Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Am J Physiol Lung Cell Mol Physiol ; 327(1): L65-L78, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38651968

ABSTRACT

Perimenstrual worsening of asthma occurs in up to 40% of women with asthma, leading to increased acute exacerbations requiring clinical care. The role of sex hormones during these times remains unclear. In the current study, we used a translational approach to determine whether progesterone exacerbates allergic inflammation in the traditional chicken egg ovalbumin (OVA) model in BALB/c mice. Simultaneously, we used peripheral blood mononuclear cells (PBMC) from healthy human donors to assess the effects of progesterone on circulating group 2 innate lymphoid cells (ILC2). Briefly, lungs of ovariectomized (OVX) or sham-operated female (F-Sham) controls were implanted with a progesterone (P4, 25 mg) (OVX-P4) or placebo pellet (OVX-Placebo), followed by sensitization and challenge with ovalbumin (OVA). Progesterone increased total inflammatory histologic scores, increased hyper-responsiveness to methacholine (MCh), increased select chemokines in the bronchoalveolar lavage (BAL) and serum, and increased ILC2 and neutrophil numbers, along the airways compared with F-Sham-OVA and OVX-Placebo-OVA animals. Lung ILC2 were sorted from F-Sham-OVA, OVX-Placebo-OVA and OVX-P4-OVA treated animals and stimulated with IL-33. OVX-P4-OVA lung ILC2 were more responsive to interleukin 33 (IL-33) compared with F-Sham-OVA treated, producing more IL-13 and chemokines following IL-33 stimulation. We confirmed the expression of the progesterone receptor (PR) on human ILC2, and showed that P4 + IL-33 stimulation also increased IL-13 and chemokine production from human ILC2. We establish that murine ILC2 are capable of responding to P4 and thereby contribute to allergic inflammation in the lung. We confirmed that human ILC2 are also hyper-responsive to P4 and IL-33 and likely contribute to airway exacerbations following allergen exposures in asthmatic women with increased symptoms around the time of menstruation.NEW & NOTEWORTHY There is a strong association between female biological sex and severe asthma. We investigated the allergic immune response, lung pathology, and airway mechanics in the well-described chicken egg ovalbumin (OVA) model with steady levels of progesterone delivered throughout the treatment period. We found that progesterone enhances the activation of mouse group 2 innate lymphoid cells (ILC2). Human ILC2 are also hyper-responsive to progesterone and interleukin 33 (IL-33), and likely contribute to airway exacerbations following allergen exposures in women with asthma.


Subject(s)
Asthma , Lung , Lymphocytes , Mice, Inbred BALB C , Ovalbumin , Progesterone , Progesterone/pharmacology , Animals , Female , Lymphocytes/immunology , Lymphocytes/metabolism , Humans , Asthma/immunology , Asthma/pathology , Asthma/metabolism , Mice , Ovalbumin/immunology , Lung/pathology , Lung/immunology , Lung/metabolism , Immunity, Innate/drug effects , Interleukin-33/metabolism , Hypersensitivity/immunology , Hypersensitivity/pathology , Hypersensitivity/metabolism , Inflammation/pathology , Inflammation/immunology , Inflammation/metabolism , Disease Models, Animal
2.
PLoS Genet ; 16(6): e1008756, 2020 06.
Article in English | MEDLINE | ID: mdl-32520939

ABSTRACT

Paternal cigarette smoke (CS) exposure is associated with increased risk of behavioral disorders and cancer in offspring, but the mechanism has not been identified. Here we use mouse models to investigate mechanisms and impacts of paternal CS exposure. We demonstrate that CS exposure induces sperm DNAme changes that are partially corrected within 28 days of removal from CS exposure. Additionally, paternal smoking is associated with changes in prefrontal cortex DNAme and gene expression patterns in offspring. Remarkably, the epigenetic and transcriptional effects of CS exposure that we observed in wild type mice are partially recapitulated in Nrf2-/- mice and their offspring, independent of smoking status. Nrf2 is a central regulator of antioxidant gene transcription, and mice lacking Nrf2 consequently display elevated oxidative stress, suggesting that oxidative stress may underlie CS-induced heritable epigenetic changes. Importantly, paternal sperm DNAme changes do not overlap with DNAme changes measured in offspring prefrontal cortex, indicating that the observed DNAme changes in sperm are not directly inherited. Additionally, the changes in sperm DNAme associated with CS exposure were not observed in sperm of unexposed offspring, suggesting the effects are likely not maintained across multiple generations.


Subject(s)
Epigenesis, Genetic , NF-E2-Related Factor 2/genetics , Paternal Exposure , Tobacco Smoke Pollution/adverse effects , Animals , DNA Methylation , Female , Male , Mice , Mice, Inbred C57BL , Prefrontal Cortex/metabolism , Spermatozoa/metabolism
3.
J Neuroinflammation ; 12: 101, 2015 May 23.
Article in English | MEDLINE | ID: mdl-25997851

ABSTRACT

BACKGROUND: Activity of cyclooxygenase 2 (COX-2) in mouse oligodendrocyte precursor cells (OPCs) modulates vulnerability to excitotoxic challenge. The mechanism by which COX-2 renders OPCs more sensitive to excitotoxicity is not known. In the present study, we examined the hypothesis that OPC excitotoxic death is augmented by COX-2-generated prostaglandin E2 (PGE2) acting on specific prostanoid receptors which could contribute to OPC death. METHODS: Dispersed OPC cultures prepared from mice brains were examined for expression of PGE2 receptors and the ability to generate PGE2 following activation of glutamate receptors with kainic acid (KA). OPC death in cultures was induced by either KA, 3'-O-(Benzoyl) benzoyl ATP (BzATP) (which stimulates the purinergic receptor P2X7), or TNFα, and the effects of EP3 receptor agonists and antagonists on OPC viability were examined. RESULTS: Stimulation of OPC cultures with KA resulted in nearly a twofold increase in PGE2. OPCs expressed all four PGE receptors (EP1-EP4) as indicated by immunofluorescence and Western blot analyses; however, EP3 was the most abundantly expressed. The EP3 receptor was identified as a candidate contributing to OPC excitotoxic death based on pharmacological evidence. Treatment of OPCs with an EP1/EP3 agonist 17 phenyl-trinor PGE2 reversed protection from a COX-2 inhibitor while inhibition of EP3 receptor protected OPCs from excitotoxicity. Inhibition with an EP1 antagonist had no effect on OPC excitotoxic death. Moreover, inhibition of EP3 was protective against toxic stimulation with KA, BzATP, or TNFα. CONCLUSION: Therefore, inhibitors of the EP3 receptor appear to enhance survival of OPCs following toxic challenge and may help facilitate remyelination.


Subject(s)
Dinoprostone/metabolism , Oligodendroglia/physiology , Receptors, Prostaglandin E/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/toxicity , Animals , Cell Death , Cells, Cultured , Cyclooxygenase 2/metabolism , Enzyme Inhibitors/pharmacology , Isoxazoles/pharmacology , Kainic Acid/toxicity , Mice , Oligodendroglia/drug effects , Receptors, Glutamate/metabolism , Receptors, IgG/metabolism , Receptors, Prostaglandin E/genetics , Stem Cells , Sulfones/pharmacology , Time Factors
4.
Am J Physiol Lung Cell Mol Physiol ; 296(3): L489-99, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19036873

ABSTRACT

Persistent hypoxia can cause pulmonary arterial hypertension that may be associated with significant remodeling of the pulmonary arteries, including smooth muscle cell proliferation and hypertrophy. We previously demonstrated that the NADPH oxidase homolog NOX4 mediates human pulmonary artery smooth muscle cell (HPASMC) proliferation by transforming growth factor-beta1 (TGF-beta1). We now show that hypoxia increases HPASMC proliferation in vitro, accompanied by increased reactive oxygen species generation and NOX4 gene expression, and is inhibited by antioxidants, the flavoenzyme inhibitor diphenyleneiodonium (DPI), and NOX4 gene silencing. HPASMC proliferation and NOX4 expression are also observed when media from hypoxic HPASMC are added to HPASMC grown in normoxic conditions, suggesting autocrine stimulation. TGF-beta1 and insulin-like growth factor binding protein-3 (IGFBP-3) are both increased in the media of hypoxic HPASMC, and increased IGFBP-3 gene expression is noted in hypoxic HPASMC. Treatment with anti-TGF-beta1 antibody attenuates NOX4 and IGFBP-3 gene expression, accumulation of IGFBP-3 protein in media, and proliferation. Inhibition of IGFBP-3 expression with small interfering RNA (siRNA) decreases NOX4 gene expression and hypoxic proliferation. Conversely, NOX4 silencing does not decrease hypoxic IGFBP-3 gene expression or secreted protein. Smad inhibition does not but the phosphatidylinositol 3-kinase (PI3K) signaling pathway inhibitor LY-294002 does inhibit NOX4 and IGFBP-3 gene expression, IGFBP-3 secretion, and cellular proliferation resulting from hypoxia. Immunoblots from hypoxic HPASMC reveal increased TGF-beta1-mediated phosphorylation of the serine/threonine kinase (Akt), consistent with hypoxia-induced activation of PI3K/Akt signaling pathways to promote proliferation. We conclude that hypoxic HPASMC produce TGF-beta1 that acts in an autocrine fashion to induce IGFBP-3 through PI3K/Akt. IGFBP-3 increases NOX4 gene expression, resulting in HPASMC proliferation. These observations add to our understanding hypoxic pulmonary vascular remodeling.


Subject(s)
Cell Hypoxia/physiology , Insulin-Like Growth Factor Binding Proteins/biosynthesis , Myocytes, Smooth Muscle/metabolism , NADPH Oxidases/metabolism , Pulmonary Artery/metabolism , Transforming Growth Factor beta1/biosynthesis , Autocrine Communication , Cell Hypoxia/genetics , Cell Proliferation , Cells, Cultured , Gene Expression , Humans , Insulin-Like Growth Factor Binding Protein 3 , Insulin-Like Growth Factor Binding Proteins/genetics , Models, Biological , Myocytes, Smooth Muscle/cytology , NADPH Oxidase 4 , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Artery/cytology , RNA Interference , Reactive Oxygen Species/metabolism , Signal Transduction
5.
Am J Physiol Lung Cell Mol Physiol ; 292(6): L1543-55, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17369289

ABSTRACT

Transforming growth factor-beta1 (TGF-beta1) plays a pivotal role in increasing airway smooth muscle mass in severe asthma by inducing proliferation and hypertrophy of human airway smooth muscle. The mechanism(s) for these effects of TGF-beta1 have not been fully elucidated. In this study, we demonstrate that TGF-beta1 is a potent inducer of expression of the nonphagocyte NAD(P)H oxidase catalytic homolog Nox4, diphenylene iodonium-inhibitable reactive oxygen species production, proliferation, and hypertrophy in cultured human airway smooth muscle cells. By confocal microscopy, TGF-beta1-induced Nox4 was localized with the endoplasmic reticulum and the nucleus, implying a role for Nox4 in regulation of both the cell cycle and protein synthesis. Consistent with this hypothesis, TGF-beta1 increased retinoblastoma protein phosphorylation at both Ser807/811 and Ser780. Silencing Nox4 prevented TGF-beta1-mediated retinoblastoma protein phosphorylation, proliferation, and cell hypertrophy. TGF-beta1 also increased phosphorylation of eukaryotic translation initiation factor 4E binding protein-1 at Thr37/46, and this was likewise blocked by silencing Nox4. This is the first report to suggest a functional role for Nox4 in cell cycle transition and to demonstrate that Nox4 influences the pathobiochemistry of asthma by generating reactive oxygen species that promote TGF-beta1-induced proliferation and hypertrophy of human airway smooth muscle.


Subject(s)
Asthma/metabolism , Bronchi/cytology , Myocytes, Smooth Muscle/enzymology , NADPH Oxidases/metabolism , Retinoblastoma Protein/metabolism , Transforming Growth Factor beta1/metabolism , Activins/metabolism , Activins/pharmacology , Asthma/pathology , CDC2 Protein Kinase/metabolism , Cell Division/drug effects , Cell Division/physiology , Cell Nucleus/enzymology , Cells, Cultured , Eukaryotic Initiation Factor-4E/metabolism , Humans , Hypertrophy , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , NADPH Oxidase 4 , NADPH Oxidases/genetics , Oxidation-Reduction , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Biosynthesis/physiology , Proto-Oncogene Proteins c-myc/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Smad3 Protein/metabolism , Transfection , Transforming Growth Factor beta1/pharmacology
6.
Am J Physiol Lung Cell Mol Physiol ; 290(4): L661-L673, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16227320

ABSTRACT

Transforming growth factor-beta1 (TGF-beta1) is abundantly expressed in pulmonary hypertension, but its effect on the pulmonary circulation remains unsettled. We studied the consequences of TGF-beta1 stimulation on freshly isolated human pulmonary artery smooth muscle cells (HPASMC). TGF-beta1 initially promoted differentiation, with upregulated expression of smooth muscle contractile proteins. TGF-beta1 also induced expression of Nox4, the only NAD(P)H oxidase membrane homolog found in HPASMC, through a signaling pathway involving Smad 2/3 but not mitogen-activated protein (MAP) kinases. TGF-beta1 likewise increased production of reactive oxygen species (ROS), an effect significantly reduced by the NAD(P)H oxidase flavoprotein inhibitor diphenylene iodonium (DPI) and by Nox4 siRNAs. In the absence of TGF-beta1, Nox4 was present in freshly cultured cells but progressively lost with each passage in culture, paralleling a decrease in ROS production by HPASMC over time. At a later time point (72 h), TGF-beta1 promoted HPASMC proliferation in a manner partially inhibited by Nox4 small interfering RNA and dominant negative Smad 2/3, indicating that TGF-beta1 stimulates HPASMC growth in part by a redox-dependent mechanism mediated through induction of Nox4. HPASMC activation of the MAP kinases ERK1/2 was reduced by the NAD(P)H oxidase inhibitors DPI and 4-(2-aminoethyl)benzenesulfonyl fluoride, suggesting that TGF-beta1 may facilitate proliferation by upregulating Nox4 and ROS production, with transient oxidative inactivation of phosphatases and augmentation of growth signaling cascades. These findings suggest that Nox4 is the relevant Nox homolog in HPASMC. This is the first observation that TGF-beta1 regulates Nox4, with important implications for mechanisms of pulmonary vascular remodeling.


Subject(s)
Myocytes, Smooth Muscle/cytology , NADPH Oxidases/metabolism , Pulmonary Artery/cytology , Reactive Oxygen Species/metabolism , Transforming Growth Factor beta/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Contractile Proteins/metabolism , Humans , Mitogen-Activated Protein Kinases/metabolism , NADPH Oxidase 4 , Pulmonary Artery/metabolism , Signal Transduction/physiology , Smad Proteins/metabolism , Transforming Growth Factor beta1
7.
Am J Physiol Lung Cell Mol Physiol ; 282(4): L782-95, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11880305

ABSTRACT

Evidence is rapidly accumulating that low-activity-reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidases homologous to that in phagocytic cells generate reactive oxygen species as signaling intermediates in both endothelium and vascular smooth muscle. We therefore explored the possibility of such an oxidase regulating growth of airway smooth muscle (AWSM). Proliferation of human AWSM cells in culture was inhibited by the antioxidants catalase and N-acetylcysteine, and by the flavoprotein inhibitor diphenylene iodonium (DPI). Membranes prepared from human AWSM cells generated superoxide anion (O) measured by superoxide dismutase-inhibitable lucigenin chemiluminescence, with a distinct preference for NADPH instead of reduced nicotinamide adenine dinucleotide as substrate. Chemiluminescence was also inhibited by DPI, suggesting the presence of a flavoprotein containing oxidase generating O as a signaling molecule for cell growth. Examination of human AWSM cells by reverse transcriptase-polymerase chain reaction consistently demonstrated transcripts with sequences identical to those reported for p22(phox). Transfection with p22(phox) antisense oligonucleotides reduced human AWSM proliferation. Inhibition of NADPH oxidase activity with DPI prevented serum-induced activation of nuclear factor-kappaB (NF-kappaB), and overexpression of a superrepressor form of the NF-kappaB inhibitor IkappaBalpha significantly reduced human AWSM growth. These findings suggest that an NADPH oxidase containing p22(phox) regulates growth-factor responsive human AWSM proliferation, and that the oxidase signals in part through activation of the prototypical redox-regulated transcription factor NF-kappaB.


Subject(s)
Bronchi/cytology , Bronchi/enzymology , Membrane Transport Proteins , Muscle, Smooth/cytology , Muscle, Smooth/enzymology , NADPH Oxidases/metabolism , NF-kappa B/metabolism , Antioxidants/metabolism , Cell Division/physiology , Cell Membrane/metabolism , Cells, Cultured , Enzyme Inhibitors/pharmacology , Flavoproteins/antagonists & inhibitors , Gene Expression Regulation, Enzymologic , Humans , NADPH Dehydrogenase/genetics , NADPH Dehydrogenase/metabolism , NADPH Oxidases/genetics , Onium Compounds/pharmacology , Phosphoproteins/genetics , Phosphoproteins/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , Superoxides/metabolism
8.
Am J Physiol Cell Physiol ; 282(6): C1212-24, 2002 Jun.
Article in English | MEDLINE | ID: mdl-11997235

ABSTRACT

Malignant melanoma cells spontaneously generate reactive oxygen species (ROS) that promote constitutive activation of the transcription factor nuclear factor-kappaB (NF-kappaB). Although antioxidants and inhibitors of NAD(P)H oxidases significantly reduce constitutive NF-kappaB activation and suppress cell proliferation (11), the nature of the enzyme responsible for ROS production in melanoma cells has not been determined. To address this issue, we now have characterized the source of ROS production in melanoma cells. We report that ROS are generated by isolated, cytosol-free melanoma plasma membranes, with inhibition by NAD(P)H oxidase inhibitors. The p22(phox), gp91(phox), and p67(phox) components of the human phagocyte NAD(P)H oxidase and the gp91(phox) homolog NOX4 were demonstrated in melanomas by RT-PCR and sequencing, and protein product for both p22(phox) and gp91(phox) was detected in cell membranes by immunoassay. Normal human epidermal melanocytes expressed only p22(phox) and NOX4. Melanoma proliferation was reduced by NAD(P)H oxidase inhibitors and by transfection of antisense but not sense oligonucleotides for p22(phox) and NOX4. Also, the flavoprotein inhibitor diphenylene iodonium inhibited constitutive DNA binding of nuclear protein to the NF-kappaB and cAMP-response element consensus oligonucleotides, without affecting DNA binding activity to activator protein-1 or OCT-1. This suggests that ROS generated in autocrine fashion by an NAD(P)H oxidase may play a role in signaling malignant melanoma growth.


Subject(s)
Melanoma, Experimental/metabolism , Membrane Transport Proteins , NADH, NADPH Oxidoreductases/metabolism , Cell Division/drug effects , Cell Line , Cell Membrane/metabolism , Enzyme Inhibitors/pharmacology , Humans , Intracellular Fluid/metabolism , Melanocytes/cytology , Melanocytes/metabolism , Melanoma, Experimental/pathology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , NADH, NADPH Oxidoreductases/antagonists & inhibitors , NADH, NADPH Oxidoreductases/genetics , NADPH Dehydrogenase/antagonists & inhibitors , NADPH Dehydrogenase/genetics , NADPH Dehydrogenase/metabolism , NADPH Oxidase 2 , NADPH Oxidase 4 , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/genetics , NF-kappa B/antagonists & inhibitors , NF-kappa B/biosynthesis , Onium Compounds/pharmacology , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/genetics , Phosphoproteins/metabolism , Polymerase Chain Reaction , Protein Subunits , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Response Elements/physiology , Transcription Factors/metabolism , Transcription, Genetic
9.
Am J Physiol Cell Physiol ; 285(2): C353-69, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12686516

ABSTRACT

Reactive oxygen species (ROS) appear to play an important role in regulating growth and survival of prostate cancer. However, the sources for ROS production in prostate cancer cells have not been determined. We report that ROS are generated by intact American Type Culture Collection DU 145 cells and by their membranes through a mechanism blocked by NAD(P)H oxidase inhibitors. ROS are critical for growth in these cells, because NAD(P)H oxidase inhibitors and antioxidants blocked proliferation. Components of the human phagocyte NAD(P)H oxidase, p22phox and gp91phox, as well as the Ca2+ concentration-responsive gp91phox homolog NOX5 were demonstrated in DU 145 cells by RT-PCR and sequencing. Although the protein product for p22phox was not detectable, both gp91phox and NOX5 were identified throughout the cell by immunostaining and confocal microscopy and NOX5 immunostaining was enhanced in a perinuclear location, corresponding to enhanced ROS production adjacent to the nuclear membrane imaged by 2',7'-dichlorofluorescin diacetate oxidation. The calcium ionophore ionomycin dramatically stimulated ferricytochrome c reduction in cell media, further supporting the importance of NOX5 for ROS production. Antisense oligonucleotides for NOX5 inhibited ROS production and cell proliferation in DU 145 cells. In contrast, antisense oligonucleotides to p22phox or gp91phox did not impair cell growth. Inhibition of ROS generation with antioxidants or NAD(P)H oxidase inhibitors increased apoptosis in cells. These results indicate that ROS generated by the newly described NOX5 oxidase are essential for prostate cancer growth, possibly by providing trophic intracellular oxidant tone that retards programmed cell death.


Subject(s)
Apoptosis/genetics , Carcinoma/enzymology , Cell Division/genetics , Membrane Proteins/metabolism , NADPH Oxidases/metabolism , Prostatic Neoplasms/enzymology , Reactive Oxygen Species/metabolism , Antioxidants/pharmacology , Apoptosis/drug effects , Cell Division/drug effects , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , Cytochrome c Group/drug effects , Cytochrome c Group/metabolism , Enzyme Inhibitors/pharmacology , Humans , Ionophores/pharmacology , Male , Membrane Glycoproteins/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , NADPH Oxidase 2 , NADPH Oxidase 5 , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/genetics , Nuclear Envelope/metabolism , Oligoribonucleotides, Antisense/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/physiology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL