side effects, and significantly reduced survival for diabetic prostate cancer patients as compared to non-diabetic prostate cancer patients. Hyperglycemia and enhanced mitochondrial damage significantly contribute to oxidative damage and disease progression after radiation therapy among diabetic prostate cancer patients. Therefore, reduction of mitochondrial damage in normal prostate fibroblasts after radiation should improve the overall clinical state of diabetic prostate cancer patients. We previously reported that MnTE-2-PyP, a manganese porphyrin, reduces oxidative damage in irradiated hyperglycemic prostate fibroblasts by scavenging superoxide and activating NRF2. In the current study, we have investigated the potential role of MnTE-2-PyP to protect mitochondrial health in irradiated hyperglycemic prostate fibroblasts. This study revealed that hyperglycemia and radiation increased mitochondrial ROS via blocking the mitochondrial electron transport chain, altered mitochondrial dynamics, and reduced mitochondrial biogenesis. Increased mitochondrial damage preceeded an increase in myofibroblast differentiation. MnTE-2-PyP reduced myofibroblast differentiation, improved mitochondrial health by releasing the block on the mitochondrial electron transport chain, enhanced ATP production efficiency, and restored mitochondrial dynamics and metabolism in the irradiated-hyperglycemic prostate fibroblasts. Therefore, we are proposing that one of the mechanisms that MnTE-2-PyP protects prostate fibroblasts from irradiation and hyperglycemia-mediated damage is by protecting the mitochondrial health in diabetic prostate cancer patients.


Asunto(s)
Diabetes Mellitus , Hiperglucemia , Metaloporfirinas , Neoplasias de la Próstata , Exposición a la Radiación , Diabetes Mellitus/metabolismo , Fibroblastos/metabolismo , Fibrosis , Humanos , Hiperglucemia/metabolismo , Masculino , Mitocondrias/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/radioterapia
11.
J Orthop Res ; 40(11): 2586-2596, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35148568

RESUMEN

As cancer survivorship increases, so does the number of patients that suffer from the late effects of radiation therapy. This includes arthrofibrosis, the development of stiff joints near the field of radiation. Previous reports have concentrated on skin fibrosis around the joint but largely ignored the deeper tissues of the joint. We hypothesized that fat, muscle, and the joint tissues themselves would play a more significant role in joint contracture after radiation than the skin surrounding the joint. To address this hypothesis, we irradiated the right hind flanks of mice with fractionated and unfractionated dose schedules, then monitored the mice for 3 months postradiotherapy. Mice were euthanized and physiological indications of arthrofibrosis including limb contracture and joint resting position were assessed. Stifle (knee) joints demonstrated significant arthrofibrosis, but none was observed in the hock (ankle) joints. During these studies, we were surprised to find that male and female mice showed a significantly different response to radiation injury. Female mice developed more injuries, had significantly worse contracture, and showed a greater difference in the expression of all markers studied. These results suggest that women undergoing radiation therapy might be at significantly greater risk for developing arthrofibrosis and may require specific adjustments to their care.


Asunto(s)
Contractura , Artropatías , Animales , Articulación del Tobillo , Contractura/etiología , Contractura/patología , Femenino , Fibrosis , Artropatías/tratamiento farmacológico , Articulación de la Rodilla/patología , Masculino , Ratones
12.
Acta Biomater ; 125: 126-137, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33549808

