Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 17 de 17
1.
J Immunother Cancer ; 12(4)2024 Apr 16.
Article En | MEDLINE | ID: mdl-38631714

BACKGROUND: Lymphocytic choriomeningitis virus (LCMV) belongs to the Arenavirus family known for inducing strong cytotoxic T-cell responses in both mice and humans. LCMV has been engineered for the development of cancer immunotherapies, currently undergoing evaluation in phase I/II clinical trials. Initial findings have demonstrated safety and an exceptional ability to activate and expand tumor-specific T lymphocytes. Combination strategies to maximize the antitumor effectiveness of LCMV-based immunotherapies are being explored. METHODS: We assessed the antitumor therapeutic effects of intratumoral administration of polyinosinic:polycytidylic acid (poly(I:C)) and systemic vaccination using an LCMV-vector expressing non-oncogenic versions of the E6 and E7 antigens of human papillomavirus 16 (artLCMV-E7E6) in a bilateral model engrafting TC-1/A9 cells. This cell line, derived from the parental TC-1, exhibits low MHC class I expression and is highly immune-resistant. The mechanisms underlying the combination's efficacy were investigated through bulk RNA-seq, flow cytometry analyses of the tumor microenvironment, selective depletions using antibodies and clodronate liposomes, Batf3 deficient mice, and in vivo bioluminescence experiments. Finally, we assessed the antitumor effectiveness of the combination of artLCMV-E7E6 with BO-112, a GMP-grade poly(I:C) formulated in polyethyleneimine, currently under evaluation in clinical trials. RESULTS: Intratumoral injection of poly(I:C) enhanced the antitumor efficacy of artLCMV-E7E6 in both injected and non-injected tumor lesions. The combined treatment resulted in a significant delay in tumor growth and often complete eradication of several tumor lesions, leading to significantly improved survival compared with monotherapies. While intratumoral administration of poly(I:C) did not impact LCMV vector biodistribution or transgene expression, it significantly modified leucocyte infiltrates within the tumor microenvironment and amplified systemic efficacy through proinflammatory cytokines/chemokines such as CCL3, CCL5, CXCL10, TNF, IFNα, and IL12p70. Upregulation of MHC on tumor cells and a reconfiguration of the gene expression programs related to tumor vasculature, leucocyte migration, and the activation profile of tumor-infiltrating CD8+ T lymphocytes were observed. Indeed, the antitumor effect relied on the functions of CD8+ T lymphocytes and macrophages. The synergistic efficacy of the combination was further confirmed when BO-112 was included. CONCLUSION: Intratumoral injection of poly(I:C) sensitizes MHClow tumors to the antitumor effects of artLCMV-E7E6, resulting in a potent therapeutic synergy.


Lymphocytic choriomeningitis virus , Neoplasms , Poly I-C , Animals , Humans , Mice , Injections, Intralesional , Tissue Distribution , Immunotherapy/methods , Adjuvants, Immunologic , Tumor Microenvironment
2.
Oncoimmunology ; 12(1): 2197370, 2023.
Article En | MEDLINE | ID: mdl-37035637

BO-112 is a poly I:C-based viral mimetic that exerts anti-tumor efficacy when intratumorally delivered in mouse models. Intratumoral BO-112 synergizes in mice with systemic anti-PD-1 mAbs and this combination has attained efficacy in PD1-refractory melanoma patients. We sought to evaluate the anti-tumor efficacy of BO-112 pre-surgically applied in neoadjuvant settings to mouse models. We have observed that repeated intratumoral injections of BO-112 prior to surgical excision of the primary tumor significantly reduced tumor metastasis from orthotopically implanted 4T1-derived tumors and subcutaneous MC38-derived tumors in mice. Such effects were enhanced when combined with systemic anti-PD-1 mAb. The anti-tumor efficacy of this neoadjuvant immunotherapy approach depended on the presence of antigen-specific effector CD8 T cells and cDC1 antigen-presenting cells. Since BO-112 has been successful in phase-two clinical trials for metastatic melanoma, these results provide a strong rationale for translating this pre-surgical strategy into clinical settings, especially in combination with standard-of-care checkpoint inhibitors.


