Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Part Fibre Toxicol ; 19(1): 10, 2022 02 09.
Article in English | MEDLINE | ID: mdl-35135577

ABSTRACT

BACKGROUND: The gut microbiota plays a vital role in host homeostasis and is associated with inflammation and cardiovascular disease (CVD) risk. Exposure to particulate matter (PM) is a known mediator of inflammation and CVD and is reported to promote dysbiosis and decreased intestinal integrity. However, the role of inhaled traffic-generated PM on the gut microbiome and its corresponding systemic effects are not well-characterized. Thus, we investigated the hypothesis that exposure to inhaled diesel exhaust particles (DEP) alters the gut microbiome and promotes microbial-related inflammation and CVD biomarkers. 4-6-week-old male C57Bl/6 mice on either a low-fat (LF, 10% fat) or high-fat (HF, 45% fat) diet were exposed via oropharyngeal aspiration to 35 µg DEP suspended in 35 µl saline or saline only (CON) 2x/week for 30 days. To determine whether probiotics could prevent diet or DEP exposure mediated alterations in the gut microbiome or systemic outcomes, a subset of animals on the HF diet were treated orally with 0.3 g/day (~ 7.5 × 108 CFU/day) of Winclove Ecologic® Barrier probiotics throughout the study. RESULTS: Our results show that inhaled DEP exposure alters gut microbial profiles, including reducing Actinobacteria and expanding Verrucomicrobia and Proteobacteria. We observed increased circulating LPS, altered circulating cytokines (IL-1α, IL-3, IL-13, IL-15, G-CSF, LIF, MIP-2, and TNF-α), and CVD biomarkers (siCAM, PAI-1, sP-Selectin, thrombomodulin, and PECAM) in DEP-exposed and/or HF diet mice. Furthermore, probiotics attenuated the observed reduction of Actinobacteria and expansion of Proteobacteria in DEP-exposed and HF-diet mice. Probiotics mitigated circulating cytokines (IL-3, IL-13, G-CSF, RANTES, and TNF- α) and CVD biomarkers (siCAM, PAI-1, sP-Selectin, thrombomodulin, and PECAM) in respect to DEP-exposure and/or HF diet. CONCLUSION: Key findings of this study are that inhaled DEP exposure alters small intestinal microbial profiles that play a role in systemic inflammation and early CVD biomarkers. Probiotic treatment in this study was fundamental in understanding the role of inhaled DEP on the microbiome and related systemic inflammatory and CVD biomarkers.


Subject(s)
Cardiovascular Diseases , Microbiota , Animals , Biomarkers , Cardiovascular Diseases/chemically induced , Cytokines , Granulocyte Colony-Stimulating Factor , Inflammation/chemically induced , Interleukin-13 , Interleukin-3 , Male , Mice , Mice, Inbred C57BL , Particulate Matter , Plasminogen Activator Inhibitor 1 , Thrombomodulin , Vehicle Emissions/toxicity
2.
F1000Res ; 9: 217, 2020.
Article in English | MEDLINE | ID: mdl-32399207

ABSTRACT

Background: The conventional dogma of treating cancer by focusing on the elimination of tumor cells has been recently refined to include consideration of the tumor microenvironment, which includes host stromal cells. Ephrin-A1, a cell surface protein involved in adhesion and migration, has been shown to be tumor suppressive in the context of the cancer cell. However, its role in the host has not been fully investigated. Here, we examine how ephrin-A1 host deficiency affects cancer growth and metastasis in a murine model of breast cancer. Methods: 4T1 cells were orthotopically implanted into the mammary fat pads or injected into the tail veins of ephrin-A1 wild-type ( Efna1+/+), heterozygous ( Efna1+/-), or knockout ( Efna1-/-) mice. Tumor growth, lung metastasis, and tumor recurrence after surgical resection were measured. Flow cytometry and immunohistochemistry (IHC) were used to analyze various cell populations in primary tumors and tumor-bearing lungs. Results: While primary tumor growth did not differ between Efna1+/+, Efna1+/-, and Efna1-/- mice, lung metastasis and primary tumor recurrence were significantly decreased in knockout mice. Efna1-/- mice had reduced lung colonization of 4T1 cells compared to Efna1+/+ littermate controls as early as 24 hours after tail vein injection. Furthermore, established lung lesions in Efna1-/- mice had reduced proliferation compared to those in Efna1+/+ controls. Conclusions: Our studies demonstrate that host deficiency of ephrin-A1 does not impact primary tumor growth but does affect metastasis by providing a less favorable metastatic niche for cancer cell colonization and growth. Elucidating the mechanisms by which host ephrin-A1 impacts cancer relapse and metastasis may shed new light on novel therapeutic strategies.