RESUMEN

Mechanical properties of vascular grafts likely play important roles in healing and tissue regeneration. Healthy arteries are compliant at low pressures but stiffen rapidly with increasing load, ensuring sufficient volumetric expansion without overstretching the vessel. Commercial synthetic vascular grafts are stiff and unable to expand under physiologic loads, which may result in altered hemodynamics, deleterious cellular responses, and compromised clinical performance. The goal of this study was to develop an Elastomeric Nanofibrillar Graft (ENG) with artery-tuned nonlinear compliance and compare its healing responses to conventional expanded polytetrafluoroethylene (ePTFE) grafts in a porcine iliac artery model. Human and porcine iliac arteries were mechanically characterized, and an ENG with similar properties was created by utilizing residual strains within electrospun nanofibers. The ENG was tested for implantation suitability and implanted onto n = 5 domestic swine iliac arteries, with control ePTFE grafts implanted onto the contralateral iliac arteries. After two weeks in vivo, all iliac arteries and grafts remained patent with no signs of thrombosis or dilation. The mechanically tuned ENG implants exhibited a more confluent CD31-positive cell monolayer (1.53 ± 0.73 µm2/mm vs 0.52 ± 0.55 µm2/mm, p = 0.042) on the graft lumenal surface and a higher fraction of αSMA-positive cells (16.2 ± 8.6% vs 1.4 ± 0.7%, p = 0.018) within the graft wall than the ePTFE controls. Despite heavy cellular infiltration, the ENG retained its artery-like mechanical characteristics after two weeks in vivo. These short-term results demonstrate potential advantages of mechanically tuned biomimetic vascular grafts over standard ePTFE grafts. STATEMENT OF SIGNIFICANCE: Off-the-shelf synthetic vascular grafts are often the only option available for treating advanced stages of vascular disease. Despite significant efforts devoted to improving their biochemical characteristics, synthetic peripheral arterial grafts continue to demonstrate poor clinical outcomes leading to costly reinterventions. Here, we hypothesized that a synthetic vascular graft with elastomeric mechanical properties tuned to a healthy peripheral artery promotes better healing responses than a synthetic stiff graft. To test this hypothesis, we developed an Elastomeric Nanofibrillar Graft (ENG) with artery-tuned mechanical properties and compared its performance to a commercial ePTFE graft in a preclinical porcine iliac artery model. Our results suggest that mechanically tuned ENGs can offer better healing responses, potentially leading to better clinical outcomes for peripheral arterial repairs.


Asunto(s)
Implantación de Prótesis Vascular , Nanofibras , Animales , Prótesis Vascular , Arteria Ilíaca/cirugía , Politetrafluoroetileno/farmacología , Porcinos
13.
Radiat Res ; 195(2): 128-139, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33264413

RESUMEN

Novel synthetic compounds, known as manganese porphyrins (MnPs), have been designed to shift the redox status of both normal cells and cancer cells. When MnPs are coupled with cancer therapies, such as radiation, they have been shown to sensitize tumor cells to treatment and protect normal tissues from damage through the modulation of the redox status of various tissue types. Until now, our preclinical studies have focused on local effects of MnPs and radiation; however, we recognize that successful outcomes for cancer patients involve control of tumor cells throughout the body. In this study, using murine orthotopic mammary tumor models, we investigated how MnPs and radiation influence the development of distant metastasis. We hypothesized that the combination of MnP (MnP/RT), such as MnTnBuOE-2-PyP5+ and radiation treatment (RT) would increase local tumor control via a shift in the intratumoral redox environment, leading to subsequent downregulation of HIF-1 in the primary tumor. Secondarily, we hypothesized that these primary tumor treatment effects would result in a reduction in pulmonary metastatic burden. Balb/c mice with orthotopic 4T1 mammary carcinomas were treated with saline, MnP, RT or MnP/RT. We found MnP/RT did extend local tumor growth delay and overall survival compared to controls and was associated with increased intratumoral oxidative stress. However, the primary tumor growth delay observed with MnP/RT was not associated with a reduced pulmonary metastatic burden. Future directions to investigate the effects of MnP/RT on the development of distant metastasis may include modifications to the radiation dose, the experimental timeline or using a murine mammary carcinoma cell line with a less aggressive metastatic behavior. Clinical trials are underway to investigate the clinical utility of MnTnBuOE-2-PyP5+ for patients undergoing radiotherapy for various tumor types. The promising preclinical data from this study, as well as others, provides support that MnP/RT has the potential to improve local tumor control for these patients.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/radioterapia , Carcinoma/radioterapia , Metaloporfirinas/farmacología , Tolerancia a Radiación/efectos de los fármacos , Animales , Neoplasias de la Mama/patología , Carcinoma/tratamiento farmacológico , Carcinoma/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Terapia Combinada , Femenino , Humanos , Manganeso/farmacología , Ratones , Ratones Endogámicos BALB C , Oxidación-Reducción/efectos de los fármacos , Oxidación-Reducción/efectos de la radiación , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/efectos de la radiación , Porfirinas/farmacología
14.
Adv Redox Res ; 32021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38831957