Melanoma , Neoadjuvant Therapy , Animals , Mice , T-Lymphocytes , Immunotherapy/methods , Melanoma/drug therapy , Antibodies, Monoclonal/pharmacology , Adjuvants, Immunologic
3.
J Immunother Cancer ; 11(1)2023 01.
Article En | MEDLINE | ID: mdl-36631161

BACKGROUND: Radioimmunotherapy combines irradiation of tumor lesions with immunotherapy to achieve local and abscopal control of cancer. Most immunotherapy agents are given systemically, but strategies for delivering immunotherapy locally are under clinical scrutiny to maximize efficacy and avoid toxicity. Local immunotherapy, by injecting various pathogen-associated molecular patterns, has shown efficacy both preclinically and clinically. BO-112 is a viral mimetic based on nanoplexed double-stranded RNA (poly I:C) which exerts immune-mediated antitumor effects in mice and humans on intratumoral delivery. BO-112 and focal irradiation were used to make the proof-of-concept for local immunotherapy plus radiation therapy combinations. METHODS: Murine transplantable tumor cell lines (TS/A, MC38 and B16-OVA) were used to show increased immunogenic features under irradiation, as well as in bilateral tumor models in which only one of the lesions was irradiated or/and injected with BO-112. Flow cytometry and multiplex tissue immunofluorescence were used to determine the effects on antitumor immunity. Depletions of immune cell populations and knockout mice for the IFNAR and BATF-3 genes were used to delineate the immune system requirements for efficacy. RESULTS: In cultures of TS/A breast cancer cells, the combination of irradiation and BO-112 showed more prominent features of immunogenic tumor cell death in terms of calreticulin exposure. Injection of BO-112 into the tumor lesion receiving radiation achieved excellent control of the treated tumor and modest delays in contralateral tumor progression. Local effects were associated with more prominent infiltrates of antitumor cytotoxic tumor lymphocytes (CTLs). Importantly, local irradiation plus BO-112 in one of the tumor lesions that enhanced the therapeutic effects of radiotherapy on distant irradiated lesions that were not injected with BO-112. Hence, this beneficial effect of local irradiation plus BO-112 on a tumor lesion enhanced the therapeutic response to radiotherapy on distant non-injected lesions. CONCLUSION: This study demonstrates that local BO-112 immunotherapy and focal irradiation may act in synergy to achieve local tumor control. Irradiation plus BO-112 in one of the tumor lesions enhanced the therapeutic effects on distant irradiated lesions that were not injected with BO-112, suggesting strategies to treat oligometastatic patients with lesions susceptible to radiotherapy and with at least one tumor accessible for repeated BO-112 intratumoral injections.


CD8-Positive T-Lymphocytes , Poly I-C , Radioimmunotherapy , Animals , Mice , Adjuvants, Immunologic/metabolism , Immunotherapy , Poly I-C/metabolism
4.
J Immunother Cancer ; 9(11)2021 11.
Article En | MEDLINE | ID: mdl-34824158

BACKGROUND: BO-112 is a nanoplexed form of polyinosinic:polycytidylic acid that acting on toll-like receptor 3 (TLR3), melanoma differentiation-associated protein 5 (MDA5) and protein kinase RNA-activated (PKR) elicits rejection of directly injected transplanted tumors, but has only modest efficacy against distant untreated tumors. Its clinical activity has also been documented in early phase clinical trials. The 5,6-dimethylxanthenone-4-acetic acid (DMXAA) stimulator of interferon genes (STING) agonist shows a comparable pattern of efficacy when used via intratumoral injections. METHODS: Mice subcutaneously engrafted with bilateral MC38 and B16.OVA-derived tumors were treated with proinflammatory immunotherapy agents known to be active when intratumorally delivered. The combination of BO-112 and DMXAA was chosen given its excellent efficacy and the requirements for antitumor effects were studied on selective depletion of immune cell types and in gene-modified mouse strains lacking basic leucine zipper ATF-like transcription factor 3 (BATF3), interferon-α/ß receptor (IFNAR) or STING. Spatial requirements for the injections were studied in mice bearing three tumor lesions. RESULTS: BO-112 and DMXAA when co-injected in one of the lesions of mice bearing concomitant bilateral tumors frequently achieved complete local and distant antitumor efficacy. Synergistic effects were contingent on CD8 T cell lymphocytes and dependent on conventional type 1 dendritic cells, responsiveness to type I interferon (IFN) and STING function in the tumor-bearing host. Efficacy was preserved even if BO-112 and DMXAA were injected in separate lesions in a manner able to control another untreated third-party tumor. Efficacy could be further enhanced on concurrent PD-1 blockade. CONCLUSION: Clinically feasible co-injections of BO-112 and a STING agonist attain synergistic efficacy able to eradicate distant untreated tumor lesions.


Dendritic Cells/immunology , Immunotherapy/methods , Poly I-C/metabolism , Animals , Disease Models, Animal , Humans , Injections, Intralesional , Mice
5.
Sci Transl Med ; 12(565)2020 10 14.
Article En | MEDLINE | ID: mdl-33055241

Intratumoral therapies, especially Toll-like receptor agonists, can trigger both the innate and adaptive immune systems. BO-112 is a nanoplexed form of polyinosinic:polycytidylic acid (poly I:C) that induces local and systemic immunotherapeutic effects in mouse models. In a multicenter phase 1 clinical trial, repeated intratumoral administrations of BO-112 induced an increase in tumor cell necrosis and apoptosis, as well as augmented immune reactivity according to gene expression profiling. The first three cohorts receiving BO-112 as a monotherapy resulted in a recommended dose of 1 mg that could be safely repeated. Two grade 3 to 4 adverse reactions in the form of reversible thrombocytopenia were reported. In a fourth cohort of 28 patients with tumors that had primary resistance to anti-programmed cell death protein-1 (PD-1), the combination of intratumoral BO-112 with nivolumab or pembrolizumab was also well tolerated, and 3 patients (2 with melanoma and 1 with renal cell carcinoma) achieved partial responses, with 10 more patients having stable disease at 8 to 12 weeks. Thus, local BO-112 combined with a systemic anti-PD-1 agent might be a strategy to revert anti-PD-1 resistance.


Carcinoma, Renal Cell , Kidney Neoplasms , Melanoma , Animals , Humans , Melanoma/drug therapy , Mice , Nivolumab/therapeutic use , Poly I
6.
Sci Transl Med ; 12(565)2020 10 14.
Article En | MEDLINE | ID: mdl-33055240

Defects in tumor-intrinsic interferon (IFN) signaling result in failure of immune checkpoint blockade (ICB) against cancer, but these tumors may still maintain sensitivity to T cell-based adoptive cell therapy (ACT). We generated models of IFN signaling defects in B16 murine melanoma observed in patients with acquired resistance to ICB. Tumors lacking Jak1 or Jak2 did not respond to ICB, whereas ACT was effective against Jak2 KO tumors, but not Jak1 KO tumors, where both type I and II tumor IFN signaling were defective. This was a direct result of low baseline class I major histocompatibility complex (MHC I) expression in B16 and the dependency of MHC I expression on either type I or type II IFN signaling. We used genetic and pharmacologic approaches to uncouple this dependency and restore MHC I expression. Through independent mechanisms, overexpression of NLRC5 (nucleotide-binding oligomerization domain-like receptor family caspase recruitment domain containing 5) and intratumoral delivery of BO-112, a potent nanoplexed version of polyinosinic:polycytidylic acid (poly I:C), each restored the efficacy of ACT against B16-Jak1 KO tumors. BO-112 activated double-stranded RNA (dsRNA) sensing (via protein kinase R and Toll-like receptor 3) and induced MHC I expression via nuclear factor κB, independent of both IFN signaling and NLRC5. In summary, we demonstrated that in the absence of tumor IFN signaling, MHC I expression is essential and sufficient for the efficacy of ACT. For tumors lacking MHC I expression due to deficient IFN signaling, activation of dsRNA sensors by BO-112 affords an alternative approach to restore the efficacy of ACT.


Antigen Presentation , Interferon-gamma , Animals , Humans , Immunotherapy , Intracellular Signaling Peptides and Proteins , Janus Kinase 1 , Mice , NF-kappa B , Signal Transduction
7.
J Immunother Cancer ; 7(1): 116, 2019 05 02.
Article En | MEDLINE | ID: mdl-31046839

Poly I:C is a powerful immune adjuvant as a result of its agonist activities on TLR-3, MDA5 and RIG-I. BO-112 is a nanoplexed formulation of Poly I:C complexed with polyethylenimine that causes tumor cell apoptosis showing immunogenic cell death features and which upon intratumoral release results in more prominent tumor infiltration by T lymphocytes. Intratumoral treatment with BO-112 of subcutaneous tumors derived from MC38, 4 T1 and B16-F10 leads to remarkable local disease control dependent on type-1 interferon and gamma-interferon. Some degree of control of non-injected tumor lesions following BO-112 intratumoral treatment was found in mice bearing bilateral B16-OVA melanomas, an activity which was enhanced with co-treatment with systemic anti-CD137 and anti-PD-L1 mAbs. More abundant CD8+ T lymphocytes were found in B16-OVA tumor-draining lymph nodes and in the tumor microenvironment following intratumoral BO-112 treatment, with enhanced numbers of tumor antigen-specific cytotoxic T lymphocytes. Genome-wide transcriptome analyses of injected tumor lesions were consistent with a marked upregulation of the type-I interferon pathway. Inspired by these data, intratumorally delivered BO-112 is being tested in cancer patients (NCT02828098).


Interferon Inducers/administration & dosage , Interferon Type I/metabolism , Melanoma, Experimental/drug therapy , Poly I-C/administration & dosage , Tumor Microenvironment/drug effects , Animals , Cell Line, Tumor/transplantation , Drug Screening Assays, Antitumor , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Injections, Intralesional , Interferon Type I/immunology , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Up-Regulation/drug effects
8.
Head Neck ; 30(2): 208-14, 2008 Feb.
Article En | MEDLINE | ID: mdl-17657783

BACKGROUND: Extracapsular nodal spread is a major prognostic indicator in head and neck cancer. Nitric oxide (NO), primarily produced by the enzyme inducible NO synthase (iNOS), has a large number of actions in cancer biology, but no studies have investigated its possible role in extracapsular spread or tumor invasion. METHODS: Immunochemistry was used to study iNOS expression in 48 patients with either extracapsular or encapsulated metastasis. In vitro invasion assays were performed using H357 (an oral squamous cell carcinoma cell line) using the iNOS inhibitor drug, 1400 W. RESULTS: iNOS expression was significantly associated with extracapsular spread, with 22/27 cases showing positive iNOS expression compared with 8/21 cases in the encapsulated group (p = .01). Invasion of H357 cells was inhibited by 1400 W at concentrations of 100 microM and 1 mM (p = .002, p = .003). CONCLUSION: iNOS protein seems to be associated with extracapsular spread and invasion in head and neck cancer. Further studies are required to understand this role more fully.


Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Nitric Oxide Synthase Type II/metabolism , Adult , Aged , Aged, 80 and over , Female , Humans , Immunohistochemistry , Male , Middle Aged , Neoplasm Invasiveness , Tumor Cells, Cultured
9.
Exp Gerontol ; 42(9): 904-10, 2007 Sep.
Article En | MEDLINE | ID: mdl-17339088

Telomerase, a reverse transcriptase involved in the maintenance of telomere function and cellular replicative capacity, is thought to be regulated by nitric oxide (NO). Here, we have used pharmacological tools and RNA interference to re-assess the role of NO in the regulation of telomerase and senescence of human umbilical vein endothelial cells. Acute or chronic treatment of these cells with the NO donors diethylenetriamine/NO (DETA-NO) or S-nitroso-N-acetylpenicillamine (SNAP) at concentrations which generated NO in the 1-300 nM range did not modulate telomerase activity. Similarly these agents did not affect cellular replicative capacity during long-term sub-cultivation. The NO synthase (NOS) inhibitor N(G)-monomethyl-L-arginine (1 mM) reduced basal levels of c-GMP by 50% but had no effect on telomerase activity or replicative capacity. Withdrawal of ascorbic acid increased the intracellular pro-oxidant capacity, reduced telomerase activity and increased the accumulation of senescent cells upon serial passage in culture. However, this shift to a more oxidative redox state did not unmask the putative capacity of NO to modulate telomerase or senescence. Infection of cells with a lentiviral vector expressing a small hairpin RNA targeted against endothelial NOS inhibited endogenous NO production completely but failed to affect the decrease of telomerase activity or the accumulation of senescent cells observed with passage in culture. Our findings suggest that physiological concentrations of NO do not modulate telomerase levels or replicative capacity of endothelial cells, regardless of their cellular oxidative status.


Endothelial Cells/enzymology , Nitric Oxide/metabolism , Telomerase/metabolism , Cell Division/physiology , Cells, Cultured , Cellular Senescence , Gene Silencing , Humans , Nitric Oxide/analysis , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Oxidation-Reduction , Penicillamine/analogs & derivatives , Penicillamine/pharmacology , RNA, Small Interfering/genetics , Time Factors , Transfection/methods , omega-N-Methylarginine/pharmacology
10.
Proc Natl Acad Sci U S A ; 104(15): 6223-8, 2007 Apr 10.
Article En | MEDLINE | ID: mdl-17384149

An increased dependency on glycolysis for ATP production is considered to be a hallmark of tumor cells. Whether this increase in glycolytic activity is due mainly to inherent metabolic alterations or to the hypoxic microenvironment remains controversial. Here we have transformed human adult mesenchymal stem cells (MSC) using genetic alterations as described for differentiated cells. Our data suggest that MSC require disruption of the same pathways as have been shown for differentiated cells to confer a fully transformed phenotype. Furthermore, we found that MSC are more glycolytic than primary human fibroblasts and, in contrast to differentiated cells, do not depend on increased aerobic glycolysis for ATP production during transformation. These data indicate that aerobic glycolysis (the Warburg effect) is not an intrinsic component of the transformation of adult stem cells, and that oncogenic adaptation to bioenergetic requirements, in some circumstances, may also rely on increases in oxidative phosphorylation. We did find, however, a reversible increase in the transcription of glycolytic enzymes in tumors generated by transformed MSC, indicating this is a secondary phenomenon resulting from adaptation of the tumor to its microenvironment.


Adenosine Triphosphate/biosynthesis , Cell Transformation, Neoplastic/metabolism , Energy Metabolism/physiology , Mesenchymal Stem Cells/physiology , Oxidative Phosphorylation , Adult , Blotting, Western , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Glucose/metabolism , Glucosephosphate Dehydrogenase/metabolism , Glycolysis/physiology , Humans , Immunophenotyping , Lactic Acid/metabolism , Male , Mesenchymal Stem Cells/metabolism , NADP/metabolism
11.
Kasmera ; 34(1): 31-39, ene.-jun. 2006. tab, graf
Article Es | LILACS | ID: lil-462813

La enfermedad diarreica representa un problema importante de salud pública, siendo las parasitosis intestinales una de sus causas. El objetivo de este estudio fue identificar especies de enteroparásitos asociados a diarrea aguda en niños menores de 12 años de edad. Se estudiaron 58 niños de ambos sexos estratificados en tres grupos de edad: menores de 1 año, de 1 a 4 años y de 5 a 12 años. A cada niño se le tomó una muestra fecal a la cual se le practicó los métodos coproparasitológicos con solución salina fisiológica, coloraciones temporales con lugol y azul de metileno amortiguado y coloraciones permanentes de Ziehl Neelsen modificado y Acido Ràpido Tricrómica. El 36,20 por ciento de los individuos presentaban enteroparásitos. Las especies parasitarias identificadas fueron las siguientes: Blastocystis hominis 17,24 por ciento, Complejo Entamoeba histolytica/E. Dispar 10,34 por ciento, Cryptosporidium parvum 6,90 por ciento, Giardia lamblia 6,90 por ciento, Entamoeba coli 3,45 por ciento, Chilomastix mesnili, Endolimax nana y Pentatrichomonas hominis 1,72 por ciento cada uno. El Chi cuadrado reveló diferencias estadísticamente significativas entre las variables parasitosis y sexo masculino, edad (5-12 años) ymonoparasitismo (p< 0.05). Estos resultados revelan la persistencia de estas infecciones en niños con diarrea aguda menores de 12 años


Humans , Male , Female , Child , Blastocystis hominis , Diarrhea, Infantile , Entamoeba histolytica , Giardia lamblia , Intestinal Diseases, Parasitic , Parasitology , Pediatrics , Venezuela
12.
Proc Natl Acad Sci U S A ; 103(14): 5379-84, 2006 Apr 04.
Article En | MEDLINE | ID: mdl-16565215

Vascular endothelial cells are highly glycolytic and consume relatively low amounts of oxygen (O(2)) compared with other cells. We have confirmed that oxidative phosphorylation is not the main source of ATP generation in these cells. We also show that at a low O(2) concentration (<1%) endogenous NO plays a key role in preventing the accumulation of the alpha-subunit of hypoxia-inducible factor 1. At higher O(2) concentrations (1-3%) NO facilitates the production of mitochondrial reactive oxygen species. This production activates the AMP-activated protein kinase by a mechanism independent of nucleotide concentrations. Thus, the primary role of mitochondria in vascular endothelial cells may not be to generate ATP but, under the control of NO, to act as signaling organelles using either O(2) or O(2)-derived species as signaling molecules. Diversion of O(2) away from endothelial cell mitochondria by NO might also facilitate oxygenation of vascular smooth muscle cells.


Endothelium, Vascular/metabolism , Mitochondria/physiology , Signal Transduction/physiology , Adenylate Kinase/metabolism , Base Sequence , Blotting, Western , Cells, Cultured , DNA Primers , Endothelium, Vascular/enzymology , Endothelium, Vascular/ultrastructure , Enzyme Activation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mitochondria/enzymology , Mitochondria/metabolism , Nitric Oxide/physiology , Oxidative Phosphorylation , Oxygen/metabolism
13.
Cancer Res ; 66(2): 770-4, 2006 Jan 15.
Article En | MEDLINE | ID: mdl-16424008

Widespread expression of the alpha-subunit of hypoxia-inducible factor (HIF-1alpha) was observed in samples of human oral squamous cell carcinoma. In all the cases, this was accompanied by a widespread distribution of nitric oxide (NO) synthases (NOS). Furthermore, in three human cell lines derived from human oral squamous cell carcinoma, the accumulation of HIF-1alpha was prevented either by inhibition of NOS activity with the nonspecific NOS inhibitor N(G)-monomethyl-L-arginine or by the antioxidants N-acetyl-L-cysteine and ascorbic acid. We suggest that, in certain forms of cancer, NO might be responsible for the accumulation of HIF-1alpha by a mechanism dependent on free radicals.


Carcinoma, Squamous Cell/physiopathology , Free Radicals , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Mouth Neoplasms/physiopathology , Nitric Oxide/toxicity , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Immunohistochemistry , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase/physiology
14.
Biochem Biophys Res Commun ; 322(3): 923-9, 2004 Sep 24.
Article En | MEDLINE | ID: mdl-15336552

2-Methoxyestradiol (2ME2), a naturally occurring metabolite of estradiol, is known to have antiproliferative, antiangiogenic, and proapoptotic activity. Mechanistically, 2ME2 has been shown to downregulate hypoxia-inducible factor 1alpha (HIF1alpha) and to induce apoptosis in tumour cells by generating reactive oxygen species (ROS). In this study we report that 2ME2 inhibits mitochondrial respiration in both intact cells and submitochondrial particles, and that this effect is due to inhibition of complex I of the mitochondrial electron transport chain (ETC). The prevention by 2ME2 of hypoxia-induced stabilisation of HIF1alpha in HEK293 cells was found not to be due to an effect on HIF1alpha synthesis but rather to an effect on protein degradation. This is in agreement with our recent observation using other inhibitors of mitochondrial respiration which bring about rapid degradation of HIF1alpha in hypoxia due to increased availability of oxygen and reactivation of prolyl hydroxylases. The concentrations of 2ME2 that inhibited complex I also induced the generation of ROS. 2ME2 did not, however, cause generation of ROS in 143B rho(-) cells, which lack a functional mitochondrial ETC. We conclude that inhibition of mitochondrial respiration explains, at least in part, the effect of 2ME2 on hypoxia-dependent HIF1alpha stabilisation and cellular ROS production. Since these actions of 2ME2 occur at higher concentrations than those known to inhibit cell proliferation, it remains to be established whether they contribute to its therapeutic effect.


Anticarcinogenic Agents/pharmacology , Estradiol/analogs & derivatives , Estradiol/pharmacology , Mitochondria/metabolism , Oxygen Consumption/drug effects , 2-Methoxyestradiol , Apoptosis/drug effects , Cell Division/drug effects , Cell Line , Electron Transport/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit , Kidney , Kinetics , Mitochondria/drug effects , Reactive Oxygen Species/metabolism , Transcription Factors/drug effects , Transcription Factors/physiology
15.
Proc Natl Acad Sci U S A ; 101(20): 7630-5, 2004 May 18.
Article En | MEDLINE | ID: mdl-15136725

We have investigated in whole cells whether, at low oxygen concentrations ([O(2)]), endogenous nitric oxide (NO) modulates the redox state of the mitochondrial electron transport chain (ETC), and whether such an action has any signaling consequences. Using a polarographic-and-spectroscopic-coupled system, we monitored redox changes in the ETC cytochromes b(H), cc(1), and aa(3) during cellular respiration. The rate of O(2) consumption (VO(2)) remained constant until [O(2)] fell below 15 microM, whereas the onset of reduction of cytochromes aa(3), part of the terminal ETC enzyme cytochrome c oxidase, occurred at approximately 50 microM O(2). Incubation of the cells with an inhibitor of NO synthase lowered significantly (P < 0.05) the [O(2)] at which reduction of the cytochromes occurred. We also measured intracellular superoxide (O(2)(-)) production at different [O(2)] and found there was no increase in O(2)(-) generation in control cells, or those treated with the NO synthase inhibitor, when incubated at 21% O(2). However, after 30-min exposure of control cells to 3% O(2), an increase in O(2)(-) generation was observed, accompanied by translocation to the nucleus of the transcription factor NF-kappa B. Both of these responses were diminished by NO synthase inhibition. Our results suggest that endogenous NO, by enhancing the reduction of ETC cytochromes, contributes to a mechanism by which cells maintain their VO(2) at low [O(2)]. This, in turn, favors the release of O(2)(-), which initiates the transcriptional activation of NF-kappa B as an early signaling stress response.


Electron Transport Complex IV/metabolism , Hypoxia/metabolism , Nitric Oxide/metabolism , Superoxides/metabolism , Animals , Cell Respiration/physiology , Mice , Monocytes , NF-kappa B/metabolism , Oxidation-Reduction , Spectrophotometry
16.
Proc Natl Acad Sci U S A ; 99(13): 8892-7, 2002 Jun 25.
Article En | MEDLINE | ID: mdl-12077295

We have found that activation of human adult T cell leukemia (Jurkat) cells with anti-Fas Ab leads, in a concentration-dependent manner, to an early burst of production of nitric oxide (NO), which inhibits cell respiration. This results in mitochondrial hyperpolarization, dependent on the hydrolysis of glycolytic ATP by the F1F(o)-ATPase acting in reverse mode. During this early phase of activation, there is a transient release of superoxide anion. All these processes can be prevented by an inhibitor of NO synthase. Approximately 2 h after stimulation with anti-Fas Ab, a distinct second phase can be detected. This comprises a concentration-dependent collapse in mitochondrial membrane potential, a second wave of free radical production, and activation of caspase-8 leading to apoptosis. This second phase is abolished by an inhibitor of caspase activation. In contrast, inhibition of NO synthesis leads to an enhancement and acceleration of these latter processes, suggesting that the early NO-dependent phase represents a protective mechanism. The significance of the two phases in relation to cell survival and death remains to be studied.


Mitochondria/physiology , Nitric Oxide/physiology , Signal Transduction , fas Receptor/metabolism , Apoptosis , Caspase 8 , Caspase 9 , Caspases/metabolism , Cell Survival , Humans , Jurkat Cells , Membrane Potentials , Mitochondria/enzymology , Oxygen Consumption , Reactive Oxygen Species , fas Receptor/immunology
17.
Arch. venez. pueric. pediatr ; 48(3/4): 86-90, jul.-dic. 1985. ilus, tab
Article Es | LILACS | ID: lil-29065

La prevalencia de portadores sanos de HBsAg en 840 mujeres embarazadas que asisten a la consulta prenatal del HUC es del 2.7% y del HBeAg 0%. Del total de 23 madres positivas han nacido hasta el momento 14 niños, que al control, sus respectivos HBsAg permanecen negativos después de haber recibido HB-VAX con o sin HBIG. Consideramos que las madres estudiadas provienen en su mayoría de estratos socio-economicos bajos por lo que son de alto riesgo, por lo tanto, el despistaje mediante la determinación de HBsAg en todas las embarazadas disminuiría el riesgo de transmisión perinatal, y el uso de la vacúna, aun sin HBIG, dado el 100% de negatividad para HBeAg, pudiera ser adecuado a un bajo costo en la profilaxis de la hepatitis a virus B en la mayoría de recién nacidos a riesgo


Adolescent , Adult , Middle Aged , Humans , Female , Hepatitis B Surface Antigens/analysis , Hepatitis B e Antigens/analysis , Hepatitis B virus/immunology , Hepatitis B/immunology , Maternal-Fetal Exchange , Pregnancy , Venezuela
...