Subject(s)
Ephrin-A1/genetics , Lung Neoplasms/genetics , Neoplasm Metastasis/genetics , Animals , Mice , Mice, Knockout , Neoplasm Recurrence, Local , Tumor Microenvironment
3.
Cancer Causes Control ; 31(4): 393-402, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32124187

ABSTRACT

BACKGROUND: Breast cancer incidence in sub-Saharan Africa (SSA) is increasing, and SSA has the highest age-standardized breast cancer mortality rate worldwide. However, high-quality breast cancer data are limited in SSA. MATERIALS AND METHODS: We examined breast cancer patient and tumor characteristics among women in Lilongwe, Malawi and evaluated risk factor associations with patient outcomes. We consecutively enrolled 100 women ≥ 18 years with newly diagnosed, pathologically confirmed breast cancer into a prospective longitudinal cohort with systematically assessed demographic data, HIV status, and clinical characteristics. Tumor subtypes were further determined by immunohistochemistry, overall survival (OS) was estimated using Kaplan-Meier methods, and hazards ratios (HR) were calculated by Cox proportional hazard analyses. RESULTS: Of the 100 participants, median age was 49 years, 19 were HIV-positive, and 75 presented with late stage (III/IV) disease. HER2-enriched and triple-negative/basal-like subtypes represented 17% and 25% tumors, respectively. One-year OS for the cohort was 74% (95% CI 62-83%). Multivariable analyses revealed mortality was associated with HIV (HR, 5.15; 95% CI 1.58-16.76; p = 0.006), stage IV disease (HR, 8.86; 95% CI 1.07-73.25; p = 0.043), and HER2-enriched (HR, 7.46; 95% CI 1.21-46.07; p = 0.031), and triple-negative subtypes (HR, 7.80; 95% CI 1.39-43.69; p = 0.020). CONCLUSION: Late stage presentation, HER2-enriched and triple-negative subtypes, and HIV coinfection were overrepresented in our cohort relative to resource-rich settings and were associated with mortality. These findings highlight robust opportunities for population- and patient-level interventions across the entire cascade of care to improve breast cancer outcomes in low-income countries in SSA.


Subject(s)
Breast Neoplasms/diagnosis , Breast Neoplasms/therapy , Adult , Aged , Aged, 80 and over , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cohort Studies , Female , HIV Infections/epidemiology , Humans , Immunohistochemistry , Incidence , Longitudinal Studies , Malawi/epidemiology , Middle Aged , Prognosis , Proportional Hazards Models , Prospective Studies , Young Adult
4.
PLoS One ; 11(10): e0164804, 2016.
Article in English | MEDLINE | ID: mdl-27780223

ABSTRACT

Squamous cell carcinoma of the head and neck (HNSCC) accounts for more than 300,000 deaths worldwide per year as a consequence of tumor cell invasion of adjacent structures or metastasis. LIM-only protein 4 (LMO4) and LIM-domain binding protein 1 (LDB1), two directly interacting transcriptional adaptors that have important roles in normal epithelial cell differentiation, have been associated with increased metastasis, decreased differentiation, and shortened survival in carcinoma of the breast. Here, we implicate two LDB1-binding proteins, single-stranded binding protein 2 (SSBP2) and 3 (SSBP3), in controlling LMO4 and LDB1 protein abundance in HNSCC and in regulating specific tumor cell functions in this disease. First, we found that the relative abundance of LMO4, LDB1, and the two SSBPs correlated very significantly in a panel of human HNSCC cell lines. Second, expression of these proteins in tumor primaries and lymph nodes involved by metastasis were concordant in 3 of 3 sets of tissue. Third, using a Matrigel invasion and organotypic reconstruct assay, CRISPR/Cas9-mediated deletion of LDB1 in the VU-SCC-1729 cell line, which is highly invasive of basement membrane and cellular monolayers, reduced tumor cell invasiveness and migration, as well as proliferation on tissue culture plastic. Finally, inactivation of the LDB1 gene in these cells decreased growth and vascularization of xenografted human tumor cells in vivo. These data show that LMO4, LDB1, and SSBP2 and/or SSBP3 regulate metastasis, proliferation, and angiogenesis in HNSCC and provide the first evidence that SSBPs control LMO4 and LDB1 protein abundance in a cancer context.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carcinoma, Squamous Cell/pathology , DNA-Binding Proteins/metabolism , LIM Domain Proteins/metabolism , Mouth Neoplasms/pathology , Transcription Factors/metabolism , Animals , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Mice , Mouth Neoplasms/metabolism , Neoplasm Metastasis , Neoplasm Transplantation , Tissue Culture Techniques
5.
Cancer Res ; 76(7): 1825-36, 2016 04 01.
Article in English | MEDLINE | ID: mdl-26833123

ABSTRACT

Dysregulation of receptor tyrosine kinases (RTK) contributes to cellular transformation and cancer progression by disrupting key metabolic signaling pathways. The EPHA2 RTK is overexpressed in aggressive forms of breast cancer, including the HER2(+) subtype, and correlates with poor prognosis. However, the role of EPHA2 in tumor metabolism remains unexplored. In this study, we used in vivo and in vitro models of HER2-overexpressing breast cancer to investigate the mechanisms by which EPHA2 ligand-independent signaling promotes tumorigenesis in the absence of its prototypic ligand, ephrin-A1. We demonstrate that ephrin-A1 loss leads to upregulated glutamine metabolism and lipid accumulation that enhanced tumor growth. Global metabolic profiling of ephrin-A1-null, HER2-overexpressing mammary tumors revealed a significant increase in glutaminolysis, a critical metabolic pathway that generates intermediates for lipogenesis. Pharmacologic inhibition of glutaminase activity reduced tumor growth in both ephrin-A1-depleted and EPHA2-overexpressing tumor allografts in vivo Mechanistically, we show that the enhanced proliferation and glutaminolysis in the absence of ephrin-A1 were attributed to increased RhoA-dependent glutaminase activity. EPHA2 depletion or pharmacologic inhibition of Rho, glutaminase, or fatty acid synthase abrogated the increased lipid content and proliferative effects of ephrin-A1 knockdown. Together, these findings highlight a novel, unsuspected connection between the EPHA2/ephrin-A1 signaling axis and tumor metabolism, and suggest potential new therapeutic targets in cancer subtypes exhibiting glutamine dependency. Cancer Res; 76(7); 1825-36. ©2016 AACR.


Subject(s)
Breast Neoplasms/genetics , Ephrin-A1/metabolism , Glutamine/metabolism , Receptor, EphA2/metabolism , Animals , Breast Neoplasms/pathology , Cell Proliferation , Female , Humans , Mice , Signal Transduction
6.
Mol Cell Biol ; 35(7): 1299-313, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25582201

ABSTRACT

Mammalian target of rapamycin (mTOR) is a serine/threonine kinase that regulates a diverse array of cellular processes, including cell growth, survival, metabolism, and cytoskeleton dynamics. mTOR functions in two distinct complexes, mTORC1 and mTORC2, whose activities and substrate specificities are regulated by complex specific cofactors, including Raptor and Rictor, respectively. Little is known regarding the relative contribution of mTORC1 versus mTORC2 in vascular endothelial cells. Using mouse models of Raptor or Rictor gene targeting, we discovered that Rictor ablation inhibited vascular endothelial growth factor (VEGF)-induced endothelial cell proliferation and assembly in vitro and angiogenesis in vivo, whereas the loss of Raptor had only a modest effect on endothelial cells (ECs). Mechanistically, the loss of Rictor reduced the phosphorylation of AKT, protein kinase Cα (PKCα), and NDRG1 without affecting the mTORC1 pathway. In contrast, the loss of Raptor increased the phosphorylation of AKT despite inhibiting the phosphorylation of S6K1, a direct target of mTORC1. Reconstitution of Rictor-null cells with myristoylated AKT (Myr-AKT) rescued vascular assembly in Rictor-deficient endothelial cells, whereas PKCα rescued proliferation defects. Furthermore, tumor neovascularization in vivo was significantly decreased upon EC-specific Rictor deletion in mice. These data indicate that mTORC2 is a critical signaling node required for VEGF-mediated angiogenesis through the regulation of AKT and PKCα in vascular endothelial cells.


Subject(s)
Cell Proliferation , Endothelial Cells/cytology , Multiprotein Complexes/metabolism , Neovascularization, Physiologic , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Vascular Endothelial Growth Factor A/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Endothelial Cells/metabolism , Gene Deletion , Human Umbilical Vein Endothelial Cells , Humans , Mechanistic Target of Rapamycin Complex 2 , Mice , Phosphorylation , Protein Kinase C-alpha/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rapamycin-Insensitive Companion of mTOR Protein , Regulatory-Associated Protein of mTOR
7.
Mol Cancer Res ; 13(3): 524-37, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25504371

ABSTRACT

UNLABELLED: Angiogenic remodeling during embryonic development and in adult tissue homeostasis is orchestrated by cooperative signaling between several distinct molecular pathways, which are often exploited by tumors. Indeed, tumors upregulate proangiogenic molecules while simultaneously suppressing angiostatic pathways to recruit blood vessels for growth, survival, and metastatic spread. Understanding how cancers exploit proangiogenic and antiangiogenic signals is a key step in developing new, molecularly targeted antiangiogenic therapies. While EphA2, a receptor tyrosine kinase (RTK), is required for VEGF-induced angiogenesis, the mechanism through which these pathways intersect remains unclear. Slit2 expression is elevated in EphA2-deficient endothelium, and here it is reported that inhibiting Slit activity rescues VEGF-induced angiogenesis in cell culture and in vivo, as well as VEGF-dependent tumor angiogenesis, in EphA2-deficient endothelial cells and animals. Moreover, blocking Slit activity or Slit2 expression in EphA2-deficient endothelial cells restores VEGF-induced activation of Src and Rac, both of which are required for VEGF-mediated angiogenesis. These data suggest that EphA2 suppression of Slit2 expression and Slit angiostatic activity enables VEGF-induced angiogenesis in vitro and in vivo, providing a plausible mechanism for impaired endothelial responses to VEGF in the absence of EphA2 function. IMPLICATIONS: Modulation of angiostatic factor Slit2 by EphA2 receptor regulates endothelial responses to VEGF-mediated angiogenesis and tumor neovascularization.


Subject(s)
Endothelium/blood supply , Intercellular Signaling Peptides and Proteins/metabolism , Mammary Neoplasms, Experimental/blood supply , Neovascularization, Physiologic , Nerve Tissue Proteins/metabolism , Receptor, EphA2/deficiency , Vascular Endothelial Growth Factor A/metabolism , Animals , Cells, Cultured , Coculture Techniques , Culture Media, Conditioned , Endothelium/metabolism , Female , Lung/blood supply , Lung/cytology , Mammary Neoplasms, Experimental/metabolism , Mice , Signal Transduction
8.
J Clin Invest ; 124(5): 2037-49, 2014 May.
Article in English | MEDLINE | ID: mdl-24713656

ABSTRACT

Genome-wide analyses determined previously that the receptor tyrosine kinase (RTK) EPHA2 is commonly overexpressed in non-small cell lung cancers (NSCLCs). EPHA2 overexpression is associated with poor clinical outcomes; therefore, EPHA2 may represent a promising therapeutic target for patients with NSCLC. In support of this hypothesis, here we have shown that targeted disruption of EphA2 in a murine model of aggressive Kras-mutant NSCLC impairs tumor growth. Knockdown of EPHA2 in human NSCLC cell lines reduced cell growth and viability, confirming the epithelial cell autonomous requirements for EPHA2 in NSCLCs. Targeting EPHA2 in NSCLCs decreased S6K1-mediated phosphorylation of cell death agonist BAD and induced apoptosis. Induction of EPHA2 knockdown within established NSCLC tumors in a subcutaneous murine model reduced tumor volume and induced tumor cell death. Furthermore, an ATP-competitive EPHA2 RTK inhibitor, ALW-II-41-27, reduced the number of viable NSCLC cells in a time-dependent and dose-dependent manner in vitro and induced tumor regression in human NSCLC xenografts in vivo. Collectively, these data demonstrate a role for EPHA2 in the maintenance and progression of NSCLCs and provide evidence that ALW-II-41-27 effectively inhibits EPHA2-mediated tumor growth in preclinical models of NSCLC.


Subject(s)
Apoptosis , Carcinoma, Non-Small-Cell Lung/enzymology , Lung Neoplasms/enzymology , Neoplasm Proteins/metabolism , Receptor, EphA2/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Survival , Heterografts , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Knockout , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Transplantation , Protein Kinase Inhibitors/pharmacology , Receptor, EphA2/antagonists & inhibitors , Receptor, EphA2/genetics , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Ribosomal Protein S6 Kinases, 90-kDa/metabolism
9.
Am J Hematol ; 89(2): 187-93, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24136375

ABSTRACT

GTP cyclohydrolase (GCH1) is rate limiting for tetrahydrobiopterin (BH4) synthesis, where BH4 is a cofactor for nitric oxide (NO) synthases and aromatic hydroxylases. GCH1 polymorphisms are implicated in the pathophysiology of pain, but have not been investigated in African populations. We examined GCH1 and pain in sickle cell anemia where GCH1 rs8007267 was a risk factor for pain crises in discovery (n = 228; odds ratio [OR] 2.26; P = 0.009) and replication (n = 513; OR 2.23; P = 0.004) cohorts. In vitro, cells from sickle cell anemia subjects homozygous for the risk allele produced higher BH4. In vivo physiological studies of traits likely to be modulated by GCH1 showed rs8007267 is associated with altered endothelial dependent blood flow in females with SCA (8.42% of variation; P = 0.002). The GCH1 pain association is attributable to an African haplotype with where its sickle cell anemia pain association is limited to females (OR 2.69; 95% CI 1.21-5.94; P = 0.01) and has the opposite directional association described in Europeans independent of global admixture. The presence of a GCH1 haplotype with high BH4 in populations of African ancestry could explain the association of rs8007267 with sickle cell anemia pain crises. The vascular effects of GCH1 and BH4 may also have broader implications for cardiovascular disease in populations of African ancestry.


Subject(s)
Anemia, Sickle Cell/complications , Anemia, Sickle Cell/genetics , GTP Cyclohydrolase/genetics , Genetic Predisposition to Disease , Haplotypes , Pain/etiology , Vascular Diseases/complications , Vascular Diseases/etiology , Adult , Alleles , Anemia, Sickle Cell/metabolism , Biopterins/analogs & derivatives , Biopterins/blood , Biopterins/metabolism , Case-Control Studies , Endothelium/metabolism , Endothelium/physiopathology , Female , Gene Expression Regulation , Gene Frequency , Genetic Association Studies , Genetic Markers , Humans , Male , Middle Aged , Odds Ratio , Pain Management , Phenotype , Plethysmography , Sex Factors , Transcriptome , Young Adult
10.
Methods Mol Biol ; 1027: 139-55, 2013.
Article in English | MEDLINE | ID: mdl-23912985

ABSTRACT

The discovery of novel genetic variants and mutations in lipoprotein and cardiovascular disease research requires DNA sequencing. Large-scale genomics facilities will increasingly accomplish this with a combination of "next-generation" DNA sequencing methodologies. However, laboratories with limited access to these emerging technologies can still support focused genomic studies with the use of automated Sanger sequencing. Here, we describe two robust methods for medium-throughput DNA sequencing from PCR-amplified fragments of genomic DNA.


Subject(s)
Cardiovascular Diseases/genetics , High-Throughput Nucleotide Sequencing/methods , Lipoproteins/genetics , Nucleic Acid Amplification Techniques/methods , Base Sequence , Biomedical Research , Cardiovascular Diseases/pathology , DNA/genetics , Genetic Predisposition to Disease , Humans , Lipoproteins/metabolism , Mutation
11.
Methods Mol Biol ; 1027: 157-79, 2013.
Article in English | MEDLINE | ID: mdl-23912986

ABSTRACT

The identification of new genomic paradigms in lipoprotein and cardiovascular diseases will be accelerated by the application of the recent technological advances in nucleic acid sequencing. Presently, large-scale genomics facilities are equipped to accomplish this objective with a combination of "next-generation" DNA sequencing chemistries, largely focused on assembling massively parallel sequence reads corresponding to complete genes, entire exomes, or whole genomes from populations of individuals. In the future, individual laboratories will also use this emerging technology for focused genomic studies with the use of a combination of next-generation sequencing and automated Sanger sequencing. In particular, -next-generation sequencing will play an increasingly important role when applied to chromatin -immunoprecipitation, RNA transcriptome analysis, and studies of human genetic variation and mutation in carefully phenotyped healthy and disease populations. In this chapter, a brief overview of recent technological advances in next-generation nucleic acid sequencing is presented, with emphasis on practical -application to clinical studies in cardiovascular diseases.


Subject(s)
Cardiovascular Diseases/genetics , High-Throughput Nucleotide Sequencing/methods , Polymorphism, Single Nucleotide/genetics , Biomedical Research , Cardiovascular Diseases/pathology , Exome/genetics , Genome, Human , Humans , Mutation , Transcriptome/genetics
12.
J Clin Invest ; 119(11): 3395-407, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19809159

ABSTRACT

Rhabdomyosarcoma (RMS) is a childhood cancer originating from skeletal muscle, and patient survival is poor in the presence of metastatic disease. Few determinants that regulate metastasis development have been identified. The receptor tyrosine kinase FGFR4 is highly expressed in RMS tissue, suggesting a role in tumorigenesis, although its functional importance has not been defined. Here, we report the identification of mutations in FGFR4 in human RMS tumors that lead to its activation and present evidence that it functions as an oncogene in RMS. Higher FGFR4 expression in RMS tumors was associated with advanced-stage cancer and poor survival, while FGFR4 knockdown in a human RMS cell line reduced tumor growth and experimental lung metastases when the cells were transplanted into mice. Moreover, 6 FGFR4 tyrosine kinase domain mutations were found among 7 of 94 (7.5%) primary human RMS tumors. The mutants K535 and E550 increased autophosphorylation, Stat3 signaling, tumor proliferation, and metastatic potential when expressed in a murine RMS cell line. These mutants also transformed NIH 3T3 cells and led to an enhanced metastatic phenotype. Finally, murine RMS cell lines expressing the K535 and E550 FGFR4 mutants were substantially more susceptible to apoptosis in the presence of a pharmacologic FGFR inhibitor than the control cell lines expressing the empty vector or wild-type FGFR4. Together, our results demonstrate that mutationally activated FGFR4 acts as an oncogene, and these are what we believe to be the first known mutations in a receptor tyrosine kinase in RMS. These findings support the potential therapeutic targeting of FGFR4 in RMS.


Subject(s)
Mutation/genetics , Receptor, Fibroblast Growth Factor, Type 4/genetics , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Rhabdomyosarcoma/physiopathology , Animals , Cell Cycle , Cell Line, Transformed , Cell Line, Tumor , Cell Proliferation , DNA Replication , Disease Models, Animal , Humans , Lung Neoplasms/physiopathology , Lung Neoplasms/secondary , Mice , Models, Molecular , Neoplasm Metastasis , Phosphorylation , Protein Structure, Tertiary , Receptor, Fibroblast Growth Factor, Type 4/chemistry , Rhabdomyosarcoma/mortality , STAT3 Transcription Factor/metabolism , Transplantation, Heterologous
13.
Am J Physiol Heart Circ Physiol ; 293(5): H2971-6, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17766485

ABSTRACT

In clinical studies, sleep apnea is associated with hypertension, oxidative stress, and increased circulating endothelin-1 (ET-1). We previously developed a model of sleep apnea by exposing rats to eucapnic intermittent hypoxia (IH-C) during sleep, which increases both blood pressure and plasma levels of ET-1. Because similar protocols in mice increase tissue and plasma markers of oxidative stress, we hypothesized that IH-C generation of reactive oxygen species (ROS) contributes to the development of ET-1-dependent hypertension in IH-C rats. To test this, male Sprague-Dawley rats were instrumented with indwelling blood pressure telemeters and drank either plain water or water containing the superoxide dismutase mimetic, Tempol (4-hydroxy-2,2,6,6-tetramethyl-piperidine-1-oxyl, 1 mM). Mean arterial pressure (MAP) and heart rate (HR) were recorded for 3 control days and 14 treatment days with rats exposed 7 h/day to IH-C or air/air cycling (Sham). On day 14, MAP in IH-C rats treated with Tempol (107 +/- 2.29 mmHg) was significantly lower than in untreated IH-C rats (118 +/- 9 mmHg, P < 0.05). Tempol did not affect blood pressure in sham-operated rats (Tempol = 101 +/- 3, water = 101 +/- 2 mmHg). Immunoreactive ET-1 was greater in plasma from IH-C rats compared with plasma from sham-operated rats but was not different from Sham in Tempol-treated IH-C rats. Small mesenteric arteries from IH-C rats but not Tempol-treated IH-C rats had increased superoxide levels as measured by ferric cytochrome c reduction, lucigenin signaling, and dihydroethidium fluorescence. The data show that IH-C increases ET-1 production and vascular ROS levels and that scavenging superoxide prevents both. Thus oxidative stress appears to contribute to increases in ET-1 production and elevated arterial pressure in this rat model of sleep apnea-induced hypertension.


Subject(s)
Endothelin-1/metabolism , Hypertension/etiology , Hypertension/metabolism , Reactive Oxygen Species/metabolism , Sleep Apnea Syndromes/complications , Sleep Apnea Syndromes/metabolism , Superoxide Dismutase/metabolism , Animals , Male , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...