301
|
Woo DK, Poyton RO. The absence of a mitochondrial genome in rho0 yeast cells extends lifespan independently of retrograde regulation. Exp Gerontol 2009; 44:390-7. [PMID: 19285548 PMCID: PMC3341797 DOI: 10.1016/j.exger.2009.03.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2008] [Revised: 02/26/2009] [Accepted: 03/03/2009] [Indexed: 10/21/2022]
Abstract
The absence of mtDNA in rho0 yeast cells affects both respiration and mitochondrial-nuclear communication (e.g., retrograde regulation, intergenomic signaling, or pleiotropic drug resistance). Previously, it has been reported that some rho0 strains have increased replicative lifespans, attributable to the lack of respiration and retrograde regulation. Here, we have been able to confirm that rho0 cells exhibit increased replicative lifespans but have found that this is not associated with the lack of respiration or reduced oxidative stress but instead, is related to the lack of mtDNA per se in rho0 cells. Also, we find no correlation between the strength of retrograde regulation and lifespan. Furthermore, we find that pdr3- or rtg2- mutations are not responsible for lifespan extension in rho0 cells, ruling out a specific role for PDR3-pleiotropic drug resistance or RGT2-retrograde regulation pathways in the extended lifespans of rho0 cells. Surprisingly, Rtg3p, which acts downstream of Rtg2p, is required for lifespan increase in rho0 cells. Together, these findings indicate that the loss of mtDNA per se and not the lack of respiration lead to extended longevity in rho0 cells. They also suggest that Rtg3p, acting independently of retrograde regulation, mediates this effect, possibly via intergenomic signaling.
Collapse
Affiliation(s)
- Dong Kyun Woo
- The Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| | | |
Collapse
|
302
|
Balligand JL, Feron O, Dessy C. eNOS activation by physical forces: from short-term regulation of contraction to chronic remodeling of cardiovascular tissues. Physiol Rev 2009; 89:481-534. [PMID: 19342613 DOI: 10.1152/physrev.00042.2007] [Citation(s) in RCA: 327] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Nitric oxide production in response to flow-dependent shear forces applied on the surface of endothelial cells is a fundamental mechanism of regulation of vascular tone, peripheral resistance, and tissue perfusion. This implicates the concerted action of multiple upstream "mechanosensing" molecules reversibly assembled in signalosomes recruiting endothelial nitric oxide synthase (eNOS) in specific subcellular locales, e.g., plasmalemmal caveolae. Subsequent short- and long-term increases in activity and expression of eNOS translate this mechanical stimulus into enhanced NO production and bioactivity through a complex transcriptional and posttranslational regulation of the enzyme, including by shear-stress responsive transcription factors, oxidant stress-dependent regulation of transcript stability, eNOS regulatory phosphorylations, and protein-protein interactions. Notably, eNOS expressed in cardiac myocytes is amenable to a similar regulation in response to stretching of cardiac muscle cells and in part mediates the length-dependent increase in cardiac contraction force. In addition to short-term regulation of contractile tone, eNOS mediates key aspects of cardiac and vascular remodeling, e.g., by orchestrating the mobilization, recruitment, migration, and differentiation of cardiac and vascular progenitor cells, in part by regulating the stabilization and transcriptional activity of hypoxia inducible factor in normoxia and hypoxia. The continuum of the influence of eNOS in cardiovascular biology explains its growing implication in mechanosensitive aspects of integrated physiology, such as the control of blood pressure variability or the modulation of cardiac remodeling in situations of hemodynamic overload.
Collapse
Affiliation(s)
- J-L Balligand
- Unit of Pharmacology and Therapeutics, Université catholique de Louvain, Brussels, Belgium.
| | | | | |
Collapse
|
303
|
Illi B, Colussi C, Grasselli A, Farsetti A, Capogrossi MC, Gaetano C. NO sparks off chromatin: tales of a multifaceted epigenetic regulator. Pharmacol Ther 2009; 123:344-52. [PMID: 19464317 DOI: 10.1016/j.pharmthera.2009.05.003] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2009] [Accepted: 05/11/2009] [Indexed: 10/20/2022]
Abstract
The discovery of nitric oxide (NO) revealed its ambiguous nature, which is related to its pleiotropic activities that control the homeostasis of every organism from bacteria to mammals in several physiological and pathological situations. The wide range of action of NO basically depends on two features: 1) the variety of chemical reactions depending on NO, and 2) the differential cellular responses elicited by distinct NO concentrations. Despite the increasing body of knowledge regarding its chemistry, biology and NO-dependent signaling pathways, little information is available on the nuclear actions of NO in terms of gene expression regulation. Indeed, studies of a putative role for this diatomic compound in regulating chromatin remodeling are still in their infancy. Only recently has the role of NO in epigenetics emerged, and some of its putative epigenetic properties are still only hypothetical. In the present review, we discuss the current evidence for NO-related mechanisms of epigenetic gene expression regulation. We link some of the well known NO chemical reactions and metabolic processes (e.g., S-nitrosylation of thiols, tyrosine nitration, cGMP production) to chromatin modification and address the most recent, striking hypothesis about NO and the control of chromosomes structure.
Collapse
Affiliation(s)
- Barbara Illi
- Laboratorio di Biologia Vascolare e Medicina Rigenerativa, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | | | | | | | | | | |
Collapse
|
304
|
Bischoff P, Altmeyer A, Dumont F. Radiosensitising agents for the radiotherapy of cancer: advances in traditional and hypoxia targeted radiosensitisers. Expert Opin Ther Pat 2009; 19:643-62. [DOI: 10.1517/13543770902824172] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
305
|
Sonveaux P, Jordan BF, Gallez B, Feron O. Nitric oxide delivery to cancer: Why and how? Eur J Cancer 2009; 45:1352-69. [DOI: 10.1016/j.ejca.2008.12.018] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2008] [Accepted: 12/12/2008] [Indexed: 02/07/2023]
|
306
|
Abstract
Nitric oxide is well established as a major signaling molecule. Evidence is accumulating that carbon monoxide and hydrogen sulfide also are physiologic mediators in the cardiovascular, immune, and nervous systems. This Review focuses on mechanisms whereby they signal by binding to metal centers in metalloproteins, such as in guanylyl cyclase, or modifying sulfhydryl groups in protein targets.
Collapse
Affiliation(s)
- Asif K. Mustafa
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Moataz M. Gadalla
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Solomon H. Snyder
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
307
|
Lima B, Lam GKW, Xie L, Diesen DL, Villamizar N, Nienaber J, Messina E, Bowles D, Kontos CD, Hare JM, Stamler JS, Rockman HA. Endogenous S-nitrosothiols protect against myocardial injury. Proc Natl Acad Sci U S A 2009; 106:6297-302. [PMID: 19325130 PMCID: PMC2669330 DOI: 10.1073/pnas.0901043106] [Citation(s) in RCA: 195] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2009] [Indexed: 11/18/2022] Open
Abstract
Despite substantial evidence that nitric oxide (NO) and/or endogenous S-nitrosothiols (SNOs) exert protective effects in a variety of cardiovascular diseases, the molecular details are largely unknown. Here we show that following left coronary artery ligation, mice with a targeted deletion of the S-nitrosoglutathione reductase gene (GSNOR(-/-)) have reduced myocardial infarct size, preserved ventricular systolic and diastolic function, and maintained tissue oxygenation. These profound physiological effects are associated with increases in myocardial capillary density and S-nitrosylation of the transcription factor hypoxia inducible factor-1alpha (HIF-1alpha) under normoxic conditions. We further show that S-nitrosylated HIF-1alpha binds to the vascular endothelial growth factor (VEGF) gene, thus identifying a role for GSNO in angiogenesis and myocardial protection. These results suggest innovative approaches to modulate angiogenesis and preserve cardiac function.
Collapse
Affiliation(s)
| | | | - Liang Xie
- Medicine, Duke University Medical Center, Durham, NC 27710; and
| | - Diana L. Diesen
- Medicine, Duke University Medical Center, Durham, NC 27710; and
| | | | | | | | | | | | - Joshua M. Hare
- Division of Cardiology, University of Miami Miller School of Medicine, Miami, FL 33136
| | | | | |
Collapse
|
308
|
Ahn GO, Brown JM. Influence of bone marrow-derived hematopoietic cells on the tumor response to radiotherapy: experimental models and clinical perspectives. Cell Cycle 2009; 8:970-6. [PMID: 19270527 PMCID: PMC2862685 DOI: 10.4161/cc.8.7.8075] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
In this review, we highlight some of recent studies underscoring the importance of the tumor microenvironment, especially the role of bone marrow-derived myeloid cells, in restoring tumor growth after irradiation. Myeloid cells are hematopoietic cells that give rise to monocytes and macrophages in the peripheral blood and tissues. These cells have been shown to be proangiogenic in tumors promoting tumor growth. We also discuss our previously unpublished results on the effect of irradiation on the tumor vasculature including pericyte and basement membrane coverage to the endothelium of tumor blood vessels. We summarize the clinical significance of these studies including the use of MMP-9 inhibitors, administering white blood cell boosters, or planning safety margin of tumor volumes, in order to improve overall clinical benefits in cancer patients treated with radiotherapy.
Collapse
Affiliation(s)
- G-One Ahn
- Division of Radiation and Cancer Biology; Department of Radiation Oncology; Stanford, California USA
| | - J. Martin Brown
- Division of Radiation and Cancer Biology; Department of Radiation Oncology; Stanford, California USA
| |
Collapse
|
309
|
Chen FH, Chiang CS, Wang CC, Tsai CS, Jung SM, Lee CC, McBride WH, Hong JH. Radiotherapy decreases vascular density and causes hypoxia with macrophage aggregation in TRAMP-C1 prostate tumors. Clin Cancer Res 2009; 15:1721-9. [PMID: 19240176 PMCID: PMC2868361 DOI: 10.1158/1078-0432.ccr-08-1471] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To investigate how single or fractionated doses of radiation change the microenvironment in transgenic adenocarcinoma of the mouse prostate (TRAMP)-C1 tumors with respect to vascularity, hypoxia, and macrophage infiltrates. EXPERIMENTAL DESIGN Murine prostate TRAMP-C1 tumors were grown in C57BL/6J mice to 4 mm tumor diameter and were irradiated with either 25 Gy in a single dose or 60 Gy in 15 fractions. Changes in vascularity, hypoxia, and macrophage infiltrates were assessed by immunohistochemistry and molecular assays. RESULTS Tumor growth was delayed for 1 week after both radiation schedules. Tumor microvascular density (MVD) progressively decreased over a 3-week period to nadirs of 25% and 40% of unirradiated tumors for single or fractionated treatment, respectively. In accord with the decrease in MVDs, mRNA levels of endothelial markers, such as CD31, endoglin, and TIE, decreased over the same time period after irradiation. Central dilated vessels developed surrounded by avascularized hypoxic regions that became infiltrated with aggregates of CD68+ tumor-associated macrophages, reaching a maximum at 3 weeks after irradiation. Necrotic regions decreased and were more dispersed. CONCLUSION Irradiation of TRAMP-C1 tumors with either single or fractionated doses decreases MVD, leading to the development of disperse chronic hypoxic regions, which are infiltrated with CD68+ tumor-associated macrophages. Approaches to interfere in the development of these effects are promising strategies to enhance the efficacy of cancer radiotherapy.
Collapse
MESH Headings
- Adenocarcinoma/blood supply
- Adenocarcinoma/metabolism
- Adenocarcinoma/pathology
- Animals
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/metabolism
- Cell Hypoxia/radiation effects
- Disease Models, Animal
- Humans
- Immunoenzyme Techniques
- Macrophages/pathology
- Macrophages/radiation effects
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Microcirculation
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/radiotherapy
- Prostatic Neoplasms/blood supply
- Prostatic Neoplasms/metabolism
- Prostatic Neoplasms/pathology
- RNA, Neoplasm/genetics
- RNA, Neoplasm/metabolism
- Radiation Dosage
- Ribonucleases/metabolism
Collapse
Affiliation(s)
- Fang-Hsin Chen
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
- Department of Radiation Oncology, Chang Gung Memorial Hospital-LinKou, Taiwan
| | - Chi-Shiun Chiang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Chun-Chieh Wang
- Department of Radiation Oncology, Chang Gung Memorial Hospital-LinKou, Taiwan
- Department of Medical Imaging and Radiological Science, Chang Gung University, Taoyuan, Taiwan
| | - Chien-Sheng Tsai
- Department of Radiation Oncology, Chang Gung Memorial Hospital-LinKou, Taiwan
- Department of Medical Imaging and Radiological Science, Chang Gung University, Taoyuan, Taiwan
| | - Shih-Ming Jung
- Department of Pathology, Chang Gung Memorial Hospital-LinKou, Taiwan
| | - Chung-Chi Lee
- Department of Medical Imaging and Radiological Science, Chang Gung University, Taoyuan, Taiwan
| | - William H. McBride
- Roy E. Coats Research Laboratories, Department of Radiation Oncology, University of California at Los Angeles, Los Angeles, California
| | - Ji-Hong Hong
- Department of Radiation Oncology, Chang Gung Memorial Hospital-LinKou, Taiwan
- Department of Medical Imaging and Radiological Science, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
310
|
Hypoxia-inducible factor 1 and related gene products in anaesthetic-induced preconditioning. Eur J Anaesthesiol 2009; 26:201-6. [DOI: 10.1097/eja.0b013e3283212cbb] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
311
|
Woo DK, Phang TL, Trawick JD, Poyton RO. Multiple pathways of mitochondrial-nuclear communication in yeast: Intergenomic signaling involves ABF1 and affects a different set of genes than retrograde regulation. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2009; 1789:135-45. [DOI: 10.1016/j.bbagrm.2008.09.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2008] [Revised: 09/14/2008] [Accepted: 09/23/2008] [Indexed: 10/21/2022]
|
312
|
WARDMAN P. The importance of radiation chemistry to radiation and free radical biology (The 2008 Silvanus Thompson Memorial Lecture). Br J Radiol 2009; 82:89-104. [DOI: 10.1259/bjr/60186130] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
|
313
|
Forrester MT, Foster MW, Benhar M, Stamler JS. Detection of protein S-nitrosylation with the biotin-switch technique. Free Radic Biol Med 2009; 46:119-26. [PMID: 18977293 PMCID: PMC3120222 DOI: 10.1016/j.freeradbiomed.2008.09.034] [Citation(s) in RCA: 257] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2008] [Revised: 09/18/2008] [Accepted: 09/25/2008] [Indexed: 01/19/2023]
Abstract
Protein S-nitrosylation, the posttranslational modification of cysteine thiols to form S-nitrosothiols, is a principle mechanism of nitric oxide-based signaling. Studies have demonstrated myriad roles for S-nitrosylation in organisms from bacteria to humans, and recent efforts have greatly advanced our scientific understanding of how this redox-based modification is dynamically regulated during physiological and pathophysiological conditions. The focus of this review is the biotin-switch technique (BST), which has become a mainstay assay for detecting S-nitrosylated proteins in complex biological systems. Potential pitfalls and modern adaptations of the BST are discussed, as are future directions for this assay in the burgeoning field of protein S-nitrosylation.
Collapse
Affiliation(s)
- Michael T. Forrester
- Department of Biochemistry, Duke University Medical Center, Durham, North Carolina, 27710
- Department of Medical Scientist Training Program, Duke University Medical Center, Durham, North Carolina, 27710
| | - Matthew W. Foster
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, 27710
| | - Moran Benhar
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, 27710
| | - Jonathan S. Stamler
- Department of Biochemistry, Duke University Medical Center, Durham, North Carolina, 27710
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, 27710
| |
Collapse
|
314
|
In vivo bioluminescence imaging monitoring of hypoxia-inducible factor 1alpha, a promoter that protects cells, in response to chemotherapy. AJR Am J Roentgenol 2009; 191:1779-84. [PMID: 19020250 DOI: 10.2214/ajr.07.4060] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Bioluminescence imaging is a powerful technique that has shown that hypoxia-inducible factor 1 (HIF-1), a transcription factor that protects tumor cells from hypoxia, is up-regulated in tumors after radiation therapy. We tested the hypothesis that bioluminescence imaging would successfully and noninvasively depict an increase in HIF-1 in the novel therapeutic environment of chemotherapy and that, as in radiation therapy, the underlying mechanism involves inducible nitric oxide synthase originating in macrophages. Active HIF-1 consists of alpha and beta subunits that bind to promoter sequences in many genes, including those that protect endothelial cells, promote angiogenesis, and alter metastasis and tumor cell metabolism. MATERIALS AND METHODS We grew 4T1 murine breast carcinoma cells with an HIF-1alpha luciferase reporter construct to 7 mm in the right rear flanks of 18 Balb-C mice. The mice were evenly randomized to receive one of the following single intraperitoneal doses: maximum tolerated dose cyclophosphamide (231.5 mg/kg), maximum tolerated dose paclitaxel (10 mg/kg), or control saline solution. Immunohistochemical analysis of tumor sections from the cyclophosphamide and control groups was performed 10 days after treatment to assess the intensity and distribution of HIF-1alpha expression, hypoxia, macrophage infiltration, and expression of macrophage-derived inducible nitric oxide synthase in tumor tissues treated with maximum tolerated dose cyclophosphamide compared with control tumors. RESULTS Cyclophosphamide, but not paclitaxel, significantly inhibited tumor growth and caused a significant increase in HIF-1alpha protein levels, which peaked at a 10-fold increase from baseline on day 10 after administration. In contrast, paclitaxel did not have an antitumor effect in this model and did not cause a significant increase in HIF-1alpha. Immunohistochemical analysis showed increased and more evenly dispersed levels of HIF-1alpha protein, macrophage infiltration, and expression of inducible nitric oxide synthase originating in macrophages after cyclophosphamide treatment. CONCLUSION We successfully monitored increased expression of a tumor protective protein in a noninvasive manner. Such monitoring may be a means of detection of resistance to therapy, and it may be possible to use the monitoring findings to alter treatment strategies in real time. The tumor microenvironment seen at immunohistochemical analysis supports the hypothesized mechanism that the cytotoxic effects of radiation therapy that attract macrophages, causing the release of macrophage-derived inducible nitric oxide synthase and production of HIF-1alpha under aerobic conditions, also underlie chemotherapy. Such noninvasive imaging may be a means to development of therapeutic strategies that prevent HIF-1 up-regulation after chemotherapy treatments.
Collapse
|
315
|
Kim H, Kasper AC, Moon EJ, Park Y, Wooten CM, Dewhirst MW, Hong J. Nucleophilic addition of organozinc reagents to 2-sulfonyl cyclic ethers: stereoselective synthesis of manassantins A and B. Org Lett 2009; 11:89-92. [PMID: 19111058 PMCID: PMC2656112 DOI: 10.1021/ol8024617] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
A convergent route to the synthesis of manassantins A and B, potent inhibitors of HIF-1, is described. Central to the synthesis is a stereoselective addition of an organozinc reagent to a 2-benzenesulfonyl cyclic ether to achieve the 2,3-cis-3,4-trans-4,5-cis-tetrahydrofuran of the natural products. Preliminary structure-activity relationships suggested that the (R)-configuration at C-7 and C-7''' is not critical for HIF-1 inhibition. In addition, the hydroxyl group at C-7 and C-7''' can be replaced with a carbonyl group without loss of activity.
Collapse
Affiliation(s)
- Hyoungsu Kim
- Department of Chemistry, Duke University, Durham, North Carolina 27708, USA
| | | | | | | | | | | | | |
Collapse
|
316
|
Martinive P, Defresne F, Quaghebeur E, Daneau G, Crokart N, Grégoire V, Gallez B, Dessy C, Feron O. Impact of cyclic hypoxia on HIF-1α regulation in endothelial cells - new insights for anti-tumor treatments. FEBS J 2008; 276:509-18. [DOI: 10.1111/j.1742-4658.2008.06798.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
317
|
Santhanam L, Gucek M, Brown TR, Mansharamani M, Ryoo S, Lemmon CA, Romer L, Shoukas AA, Berkowitz DE, Cole RN. Selective fluorescent labeling of S-nitrosothiols (S-FLOS): a novel method for studying S-nitrosation. Nitric Oxide 2008; 19:295-302. [PMID: 18706513 PMCID: PMC3705760 DOI: 10.1016/j.niox.2008.07.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2007] [Revised: 07/08/2008] [Accepted: 07/08/2008] [Indexed: 11/21/2022]
Abstract
Protein S-nitrosation is a reversible post-translation modification critical for redox-sensitive cell signaling that is typically studied using the Biotin Switch method. This method and subsequent modifications usually require avidin binding or Western blot analysis to detect biotin labeled proteins. We describe here a modification of the Biotin Switch assay that eliminates the need for Western blot or avidin enrichment protocols and allows direct comparison of the S-nitrosation state proteins from two different samples in the same gel lane or on the same 2D gel. This S-FLOS method offers detection, identification and quantification of S-nitrosated proteins, with the potential for site-specific identification of nitrosation events.
Collapse
Affiliation(s)
- Lakshmi Santhanam
- The Johns Hopkins School of Medicine, Anesthesiology and CCM, 720 Rutland Avenue, Traylor 621, Baltimore, MD 21205, USA
| | - Marjan Gucek
- The Johns Hopkins School of Medicine, Mass Spectrometry and Proteomics Facility, IBBS, 733 Broadway St., BRB 371, Baltimore, MD 21205, USA
| | - Tashalee R. Brown
- The Johns Hopkins School of Medicine, Anesthesiology and CCM, 720 Rutland Avenue, Traylor 621, Baltimore, MD 21205, USA
| | - Malini Mansharamani
- The Johns Hopkins School of Medicine, Mass Spectrometry and Proteomics Facility, IBBS, 733 Broadway St., BRB 371, Baltimore, MD 21205, USA
| | - Sungwoo Ryoo
- The Johns Hopkins School of Medicine, Anesthesiology and CCM, 720 Rutland Avenue, Traylor 621, Baltimore, MD 21205, USA
| | - Christopher A. Lemmon
- The Johns Hopkins School of Medicine, Anesthesiology and CCM, 720 Rutland Avenue, Traylor 621, Baltimore, MD 21205, USA
| | - Lewis Romer
- The Johns Hopkins School of Medicine, Anesthesiology and CCM, 720 Rutland Avenue, Traylor 621, Baltimore, MD 21205, USA
| | - Artin A. Shoukas
- The Johns Hopkins School of Medicine, Anesthesiology and CCM, 720 Rutland Avenue, Traylor 621, Baltimore, MD 21205, USA
| | - Dan E. Berkowitz
- The Johns Hopkins School of Medicine, Anesthesiology and CCM, 720 Rutland Avenue, Traylor 621, Baltimore, MD 21205, USA
| | - Robert N. Cole
- The Johns Hopkins School of Medicine, Mass Spectrometry and Proteomics Facility, IBBS, 733 Broadway St., BRB 371, Baltimore, MD 21205, USA
| |
Collapse
|
318
|
Wink DA, Ridnour LA, Hussain SP, Harris CC. The reemergence of nitric oxide and cancer. Nitric Oxide 2008; 19:65-7. [PMID: 18638716 PMCID: PMC2565861 DOI: 10.1016/j.niox.2008.05.003] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2008] [Accepted: 05/16/2008] [Indexed: 01/19/2023]
Affiliation(s)
- David A Wink
- Radiation Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | |
Collapse
|
319
|
Ridnour LA, Thomas DD, Switzer C, Flores-Santana W, Isenberg JS, Ambs S, Roberts DD, Wink DA. Molecular mechanisms for discrete nitric oxide levels in cancer. Nitric Oxide 2008; 19:73-6. [PMID: 18472020 PMCID: PMC2574989 DOI: 10.1016/j.niox.2008.04.006] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2008] [Revised: 04/12/2008] [Accepted: 04/14/2008] [Indexed: 01/02/2023]
Abstract
Nitric oxide (NO) has been invoked in nearly every normal and pathological condition associated with human physiology. In tumor biology, nitrogen oxides have both positive and negative affects as they have been implicated in both promoting and preventing cancer. Our work has focused on NO chemistry and how it correlates with cytotoxicity and cancer. Toward this end, we have studied both concentration- and time-dependent NO regulation of specific signaling pathways in response to defined nitrosative stress levels that may occur within the tumor microenvironment. Threshold levels of NO required for activation and stabilization of key proteins involved in carcinogenesis including p53, ERK, Akt and HIF have been identified. Importantly, threshold NO levels are further influenced by reactive oxygen species (ROS) including superoxide, which can shift or attenuate NO-mediated signaling as observed in both tumor and endothelial cells. Our studies have been extended to determine levels of NO that are critical during angiogenic response through regulation of the anti-angiogenic agent thrombospondin-1 (TSP-1) and pro-angiogenic agent matrix metalloproteinase-9 (MMP-9). The quantification of redox events at the cellular level has revealed potential mechanisms that may either limit or potentiate tumor growth, and helped define the positive and negative function of nitric oxide in cancer.
Collapse
Affiliation(s)
- Lisa A. Ridnour
- Radiation Biology Branch National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Douglas D. Thomas
- Department of Medicinal Chemistry and Pharmacology, School of Pharmacy University of Illinois at Chicago, Illinois
| | - Christopher Switzer
- Radiation Biology Branch National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Wilmarie Flores-Santana
- Radiation Biology Branch National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Jeffrey S. Isenberg
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Stefan Ambs
- Laboratory of Human Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - David D. Roberts
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - David A. Wink
- Radiation Biology Branch National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
320
|
Trachootham D, Lu W, Ogasawara MA, Valle NRD, Huang P. Redox regulation of cell survival. Antioxid Redox Signal 2008; 10:1343-74. [PMID: 18522489 PMCID: PMC2932530 DOI: 10.1089/ars.2007.1957] [Citation(s) in RCA: 1288] [Impact Index Per Article: 75.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2007] [Revised: 02/06/2008] [Accepted: 02/06/2008] [Indexed: 12/18/2022]
Abstract
Reactive oxygen species (ROS) and reactive nitrogen species (RNS) play important roles in regulation of cell survival. In general, moderate levels of ROS/RNS may function as signals to promote cell proliferation and survival, whereas severe increase of ROS/RNS can induce cell death. Under physiologic conditions, the balance between generation and elimination of ROS/RNS maintains the proper function of redox-sensitive signaling proteins. Normally, the redox homeostasis ensures that the cells respond properly to endogenous and exogenous stimuli. However, when the redox homeostasis is disturbed, oxidative stress may lead to aberrant cell death and contribute to disease development. This review focuses on the roles of key transcription factors, signal-transduction pathways, and cell-death regulators in affecting cell survival, and how the redox systems regulate the functions of these molecules. The current understanding of how disturbance in redox homeostasis may affect cell death and contribute to the development of diseases such as cancer and degenerative disorders is reviewed. We also discuss how the basic knowledge on redox regulation of cell survival can be used to develop strategies for the treatment or prevention of those diseases.
Collapse
Affiliation(s)
- Dunyaporn Trachootham
- Department of Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
- Faculty of Dentistry, Thammasat University (Rangsit Campus), Pathum-thani, Thailand
| | - Weiqin Lu
- Department of Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
| | - Marcia A. Ogasawara
- Department of Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
| | - Nilsa Rivera-Del Valle
- Department of Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
| | - Peng Huang
- Department of Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
| |
Collapse
|
321
|
Park YK, Ahn DR, Oh M, Lee T, Yang EG, Son M, Park H. Nitric oxide donor, (+/-)-S-nitroso-N-acetylpenicillamine, stabilizes transactive hypoxia-inducible factor-1alpha by inhibiting von Hippel-Lindau recruitment and asparagine hydroxylation. Mol Pharmacol 2008; 74:236-45. [PMID: 18426857 DOI: 10.1124/mol.108.045278] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We have confirmed that the NO donor (+/-)-S-nitroso-N-acetylpenicillamine (SNAP) stabilizes the transactive form of hypoxia-inducible factor-1alpha (HIF-1alpha), leading to the induction of HIF-1alpha target genes such as vascular endothelial growth factor and carbonic anhydrase 9. Activation of HIF-1alpha should require inhibition of the dual system that keeps it inactive. One is ubiquitination, which is triggered by hydroxylation of HIF-1alpha-proline and the subsequent binding of E3 ubiquitin ligase, the von Hippel Lindau (VHL) protein. The other is hydroxylation of HIF-1alpha-asparagine, which reduces the affinity of HIF-1alpha for its coactivator, cAMP responsive element binding protein/p300. We examined the effects of the NO donor SNAP on proline and asparagine hydroxylation of HIF-1alpha peptides by measuring the activities of the corresponding enzymes, HIF-1alpha-specific proline hydroxylase 2 (PHD2) and the HIF-1alpha-specific asparagine hydroxylase, designated factor inhibiting HIF-1alpha (FIH-1), respectively. We found that the SNAP did not prevent PHD2 from hydroxylating the proline of HIF-1alpha. Instead, it blocked the interaction between VHL and the proline-hydroxylated HIF-1alpha, but only when the reducing agents Fe(II) and vitamin C were limiting. The fact that the absence of cysteine 520 of HIF-1alpha abolishes its responsiveness to SNAP suggests that this residue mediates the inhibition by SNAP of the interaction between VHL and HIF-1alpha, presumably by S-nitrosylation of HIF-1alpha. Un-like PHD2, asparagine hydroxylation by FIH-1 was directly inhibited by SNAP, but again only when reducing agents were limiting. Substitution of cysteine 800 of HIF-1alpha with alanine failed to reverse the inhibitory effects of SNAP on asparagine hydroxylation, implying that FIH-1, not its substrate HIF-1alpha, is inhibited by SNAP.
Collapse
Affiliation(s)
- Young-Kwon Park
- Department of Life Science, University of Seoul, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
322
|
Abstract
S-Nitrosylation is a ubiquitous signaling process in biological systems. Research regarding this signaling has been hampered, however, by assays that lack sensitivity and specificity. In particular, iodine-based assays for S-nitrosothiols (1) produce nitrosyliodide, a potent nitrosating agent that can be lost to reactions in the biological sample being studied; (2) require pretreatment of biological samples with several reagents that react with proteins, artifactually forming or breaking S-NO bonds before the assay; and (3) are not sensitive or specific for nitrogen oxides in biological samples, reporting a wide range of different concentrations and falsely reporting NO-modified proteins, to be nitrite. These data, therefore, suggest that iodine-based assays should never be used for biological S-nitrosothiols. There are other assays that provide reasonably sensitive and accurate data regarding biological S-nitrosothiols, including assays based on mass spectrometry, spectrophotometry, chemiluminescence, fluorescence, and immunostaining. Each assay, however, has limitations and should be quantitatively complemented by separate assays. Continued improvement in assays will facilitate improved understanding of S-nitrosylation signaling.
Collapse
Affiliation(s)
- Lisa A Palmer
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | | |
Collapse
|
323
|
Dewhirst MW, Cao Y, Moeller B. Cycling hypoxia and free radicals regulate angiogenesis and radiotherapy response. Nat Rev Cancer 2008; 8:425-37. [PMID: 18500244 PMCID: PMC3943205 DOI: 10.1038/nrc2397] [Citation(s) in RCA: 777] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hypoxia and free radicals, such as reactive oxygen and nitrogen species, can alter the function and/or activity of the transcription factor hypoxia-inducible factor 1 (HIF1). Interplay between free radicals, hypoxia and HIF1 activity is complex and can influence the earliest stages of tumour development. The hypoxic environment of tumours is heterogeneous, both spatially and temporally, and can change in response to cytotoxic therapy. Free radicals created by hypoxia, hypoxia-reoxygenation cycling and immune cell infiltration after cytotoxic therapy strongly influence HIF1 activity. HIF1 can then promote endothelial and tumour cell survival. As discussed here, a constant theme emerges: inhibition of HIF1 activity will have therapeutic benefit.
Collapse
Affiliation(s)
- Mark W Dewhirst
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | |
Collapse
|
324
|
Park H. Novel Dioxygenases, HIF-α Specific Prolyl-hydroxylase and Asparanginyl-hydroxylase: O 2 Switch for Cell Survival. Toxicol Res 2008; 24:101-107. [PMID: 32038783 PMCID: PMC7006278 DOI: 10.5487/tr.2008.24.2.101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2008] [Revised: 05/28/2008] [Accepted: 05/29/2008] [Indexed: 11/20/2022] Open
Abstract
Studies on hypoxia-signaling pathways have revealed novel Fe(II) and α-ketoglutarate-dependent dioxygenases that hydroxylate prolyl or asparaginyl residues of a transactivator, Hypoxia-Inducible Factor-α (HIF-α) protein. The recognition of these unprecedented dioxygenases has led to open a new paradigm that the hydroxylation mediates an instant post-translational modification of a protein in response to the changes in cellular concentrations of oxygen, reducing agents, or α-ketoglutarate. Activity of HIF-α is repressed by two hydroxylases. One is HIF-α specific prolyl-hydroxylases, referred as prolyl-hydroxylase domain (PHD). The other is HIF-α specific asparaginyl-hydroxylase, referred as factor-inhibiting HIF-1 (FIH-1). The facts (i) that many dioxygenases commonly use molecular oxygen and reducing agents during detoxification of xenobiotics, (ii) that detoxification reaction produces radicals and reactive oxygen species, and (iii) that activities of both PHD and FIH-1 are regulated by the changes in the balance between oxygen species and reducing agents, imply the possibility that the activity of HIF-α can be increased during detoxification process. The importance of HIF-α in cancer and ischemic diseases has been emphasized since its target genes mediate various hypoxic responses including angiogenesis, erythropoiesis, glycolysis, pH balance, metastasis, invasion and cell survival. Therefore, activators of PHDs and FIH-1 can be potential anticancer drugs which could reduce the activity of HIF, whereas inhibitors, for preventing ischemic diseases. This review highlights these novel dioxygenases, PHDs and FIH-1 as specific target against not only cancers but also ischemic diseases.
Collapse
Affiliation(s)
- Hyunsung Park
- Department of Life Science, University of Seoul, Tongdaemun-gu, Seoul, 130-743 Korea
| |
Collapse
|
325
|
iNOS as a therapeutic target for treatment of human tumors. Nitric Oxide 2008; 19:217-24. [PMID: 18515106 DOI: 10.1016/j.niox.2008.05.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2008] [Revised: 05/02/2008] [Accepted: 05/05/2008] [Indexed: 01/25/2023]
Abstract
Nitric oxide synthase (NOS) has been shown to be overexpressed in a number of human tumors compared to normal tissues and therefore potentially represents an exploitable target in future anticancer therapies. To achieve this, there will be a need to profile tumors to identify those expressing high levels of NOS; alternatively, endogenous (low) levels of NOS could be modulated by induction or through gene therapy approaches. NOS consists of a reductase domain which shares a high degree of sequence homology with P450 reductase and this domain supplies reducing equivalents to a haem containing oxygenase domain that is responsible for the production of nitric oxide. Thus, there are a number of routes of exploitation. Firstly, to take advantage of the reductase domain to activate bioreductive drugs as has been exemplified with tirapazamine and now extended to AQ4N (1,4-bis{2-(dimethylamino-N-oxide)ethylamino}5,8-dihydroxy-anthracene-9,10-dione). Secondly, to take advantage of nitric oxide production for its ability to increase the sensitivity of resistant hypoxic cells to radiation. Lastly, to utilize inhibition of HIF-1 to amplify NO based therapies. In this review we provide examples/evidence of how these objectives can be achieved.
Collapse
|
326
|
Li Q, Michaud M, Stewart W, Schwartz M, Madri JA. Modeling the neurovascular niche: murine strain differences mimic the range of responses to chronic hypoxia in the premature newborn. J Neurosci Res 2008; 86:1227-42. [PMID: 18092360 PMCID: PMC2644407 DOI: 10.1002/jnr.21597] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Preterm birth results in significant cognitive and motor disabilities, but recent evidence suggests that there is variable recovery over time. One possibility that may explain this variable recovery entails variable neurogenic responses in the subventricular zone (SVZ) following the period of chronic hypoxia experienced by these neonates. In this report, we have characterized the responses to chronic hypoxia of two mouse strains that represent a wide range of susceptibility to chronic hypoxia. We determined that C57BL/6 pups and neural progenitor cells (NPCs) derived from them exhibit a blunted response to hypoxic insult compared with CD-1 pups and NPCs. Specifically, C57BL/6 pups and NPCs exhibited blunted in vivo and in vitro proliferative and increased apoptotic responses to hypoxic insult. Additionally, C57BL/6 NPCs exhibited lower baseline levels and hypoxia-induced levels of selected transcription factors, growth factors, and receptors (including HIF-1alpha, PHD2, BDNF, VEGF, SDF-1, TrkB, Nrp-1, CXCR4, and NO) that determine, in part, the responsiveness to chronic hypoxic insult compared with CD-1 pups and NPCs, providing insight into this important and timely problem in perinatology.
Collapse
Affiliation(s)
- Qi Li
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Michael Michaud
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - William Stewart
- Department of Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - Michael Schwartz
- Department of Neurobiology, Yale University School of Medicine, New Haven, Connecticut
| | - Joseph A. Madri
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
327
|
Weigert A, Brüne B. Nitric oxide, apoptosis and macrophage polarization during tumor progression. Nitric Oxide 2008; 19:95-102. [PMID: 18486631 DOI: 10.1016/j.niox.2008.04.021] [Citation(s) in RCA: 113] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2008] [Accepted: 04/18/2008] [Indexed: 12/31/2022]
Abstract
Decreased oxygen availability evokes adaptive responses, which are primarily under the gene regulatory control of hypoxia inducible factor-1 (HIF-1). Hypoxic cores of a growing tumor cell mass use this signaling circuit to gain access to further blood and nutrient supply that guarantees their continuing growth. Interestingly, NO shares with hypoxia the ability to block prolyl-hydroxylase (PHD) activity, and thus the ability to stabilize hypoxia inducible factor 1 alpha (HIF-1 alpha). Under these conditions NO mimics hypoxia, which might contribute to tumor development. Stimulating/triggering innate immune responses associated with macrophage activation often correlated with iNOS induction and massive NO release, which is known to kill NO-sensitive tumors. However, this safeguard mechanism will only be effective if all tumor cells are eliminated because apoptotic death of tumor cells implies mechanisms to stop macrophages from attacking the survivors. Apoptotic cells release factors, among others sphingosine-1-phosphate (S1P), which reprogram macrophages. Macrophage reprogramming shifts responses from a M1 and thus pro-inflammatory and killing phenotype, to a M2 phenotype, which is anti-inflammatory and pro-angiogenic. These polarized tumor associated macrophages (TAM) are actively contributing to tumor development. Apparently NO uses distinct signaling pathways that could serve as an explanation to understand how NO affects tumor development. Some of these pathways, especially the ability of NO to mimic hypoxia at the level of HIF-1 alpha, as well as the role of macrophage polarization by apoptotic cells with accompanying changes in the iNOS versus arginase ratio and activities, will be discussed to better understand how NO affects tumor growth.
Collapse
Affiliation(s)
- Andreas Weigert
- Institute of Biochemistry I/ZAFES, Goethe-University, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | | |
Collapse
|
328
|
Bove PF, Hristova M, Wesley UV, Olson N, Lounsbury KM, van der Vliet A. Inflammatory levels of nitric oxide inhibit airway epithelial cell migration by inhibition of the kinase ERK1/2 and activation of hypoxia-inducible factor-1 alpha. J Biol Chem 2008; 283:17919-28. [PMID: 18424783 DOI: 10.1074/jbc.m709914200] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Increased synthesis of NO during airway inflammation, caused by induction of nitric-oxide synthase 2 in several lung cell types, may contribute to epithelial injury and permeability. To investigate the consequence of elevated NO production on epithelial function, we exposed cultured monolayers of human bronchial epithelial cells to the NO donor diethylenetriaamine NONOate. At concentrations generating high nanomolar levels of NO, representative of inflammatory conditions, diethylenetriaamine NONOate markedly reduced wound closure in an in vitro scratch injury model, primarily by inhibiting epithelial cell migration. Analysis of signaling pathways and gene expression profiles indicated a rapid induction of the mitogen-activated protein kinase phosphatase (MPK)-1 and decrease in extracellular signal-regulated kinase (ERK)1/2 activation, as well as marked stabilization of hypoxia-inducible factor (HIF)-1alpha and activation of hypoxia-responsive genes, under these conditions. Inhibition of ERK1/2 signaling using U0126 enhanced HIF-1alpha stabilization, implicating ERK1/2 dephosphorylation as a contributing mechanism in NO-mediated HIF-1alpha activation. Activation of HIF-1alpha by the hypoxia mimic cobalt chloride, or cell transfection with a degradation-resistant HIF-1alpha mutant construct inhibited epithelial wound repair, implicating HIF-1alpha in NO-mediated inhibition of cell migration. Conversely, NO-mediated inhibition of epithelial wound closure was largely prevented after small interfering RNA suppression of HIF-1alpha. Finally, NO-mediated inhibition of cell migration was associated with HIF-1alpha-dependent induction of PAI-1 and activation of p53, both negative regulators of epithelial cell migration. Collectively, our results demonstrate that inflammatory levels of NO inhibit epithelial cell migration, because of suppression of ERK1/2 signaling, and activation of HIF-1alpha and p53, with potential consequences for epithelial repair and remodeling during airway inflammation.
Collapse
Affiliation(s)
- Peter F Bove
- Department of Pathology, College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | | | | | | | | | | |
Collapse
|
329
|
Biswas SK, Sica A, Lewis CE. Plasticity of macrophage function during tumor progression: regulation by distinct molecular mechanisms. THE JOURNAL OF IMMUNOLOGY 2008; 180:2011-7. [PMID: 18250403 DOI: 10.4049/jimmunol.180.4.2011] [Citation(s) in RCA: 321] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Recent studies have shown that macrophages play an important part in both tumor initiation and various key steps in growth and metastasis. These cells show a remarkable degree of plasticity during tumor development with a "switch" in macrophage phenotypes occurring during the course of tumor progression. During chronic inflammation they appear to predispose a given tissue to tumor initiation by the release of factors that promote neoplastic transformation. Following this, their phenotype shifts more toward one that is immunosuppressive and supports tumor growth, angiogenesis, and metastasis. In this review, we discuss the evidence for this plasticity of macrophage functions, the specific signaling mechanisms that may be regulating it, and the new targets for anticancer therapies highlighted by these findings.
Collapse
Affiliation(s)
- Subhra K Biswas
- Singapore Immunology Network, Biomedical Sciences Institutes, Agency for Science, Technology and Research, Singapore
| | | | | |
Collapse
|
330
|
Oxygen-regulated isoforms of cytochrome c oxidase have differential effects on its nitric oxide production and on hypoxic signaling. Proc Natl Acad Sci U S A 2008; 105:8203-8. [PMID: 18388202 DOI: 10.1073/pnas.0709461105] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Recently, it has been reported that mitochondria possess a novel pathway for nitric oxide (NO) synthesis. This pathway is induced when cells experience hypoxia, is nitrite (NO(2)(-))-dependent, is independent of NO synthases, and is catalyzed by cytochrome c oxidase (Cco). It has been proposed that this mitochondrially produced NO is a component of hypoxic signaling and the induction of nuclear hypoxic genes. In this study, we examine the NO(2)(-)-dependent NO production in yeast engineered to contain alternative isoforms, Va or Vb, of Cco subunit V. Previous studies have shown that these isoforms have differential effects on oxygen reduction by Cco, and that their genes (COX5a and COX5b, respectively) are inversely regulated by oxygen. Here, we find that the Vb isozyme has a higher turnover rate for NO production than the Va isozyme and that the Vb isozyme produces NO at much higher oxygen concentrations than the Va isozyme. We have also found that the hypoxic genes CYC7 and OLE1 are induced to higher levels in a strain carrying the Vb isozyme than in a strain carrying the Va isozyme. Together, these results demonstrate that the subunit V isoforms have differential effects on NO(2)(-)-dependent NO production by Cco and provide further support for a role of Cco in hypoxic signaling. These findings also suggest a positive feedback mechanism in which mitochondrially produced NO induces expression of COX5b, whose protein product then functions to enhance the ability of Cco to produce NO in hypoxic/anoxic cells.
Collapse
|
331
|
Richardson DR, Lok HC. The nitric oxide–iron interplay in mammalian cells: Transport and storage of dinitrosyl iron complexes. Biochim Biophys Acta Gen Subj 2008; 1780:638-51. [DOI: 10.1016/j.bbagen.2007.12.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2007] [Revised: 12/03/2007] [Accepted: 12/18/2007] [Indexed: 02/05/2023]
|
332
|
Gauter-Fleckenstein B, Fleckenstein K, Owzar K, Jiang C, Batinic-Haberle I, Vujaskovic Z. Comparison of two Mn porphyrin-based mimics of superoxide dismutase in pulmonary radioprotection. Free Radic Biol Med 2008; 44:982-9. [PMID: 18082148 PMCID: PMC3684016 DOI: 10.1016/j.freeradbiomed.2007.10.058] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2007] [Revised: 10/30/2007] [Accepted: 10/31/2007] [Indexed: 02/02/2023]
Abstract
Development of radiation therapy (RT)-induced lung injury is associated with chronic production of reactive oxygen and nitrogen species (ROS/RNS). MnTE-2-PyP5+ is a catalytic Mn porphyrin mimic of SOD, already shown to protect lungs from RT-induced injury by scavenging ROS/RNS. The purpose of this study was to compare MnTE-2-PyP5+ with a newly introduced analogue MnTnHex-2-PyP5+, which is expected to be a more effective radioprotector due to its lipophilic properties. This study shows that Fischer rats which were irradiated to their right hemithorax (28 Gy) have less pulmonary injury as measured using breathing frequencies when treated with daily subcutaneous injections of MnTE-2-PyP5+ (3 and 6 mg/kg) or MnTnHex-2-PyP5+ (0.3, 0.6, or 1.0 mg/kg) for 2 weeks after RT. However, at 16 weeks post-RT, only MnTE-2-PyP5+ at a dose of 6 mg/kg is able to ameliorate oxidative damage, block activation of HIF-1alpha and TGF-beta, and impair upregulation of CA-IX and VEGF. MnTnHex-2-PyP5+ at a dose of 0.3 mg/kg is effective only in reducing RT-induced TGF-beta and CA-IX expression. Significant loss of body weight was observed in animals receiving MnTnHex-2-PyP5+ (0.3 and 0.6 mg/kg). MnTnHex-2-PyP5+ has the ability to dissolve lipid membranes, causing local irritation/necrosis at injection sites if given at doses of 1 mg/kg or higher. In conclusion, both compounds show an ability to ameliorate lung damage as measured using breathing frequencies and histopathologic evaluation. However, MnTE-2-PyP5+ at 6 mg/kg proved to be more effective in reducing expression of key molecular factors known to play an important role in radiation-induced lung injury.
Collapse
|
333
|
Kunieda T, Minamino T, Miura K, Katsuno T, Tateno K, Miyauchi H, Kaneko S, Bradfield CA, FitzGerald GA, Komuro I. Reduced Nitric Oxide Causes Age-Associated Impairment of Circadian Rhythmicity. Circ Res 2008; 102:607-14. [DOI: 10.1161/circresaha.107.162230] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Takeshige Kunieda
- From the Department of Cardiovascular Science and Medicine (T. Kunieda, T.M., K.M., T. Katsuno, K.T., H.M., I.K.), Chiba University Graduate School of Medicine, Japan; Institute for Translational Medicine and Therapeutics (T. Kunieda, G.A.F.), University of Pennsylvania, Philadelphia; PRESTO (T.M.), Japan Science and Technology Agency, Saitama, Japan; Department of Disease Control of Homeostasis (S.K.), Kanazawa University Graduate School of Medicine, Ishikawa, Japan; and McArdle Laboratory for
| | - Tohru Minamino
- From the Department of Cardiovascular Science and Medicine (T. Kunieda, T.M., K.M., T. Katsuno, K.T., H.M., I.K.), Chiba University Graduate School of Medicine, Japan; Institute for Translational Medicine and Therapeutics (T. Kunieda, G.A.F.), University of Pennsylvania, Philadelphia; PRESTO (T.M.), Japan Science and Technology Agency, Saitama, Japan; Department of Disease Control of Homeostasis (S.K.), Kanazawa University Graduate School of Medicine, Ishikawa, Japan; and McArdle Laboratory for
| | - Kentaro Miura
- From the Department of Cardiovascular Science and Medicine (T. Kunieda, T.M., K.M., T. Katsuno, K.T., H.M., I.K.), Chiba University Graduate School of Medicine, Japan; Institute for Translational Medicine and Therapeutics (T. Kunieda, G.A.F.), University of Pennsylvania, Philadelphia; PRESTO (T.M.), Japan Science and Technology Agency, Saitama, Japan; Department of Disease Control of Homeostasis (S.K.), Kanazawa University Graduate School of Medicine, Ishikawa, Japan; and McArdle Laboratory for
| | - Taro Katsuno
- From the Department of Cardiovascular Science and Medicine (T. Kunieda, T.M., K.M., T. Katsuno, K.T., H.M., I.K.), Chiba University Graduate School of Medicine, Japan; Institute for Translational Medicine and Therapeutics (T. Kunieda, G.A.F.), University of Pennsylvania, Philadelphia; PRESTO (T.M.), Japan Science and Technology Agency, Saitama, Japan; Department of Disease Control of Homeostasis (S.K.), Kanazawa University Graduate School of Medicine, Ishikawa, Japan; and McArdle Laboratory for
| | - Kaoru Tateno
- From the Department of Cardiovascular Science and Medicine (T. Kunieda, T.M., K.M., T. Katsuno, K.T., H.M., I.K.), Chiba University Graduate School of Medicine, Japan; Institute for Translational Medicine and Therapeutics (T. Kunieda, G.A.F.), University of Pennsylvania, Philadelphia; PRESTO (T.M.), Japan Science and Technology Agency, Saitama, Japan; Department of Disease Control of Homeostasis (S.K.), Kanazawa University Graduate School of Medicine, Ishikawa, Japan; and McArdle Laboratory for
| | - Hideyuki Miyauchi
- From the Department of Cardiovascular Science and Medicine (T. Kunieda, T.M., K.M., T. Katsuno, K.T., H.M., I.K.), Chiba University Graduate School of Medicine, Japan; Institute for Translational Medicine and Therapeutics (T. Kunieda, G.A.F.), University of Pennsylvania, Philadelphia; PRESTO (T.M.), Japan Science and Technology Agency, Saitama, Japan; Department of Disease Control of Homeostasis (S.K.), Kanazawa University Graduate School of Medicine, Ishikawa, Japan; and McArdle Laboratory for
| | - Shuichi Kaneko
- From the Department of Cardiovascular Science and Medicine (T. Kunieda, T.M., K.M., T. Katsuno, K.T., H.M., I.K.), Chiba University Graduate School of Medicine, Japan; Institute for Translational Medicine and Therapeutics (T. Kunieda, G.A.F.), University of Pennsylvania, Philadelphia; PRESTO (T.M.), Japan Science and Technology Agency, Saitama, Japan; Department of Disease Control of Homeostasis (S.K.), Kanazawa University Graduate School of Medicine, Ishikawa, Japan; and McArdle Laboratory for
| | - Christopher A. Bradfield
- From the Department of Cardiovascular Science and Medicine (T. Kunieda, T.M., K.M., T. Katsuno, K.T., H.M., I.K.), Chiba University Graduate School of Medicine, Japan; Institute for Translational Medicine and Therapeutics (T. Kunieda, G.A.F.), University of Pennsylvania, Philadelphia; PRESTO (T.M.), Japan Science and Technology Agency, Saitama, Japan; Department of Disease Control of Homeostasis (S.K.), Kanazawa University Graduate School of Medicine, Ishikawa, Japan; and McArdle Laboratory for
| | - Garret A. FitzGerald
- From the Department of Cardiovascular Science and Medicine (T. Kunieda, T.M., K.M., T. Katsuno, K.T., H.M., I.K.), Chiba University Graduate School of Medicine, Japan; Institute for Translational Medicine and Therapeutics (T. Kunieda, G.A.F.), University of Pennsylvania, Philadelphia; PRESTO (T.M.), Japan Science and Technology Agency, Saitama, Japan; Department of Disease Control of Homeostasis (S.K.), Kanazawa University Graduate School of Medicine, Ishikawa, Japan; and McArdle Laboratory for
| | - Issei Komuro
- From the Department of Cardiovascular Science and Medicine (T. Kunieda, T.M., K.M., T. Katsuno, K.T., H.M., I.K.), Chiba University Graduate School of Medicine, Japan; Institute for Translational Medicine and Therapeutics (T. Kunieda, G.A.F.), University of Pennsylvania, Philadelphia; PRESTO (T.M.), Japan Science and Technology Agency, Saitama, Japan; Department of Disease Control of Homeostasis (S.K.), Kanazawa University Graduate School of Medicine, Ishikawa, Japan; and McArdle Laboratory for
| |
Collapse
|
334
|
Ahn GO, Brown JM. Matrix metalloproteinase-9 is required for tumor vasculogenesis but not for angiogenesis: role of bone marrow-derived myelomonocytic cells. Cancer Cell 2008; 13:193-205. [PMID: 18328424 PMCID: PMC2967441 DOI: 10.1016/j.ccr.2007.11.032] [Citation(s) in RCA: 357] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2007] [Revised: 10/16/2007] [Accepted: 11/28/2007] [Indexed: 11/23/2022]
Abstract
Tumor vasculature is derived from sprouting of local vessels (angiogenesis) and bone marrow (BM)-derived circulating cells (vasculogenesis). By using a model system of transplanting tumors into an irradiated normal tissue to prevent angiogenesis, we found that tumors were unable to grow in matrix metalloproteinase-9 (MMP-9) knockout mice, but tumor growth could be restored by transplantation of wild-type BM. Endothelial progenitor cells did not contribute significantly to this process. Rather, CD11b-positive myelomonocytic cells from the transplanted BM were responsible for tumor growth and the development of immature blood vessels in MMP-9 knockout mice receiving wild-type BM. Our results suggest that MMP-9 could be an important target for adjunct therapy to enhance the response of tumors to radiotherapy.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Bone Marrow Cells/drug effects
- Bone Marrow Cells/enzymology
- Bone Marrow Cells/immunology
- Bone Marrow Transplantation
- CD11b Antigen/metabolism
- Diphosphonates/pharmacology
- Diphosphonates/therapeutic use
- Endothelial Cells/enzymology
- Imidazoles/pharmacology
- Imidazoles/therapeutic use
- Matrix Metalloproteinase 9/deficiency
- Matrix Metalloproteinase 9/genetics
- Matrix Metalloproteinase 9/metabolism
- Matrix Metalloproteinase Inhibitors
- Melanoma, Experimental
- Mice
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Mice, Knockout
- Monocytes/drug effects
- Monocytes/enzymology
- Monocytes/immunology
- Monocytes/transplantation
- Myeloid Cells/drug effects
- Myeloid Cells/enzymology
- Myeloid Cells/immunology
- Myeloid Cells/transplantation
- Neoplasms, Experimental/blood supply
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/enzymology
- Neoplasms, Experimental/pathology
- Neoplasms, Experimental/radiotherapy
- Neovascularization, Pathologic/enzymology
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/prevention & control
- Protease Inhibitors/pharmacology
- Protease Inhibitors/therapeutic use
- Signal Transduction
- Stem Cells/enzymology
- Subcutaneous Tissue/blood supply
- Subcutaneous Tissue/radiation effects
- Subcutaneous Tissue/surgery
- Time Factors
- Zoledronic Acid
Collapse
Affiliation(s)
- G-One Ahn
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, 269 Campus Drive, CCSR-South, Room 1255, Stanford, CA 94305, USA
| | | |
Collapse
|
335
|
Sonveaux P. Provascular strategy: Targeting functional adaptations of mature blood vessels in tumors to selectively influence the tumor vascular reactivity and improve cancer treatment. Radiother Oncol 2008; 86:300-13. [PMID: 18313779 DOI: 10.1016/j.radonc.2008.01.024] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2007] [Revised: 01/30/2008] [Accepted: 01/30/2008] [Indexed: 12/22/2022]
|
336
|
Abstract
The increase in body size of humans and other vertebrates requires a physiological infrastructure to provide adequate delivery of oxygen to tissues and cells to maintain oxygen homeostasis. The heart, lungs and the vasculature are all part of a highly regulated system that ensures the distribution of the precise amount of oxygen needed throughout the mammalian organism. Given its fundamental impact on physiology and pathology, it is no surprise that the response of cells to a lack of oxygen, termed hypoxia, has been the focus of many research groups worldwide for many decades now. The transcriptional complex hypoxia-inducible factor has emerged as a key regulator of the molecular hypoxic response, mediating a wide range of physiological and cellular mechanisms necessary to adapt to reduced oxygen.
Collapse
|
337
|
Sell S, Lindermayr C, Durner J. Identification of S‐Nitrosylated Proteins in Plants. Methods Enzymol 2008; 440:283-93. [DOI: 10.1016/s0076-6879(07)00818-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
338
|
Sumbayev VV, Yasinska IM. Protein S‐Nitrosation in Signal Transduction: Assays for Specific Qualitative and Quantitative Analysis. Methods Enzymol 2008; 440:209-19. [DOI: 10.1016/s0076-6879(07)00812-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
339
|
Yang L, Jiang Y, Wu SF, Zhou MY, Wu YL, Chen GQ. CCAAT/enhancer-binding protein alpha antagonizes transcriptional activity of hypoxia-inducible factor 1 alpha with direct protein-protein interaction. Carcinogenesis 2007; 29:291-8. [PMID: 18024476 DOI: 10.1093/carcin/bgm262] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Hypoxia-inducible factor 1 (HIF-1), a master heterodimeric transcriptional regulator consisting of HIF-1alpha and HIF-1beta subunits for cellular response to hypoxia, plays an important role in carcinogenesis, while CCAAT/enhancer-binding protein alpha (C/EBPalpha) is proposed to act as a tumor suppressor in C/EBPalpha-expressing tissues. Previously, we reported that ectopically expressed HIF-1alpha protein interacts with and enhances transcriptional activity of C/EBPalpha, which favors leukemic cell differentiation. Here we further showed that such an interaction also occurred in their endogenously expressing state of leukemic U937 cells. Glutathione S-transferase pull-down assay proposed that the protein-protein interaction was direct, and transactivation domains of C/EBPalpha and the basic helix-loop-helix domain of HIF-1alpha were essential for such an interaction. More intriguingly, we provided the first demonstration that C/EBPalpha competed with HIF-1beta for direct binding to HIF-1alpha protein. Correspondingly, C/EBPalpha overexpression significantly inhibited the DNA-binding ability of HIF-1 and expressions of hypoxia-responsive element-driven luciferase and HIF-1-targeted genes vascular endothelial growth factor, glucose transporter-1 and phosphoglycerate kinase 1. In parallel, suppression of C/EBPalpha expression by specific small hairpin RNA increased DNA-binding ability of HIF-1 and expression of these HIF-1-targeted genes in leukemic U937 cells. These results would provide new insights for antitumor potential of C/EBPalpha protein.
Collapse
Affiliation(s)
- L Yang
- Institute of Health Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences-Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | | | | | | | | | | |
Collapse
|
340
|
Hypoxia-inducible factors: Crosstalk between their protein stability and protein degradation. Cancer Lett 2007; 257:145-56. [DOI: 10.1016/j.canlet.2007.08.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2007] [Revised: 08/02/2007] [Accepted: 08/06/2007] [Indexed: 11/21/2022]
|
341
|
Abstract
PURPOSE OF REVIEW Angiogenesis and inflammation are important features in atherosclerotic plaque destabilization. The transcription factors hypoxia-inducible factor-1alpha and Notch are key regulators of angiogenesis. In addition, hypoxia-inducible factor-1alpha has been linked to regulation of inflammatory processes and innate immunity. This review will document how hypoxia-inducible factor-mediated signaling pathways are initiated in hypoxic cells, and how the hypoxia-inducible factor and Notch-dependent signaling pathways are functionally integrated. RECENT FINDINGS Activation of the hypoxia-inducible factor-mediated signaling events by hypoxia is complex and regulated by a cascade of molecular events that will be reviewed in detail. The activated form of hypoxia-inducible factor enhances Notch-dependent activation of Notch target genes, thereby providing a mechanism by which hypoxia can regulate the differentiation status of a cell. Recent observations implicate the Notch signaling pathway in proper specification of cell identity, position and behavior in a developing blood vessel sprout, and the hypoxia-inducible factor-mediated signaling pathway is critical for induction of expression of vascular endothelial growth factor. SUMMARY Hypoxia-inducible factor and Notch transcription factors represent potentially attractive targets for regulation of angiogenesis and possibly inflammation. In view of the pleiotropic effects of these transcription factors, however, successful targeting of these signaling pathways will require the development of gene specific (and possibly tissue-specific) modulators and extensive validation in relevant model systems.
Collapse
Affiliation(s)
- Jorge L Ruas
- Department of Cancer Biology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
| | | | | |
Collapse
|
342
|
Chung KKK. Say NO to neurodegeneration: role of S-nitrosylation in neurodegenerative disorders. Neurosignals 2007; 15:307-13. [PMID: 17901712 DOI: 10.1159/000109071] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2007] [Accepted: 08/10/2007] [Indexed: 01/19/2023] Open
Abstract
Nitric oxide (NO) is an important signaling molecule that controls a wide range of biological processes. One of the signaling mechanisms of NO is through the S-nitrosylation of cysteine residues on proteins. S-nitrosylation is now regarded as an important redox signaling mechanism in the regulation of different cellular and physiological functions. However, deregulation of S-nitrosylation has also been linked to various human diseases such as neurodegenerative disorders. Nitrosative stress has long been considered as a major mediator in the development of neurodegeneration, but the molecular mechanism of how NO can contribute to neurodegeneration is not completely clear. Early studies suggested that nitration of proteins, which can induce protein aggregation might contribute to the neurodegenerative process. However, several recent studies suggest that S-nitrosylation of proteins that are important for neuronal survival contributes substantially in the development of various neurodegenerative disorders. Thus, in-depth understanding of the mechanism of neurodegeneration in relation to S-nitrosylation will be critical for the development of therapeutic treatment against these neurodegenerative diseases.
Collapse
Affiliation(s)
- Kenny K K Chung
- Department of Biochemistry, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, SAR China.
| |
Collapse
|
343
|
Ohtani N, Imamura Y, Yamakoshi K, Hirota F, Nakayama R, Kubo Y, Ishimaru N, Takahashi A, Hirao A, Shimizu T, Mann DJ, Saya H, Hayashi Y, Arase S, Matsumoto M, Kazuki N, Hara E. Visualizing the dynamics of p21(Waf1/Cip1) cyclin-dependent kinase inhibitor expression in living animals. Proc Natl Acad Sci U S A 2007; 104:15034-9. [PMID: 17848507 PMCID: PMC1975854 DOI: 10.1073/pnas.0706949104] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2007] [Indexed: 12/29/2022] Open
Abstract
Although the role of p21(Waf1/Cip1) gene expression is well documented in various cell culture studies, its in vivo roles are poorly understood. To gain further insight into the role of p21(Waf1/Cip1) gene expression in vivo, we attempted to visualize the dynamics of p21(Waf1/Cip1) gene expression in living animals. In this study, we established a transgenic mice line (p21-p-luc) expressing the firefly luciferase under the control of the p21(Waf1/Cip1) gene promoter. In conjunction with a noninvasive bioluminescent imaging technique, p21-p-luc mice enabled us to monitor the endogenous p21(Waf1/Cip1) gene expression in vivo. By monitoring and quantifying the p21(Waf1/Cip1) gene expression repeatedly in the same mouse throughout its entire lifespan, we were able to unveil the dynamics of p21(Waf1/Cip1) gene expression in the aging process. We also applied this system to chemically induced skin carcinogenesis and found that the levels of p21(Waf1/Cip1) gene expression rise dramatically in benign skin papillomas, suggesting that p21(Waf1/Cip1) plays a preventative role(s) in skin tumor formation. Surprisingly, moreover, we found that the level of p21(Waf1/Cip1) expression strikingly increased in the hair bulb and oscillated with a 3-week period correlating with hair follicle cycle progression. Notably, this was accompanied by the expression of p63 but not p53. This approach, together with the analysis of p21(Waf1/Cip1) knockout mice, has uncovered a novel role for the p21(Waf1/Cip1) gene in hair development. These data illustrate the unique utility of bioluminescence imaging in advancing our understanding of the timing and, hence, likely roles of specific gene expression in higher eukaryotes.
Collapse
Affiliation(s)
| | | | | | | | - Rika Nakayama
- Center for Developmental Biology, RIKEN, Kobe 650-0047, Japan
| | - Yoshiaki Kubo
- Institute of Health Biosciences, University of Tokushima, Tokushima 770-8503, Japan
| | - Naozumi Ishimaru
- Institute of Health Biosciences, University of Tokushima, Tokushima 770-8503, Japan
| | | | - Atsushi Hirao
- Cancer Research Institute, Kanazawa University, Kanazawa 920-0934, Japan
- **CREST, Japan Science and Technology Agency, Tokyo 102-0075, Japan
| | - Takatsune Shimizu
- Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo 160-8582, Japan; and
| | - David J. Mann
- Division of Cell and Molecular Biology, Imperial College London, London SW7 2AZ, United Kingdom
| | - Hideyuki Saya
- Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo 160-8582, Japan; and
| | - Yoshio Hayashi
- Institute of Health Biosciences, University of Tokushima, Tokushima 770-8503, Japan
| | - Seiji Arase
- Institute of Health Biosciences, University of Tokushima, Tokushima 770-8503, Japan
| | | | - Nakao Kazuki
- Center for Developmental Biology, RIKEN, Kobe 650-0047, Japan
| | | |
Collapse
|
344
|
Santhanam L, Lim HK, Lim HK, Miriel V, Brown T, Patel M, Balanson S, Ryoo S, Anderson M, Irani K, Khanday F, Di Costanzo L, Nyhan D, Hare JM, Christianson DW, Rivers R, Shoukas A, Berkowitz DE. Inducible NO synthase dependent S-nitrosylation and activation of arginase1 contribute to age-related endothelial dysfunction. Circ Res 2007; 101:692-702. [PMID: 17704205 DOI: 10.1161/circresaha.107.157727] [Citation(s) in RCA: 149] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Endothelial function is impaired in aging because of a decrease in NO bioavailability. This may be, in part, attributable to increased arginase activity, which reciprocally regulates NO synthase (NOS) by competing for the common substrate, L-arginine. However, the high Km of arginase (>1 mmol/L) compared with NOS (2 to 20 micromol/L) seemingly makes direct competition for substrate unlikely. One of the mechanisms by which NO exerts its effects is by posttranslational modification through S-nitrosylation of protein cysteines. We tested the hypothesis that arginase1 activity is modulated by this mechanism, which serves to alter its substrate affinity, allowing competition with NOS for L-arginine. We demonstrate that arginase1 activity is altered by S-nitrosylation, both in vitro and ex vivo. Furthermore, using site-directed mutagenesis we demonstrate that 2 cysteine residues (C168 and C303) are able to undergo nitrosylation. S-Nitrosylation of C303 stabilizes the arginase1 trimer and reduces its Km value 6-fold. Finally, arginase1 nitrosylation is increased (and thus its Km decreased) in blood vessels from aging rats, likely contributing to impaired NO bioavailability and endothelial dysfunction. This is mediated by inducible NOS, which is expressed in the aging endothelium. These findings suggest that S-nitrosylated arginase1 can compete with NOS for L-arginine and contribute to endothelial dysfunction in the aging cardiovascular system.
Collapse
Affiliation(s)
- Lakshmi Santhanam
- Johns Hopkins University School of Medicine, 720 Rutland Ave, Traylor 621, Baltimore MD 21209, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
345
|
Toffoli S, Feron O, Raes M, Michiels C. Intermittent hypoxia changes HIF-1alpha phosphorylation pattern in endothelial cells: unravelling of a new PKA-dependent regulation of HIF-1alpha. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2007; 1773:1558-71. [PMID: 17662481 DOI: 10.1016/j.bbamcr.2007.06.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2007] [Revised: 05/22/2007] [Accepted: 06/06/2007] [Indexed: 01/09/2023]
Abstract
Vascularized tumors are exposed to intermittent hypoxia, that is, hypoxia followed by periods of reoxygenation. Abnormal structure and dysfunction of tumor blood vessels are responsible for these conditions. These repeated short periods of hypoxia concern tumor cells as well as endothelial cells. However, the effects of intermittent hypoxia are poorly understood. The aim of this study was to investigate the effects of intermittent hypoxia on endothelial cells and particularly on HIF-1alpha, a central actor in adaptive response to hypoxia. For that, endothelial cells were exposed to four repeated cycles of 1-h hypoxia followed by 30 min of reoxygenation. We showed that repeated cycles of hypoxia/reoxygenation induced a modification in HIF-l alpha phosphorylation pattern: a progressive increase in HIF-1alpha phosphorylated form was observed during the hypoxic periods. Activation of p42/p44, Akt and PKA was observed in parallel. PKA was shown to be involved in the phosphorylation of HIF-lalpha under intermittent hypoxia, while p42/p44 and Akt were not. As HIF-1 activity is often associated with enhanced cell survival, a better knowledge of the effects of intermittent hypoxia on endothelial cells and the highlight of particular mechanisms induced by intermittent hypoxia are essential to understand the behavior of endothelial cells during neo-angiogenesis.
Collapse
Affiliation(s)
- Sébastien Toffoli
- Laboratory of Biochemistry and Cellular Biology, FUNDP-University of Namur, 61 rue de Bruxelles, 5000 Namur, Belgium
| | | | | | | |
Collapse
|
346
|
Abstract
In a recent issue of Molecular Cell, Li et al. (2007) reported that ionizing radiation stabilizes hypoxia-inducible factor 1alpha (HIF-1alpha) in normoxic tumor tissues through S-nitrosylation by nitric oxide (NO) generated from neighboring activated macrophages.
Collapse
Affiliation(s)
- Randall S Johnson
- Molecular Biology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | | |
Collapse
|
347
|
Dewhirst MW, Cao Y, Li CY, Moeller B. Exploring the role of HIF-1 in early angiogenesis and response to radiotherapy. Radiother Oncol 2007; 83:249-55. [PMID: 17560674 PMCID: PMC2694841 DOI: 10.1016/j.radonc.2007.05.016] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2007] [Revised: 05/19/2007] [Accepted: 05/19/2007] [Indexed: 01/12/2023]
Abstract
The objective of this review is to examine the role that HIF-1 plays in the initiation of angiogenesis and in radiotherapy response. Although these two phenomena may at first seem unrelated, there are parallelisms to be drawn associated with the importance of reactive oxygen species in controlling the transcriptional activity of HIF-1, independently of its main driving force, hypoxia. Knowledge of the mechanisms underlying the control of HIF-1 leads to rationale for its inhibition in a range of clinical scenarios.
Collapse
|