RESUMEN

The pathogenesis of hypertension has been linked to excessive levels of reactive oxygen species (ROS), particularly superoxide (O2•-), in multiple tissues and organ systems. Overexpression of superoxide dismutase (SOD) to scavenge O2•- has been shown to decrease blood pressure in hypertensive animals. We have previously shown that MnTnBuOE-2-PyP5+ (BuOE), a manganese porphyrin SOD mimic currently in clinical trials as a normal tissue protector for cancer patients undergoing radiation therapy, can scavenge O2•- and acutely decrease normotensive blood pressures. Herein, we hypothesized that BuOE decreases hypertensive blood pressures. Using angiotensin II (AngII)-hypertensive mice, we demonstrate that BuOE administered both intraperitoneally and intravenously (IV) acutely decreases elevated blood pressure. Further investigation using renal sympathetic nerve recordings in spontaneously hypertensive rats (SHRs) reveals that immediately following IV injection of BuOE, blood pressure and renal sympathetic nerve activity (RSNA) decrease. BuOE also induces dose-dependent vasodilation of femoral arteries from AngII-hypertensive mice, a response that is mediated, at least in part, by nitric oxide, as demonstrated by ex vivo video myography. We confirmed this vasodilation in vivo using doppler imaging of the superior mesenteric artery in AngII-hypertensive mice. Together, these data demonstrate that BuOE acutely decreases RSNA and induces vasodilation, which likely contribute to its ability to rapidly decrease hypertensive blood pressure.

15.
Redox Biol ; 36: 101610, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32863236

RESUMEN

Scavenging superoxide (O2•-) via overexpression of superoxide dismutase (SOD) or administration of SOD mimics improves outcomes in multiple experimental models of human disease including cardiovascular disease, neurodegeneration, and cancer. While few SOD mimics have transitioned to clinical trials, MnTnBuOE-2-PyP5+ (BuOE), a manganese porphyrin SOD mimic, is currently in clinical trials as a radioprotector for cancer patients; thus, providing hope for the use of SOD mimics in the clinical setting. However, BuOE transiently alters cardiovascular function including a significant and precipitous decrease in blood pressure. To limit BuOE's acute hypotensive action, we developed a mesoporous silica nanoparticle and lipid bilayer nanoformulation of BuOE (nanoBuOE) that allows for slow and sustained release of the drug. Herein, we tested the hypothesis that unlike native BuOE, nanoBuOE does not induce an acute hypotensive response, as the nanoformulation prevents BuOE from scavenging O2•- while the drug is still encapsulated in the formulation. We report that intact nanoBuOE does not effectively scavenge O2•-, whereas BuOE released from the nanoformulation does retain SOD-like activity. Further, in mice, native BuOE, but not nanoBuOE, rapidly, acutely, and significantly decreases blood pressure, as measured by radiotelemetry. To begin exploring the physiological mechanism by which native BuOE acutely decreases blood pressure, we recorded renal sympathetic nerve activity (RSNA) in rats. RSNA significantly decreased immediately following intravenous injection of BuOE, but not nanoBuOE. These data indicate that nanoformulation of BuOE, a SOD mimic currently in clinical trials in cancer patients, prevents BuOE's negative side effects on blood pressure homeostasis.


Asunto(s)
Metaloporfirinas , Preparaciones Farmacéuticas , Porfirinas , Animales , Humanos , Ratones , Ratas , Superóxido Dismutasa
16.
Sci Rep ; 10(1): 12616, 2020 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-32724116

RESUMEN

Prostate and colon cancers are among the most common cancers diagnosed annually, and both often require treatment with radiation therapy. Advancement in radiation delivery techniques has led to highly accurate targeting of tumor and sparing of normal tissue; however, in the pelvic region it is anatomically difficult to avoid off-target radiation exposure to other organs. Chronically the effects of normal urogenital tissue exposure can lead to urinary frequency, urinary incontinence, proctitis, and erectile dysfunction. Most of these symptoms are caused by radiation-induced fibrosis and reduce the quality of life for cancer survivors. We have observed in animal models that the severity of radiation-induced fibrosis in normal tissue correlates to damaged fat reservoirs in the pelvic region. We hypothesize that adipocytes may secrete a factor that prevents the induction of radiation-associated fibrosis in normal tissues. In these studies we show that the adipokine, adiponectin, is secreted by primary mouse adipocytes and protects fibroblasts from radiation-induced cell death, myofibroblast formation, and senescence. Further, we demonstrated that adiponectin does not protect colorectal or prostate cancer cells from radiation-induced death. Thus, we propose that adiponectin, or its downstream pathway, would provide a novel target for adjuvant therapy when treating pelvic cancers with radiation therapy.


Asunto(s)
Adipocitos/patología , Adiponectina/metabolismo , Citoprotección , Fibroblastos/patología , Adipocitos/efectos de los fármacos , Adipocitos/efectos de la radiación , Animales , Muerte Celular/efectos de los fármacos , Muerte Celular/efectos de la radiación , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Senescencia Celular/efectos de los fármacos , Técnicas de Cocultivo , Medios de Cultivo Condicionados/farmacología , Citoprotección/efectos de los fármacos , Citoprotección/efectos de la radiación , Fibroblastos/efectos de los fármacos , Fibroblastos/efectos de la radiación , Humanos , Masculino , Ratones Endogámicos C57BL , Proteínas Recombinantes/farmacología , Rayos X
17.
Antioxidants (Basel) ; 9(6)2020 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-32512786

RESUMEN

Prostate cancer patients are often treated with radiotherapy. MnTE-2-PyP, a superoxide dismutase (SOD) mimic, is a known radioprotector of normal tissues. Our recent work demonstrated that MnTE-2-PyP also inhibits prostate cancer progression with radiotherapy; however, the mechanisms remain unclear. In this study, we identified that MnTE-2-PyP-induced intracellular H2O2 levels are critical in inhibiting the growth of PC3 and LNCaP cells, but the increased H2O2 levels affected the two cancer cells differently. In PC3 cells, many proteins were thiol oxidized with MnTE-2-PyP treatment, including Ser/Thr protein phosphatase 1 beta catalytic subunit (PP1CB). This resulted in reduced PP1CB activity; however, overall cell cycle progression was not altered, so this is not the main mechanism of PC3 cell growth inhibition. High H2O2 levels by MnTE-2-PyP treatment induced nuclear fragmentation, which could be synergistically enhanced with radiotherapy. In LNCaP cells, thiol oxidation by MnTE-2-PyP treatment was not observed previously and, similarly to PC3 cells, there was no effect of MnTE-2-PyP treatment on cell cycle progression. However, in LNCaP cells, MnTE-2-PyP caused an increase in low RNA population and sub-G1 population of cells, which indicates that MnTE-2-PyP treatment may cause cellular quiescence or direct cancer cell death. The protein oxidative modifications and mitotic catastrophes caused by MnTE-2-PyP may be the major contributors to cell growth inhibition in PC3 cells, while in LNCaP cells, tumor cell quiescence or cell death appears to be major factors in MnTE-2-PyP-induced growth inhibition.

18.
Redox Biol ; 34: 101542, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32361681

RESUMEN

Radiation is a common anticancer therapy for many cancer patients, including prostate cancer. Diabetic prostate cancer patients suffer from increased lymph node metastasis, tumor recurrence and decreased survival as compared to non-diabetic prostate cancer patients. These patients are also at increased risk for enhanced radiation-induced normal tissue damage such as proctitis. Diabetics are oxidatively stressed and radiation causes additional oxidative damage. We and others have reported that, MnTE-2-PyP, a manganese porphyrin, protects normal prostate tissue from radiation damage. We have also reported that, in an in vivo mouse model of prostate cancer, MnTE-2-PyP decreases tumor volume and increases survival of the mice. In addition, MnTE-2-PyP has also been shown to reduce blood glucose and inhibits pro-fibrotic signaling in a diabetic model. Therefore, to investigate the role of MnTE-2-PyP in normal tissue protection in an irradiated diabetic environment, we have treated human prostate fibroblast cells with MnTE-2-PyP in an irradiated hyperglycemic environment. This study revealed that hyperglycemia causes increased cell death after radiation as compared to normo-glycemia. MnTE-2-PyP protects against hyperglycemia-induced cell death after radiation. MnTE-2-PyP decreases expression of NOX4 and α-SMA, one of the major oxidative enzymes and pro-fibrotic molecules respectively. MnTE-2-PyP obstructs NF-κB activity by decreasing DNA binding of the p50-p50 homodimer in the irradiated hyperglycemic environment. MnTE-2-PyP increases NRF2 mediated cytoprotection by increasing NRF2 protein expression and DNA binding. Therefore, we are proposing that, MnTE-2-PyP protects fibroblasts from irradiation and hyperglycemia damage by enhancing the NRF2- mediated pathway in diabetic prostate cancer patients, undergoing radiotherapy.


Asunto(s)
Diabetes Mellitus , Metaloporfirinas , Porfirinas , Animales , Antioxidantes , Humanos , Masculino , Manganeso , Metaloporfirinas/farmacología , Ratones
19.
J Exp Med ; 217(7)2020 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-32441762

RESUMEN

Approximately one third of cancer patients die due to complexities related to cachexia. However, the mechanisms of cachexia and the potential therapeutic interventions remain poorly studied. We observed a significant positive correlation between SIRT1 expression and muscle fiber cross-sectional area in pancreatic cancer patients. Rescuing Sirt1 expression by exogenous expression or pharmacological agents reverted cancer cell-induced myotube wasting in culture conditions and mouse models. RNA-seq and follow-up analyses showed cancer cell-mediated SIRT1 loss induced NF-κB signaling in cachectic muscles that enhanced the expression of FOXO transcription factors and NADPH oxidase 4 (Nox4), a key regulator of reactive oxygen species production. Additionally, we observed a negative correlation between NOX4 expression and skeletal muscle fiber cross-sectional area in pancreatic cancer patients. Knocking out Nox4 in skeletal muscles or pharmacological blockade of Nox4 activity abrogated tumor-induced cachexia in mice. Thus, we conclude that targeting the Sirt1-Nox4 axis in muscles is an effective therapeutic intervention for mitigating pancreatic cancer-induced cachexia.


Asunto(s)
Caquexia/complicaciones , Caquexia/metabolismo , NADPH Oxidasa 4/metabolismo , Neoplasias/complicaciones , Neoplasias/metabolismo , Transducción de Señal , Sirtuina 1/metabolismo , Tejido Adiposo/patología , Animales , Línea Celular , Línea Celular Tumoral , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Factores de Transcripción Forkhead/metabolismo , Células HEK293 , Humanos , Metaboloma/efectos de los fármacos , Ratones , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Atrofia Muscular/metabolismo , Atrofia Muscular/patología , FN-kappa B/metabolismo , Oxidación-Reducción , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Estabilidad Proteica/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Resveratrol/farmacología , Transducción de Señal/efectos de los fármacos , Síndrome Debilitante/patología
20.
Int J Mol Sci ; 21(5)2020 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-32182883

RESUMEN

BACKGROUND: Chemotherapy treatment for breast cancer can induce cognitive impairments often involving oxidative stress. The brain, as a whole, is susceptible to oxidative stress due to its high-energy requirements, limited anaerobic respiration capacities, and limited antioxidant defenses. The goal of the current study was to determine if the manganese porphyrin superoxide dismutase mimetic MnTnBuOE-2-PyP (MnBuOE) could ameliorate the effects of doxorubicin, cyclophosphamide, and paclitaxel (AC-T) on mature dendrite morphology and cognitive function. METHODS: Four-month-old female C57BL/6 mice received intraperitoneal injections of chemotherapy followed by subcutaneous injections of MnBuOE. Four weeks following chemotherapy treatment, mice were tested for hippocampus-dependent cognitive performance in the Morris water maze. After testing, brains were collected for Golgi staining and molecular analyses. RESULTS: MnBuOE treatment preserved spatial memory during the Morris water-maze. MnBuOE/AC-T showed spatial memory retention during all probe trials. AC-T treatment significantly impaired spatial memory retention in the first and third probe trial (no platform). AC-T treatment decreased dendritic length in the Cornu Ammonis 1 (CA1) and dentate gyrus (DG) areas of the hippocampus while AC-T/MnBuOE maintained dendritic length. Comparative proteomic analysis revealed affected protein networks associated with cell morphology and behavior functions in both the AC-T and AC-T/MnBuOE treatment groups.


Asunto(s)
Cognición/efectos de los fármacos , Ciclofosfamida/farmacología , Doxorrubicina/farmacología , Hipocampo/efectos de los fármacos , Metaloporfirinas/farmacología , Oxidación-Reducción/efectos de los fármacos , Paclitaxel/farmacología , Animales , Antioxidantes/farmacología , Región CA1 Hipocampal/efectos de los fármacos , Giro Dentado/efectos de los fármacos , Femenino , Ratones , Ratones Endogámicos C57BL , Neurogénesis/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Proteómica/métodos , Protectores contra Radiación/farmacología , Memoria Espacial/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA