1
|
Ehrenreich H, Gassmann M, Poustka L, Burtscher M, Hammermann P, Sirén AL, Nave KA, Miskowiak K. Exploiting moderate hypoxia to benefit patients with brain disease: Molecular mechanisms and translational research in progress. NEUROPROTECTION 2023; 1:9-19. [PMID: 37671067 PMCID: PMC7615021 DOI: 10.1002/nep3.15] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 11/17/2022] [Indexed: 09/07/2023]
Abstract
Hypoxia is increasingly recognized as an important physiological driving force. A specific transcriptional program, induced by a decrease in oxygen (O2) availability, for example, inspiratory hypoxia at high altitude, allows cells to adapt to lower O2 and limited energy metabolism. This transcriptional program is partly controlled by and partly independent of hypoxia-inducible factors. Remarkably, this same transcriptional program is stimulated in the brain by extensive motor-cognitive exercise, leading to a relative decrease in O2 supply, compared to the acutely augmented O2 requirement. We have coined the term "functional hypoxia" for this important demand-responsive, relative reduction in O2 availability. Functional hypoxia seems to be critical for enduring adaptation to higher physiological challenge that includes substantial "brain hardware upgrade," underlying advanced performance. Hypoxia-induced erythropoietin expression in the brain likely plays a decisive role in these processes, which can be imitated by recombinant human erythropoietin treatment. This article review presents hints of how inspiratory O2 manipulations can potentially contribute to enhanced brain function. It thereby provides the ground for exploiting moderate inspiratory plus functional hypoxia to treat individuals with brain disease. Finally, it sketches a planned multistep pilot study in healthy volunteers and first patients, about to start, aiming at improved performance upon motor-cognitive training under inspiratory hypoxia.
Collapse
Affiliation(s)
- Hannelore Ehrenreich
- Clinical Neuroscience, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Max Gassmann
- Institute of Veterinary Physiology and Zürich Center for Integrative Human Physiology, University of Zürich, Zürich, Switzerland
| | - Luise Poustka
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Martin Burtscher
- Faculty of Sports Science, University of Innsbruck, Innsbruck, Austria
| | | | - Anna-Leena Sirén
- Departments of Neurophysiology and Neurosurgery, University of Würzburg, Würzburg, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Kamilla Miskowiak
- Psychiatric Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Psychology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
2
|
Otsuka H, Takito J, Endo Y, Yagi H, Soeta S, Yanagisawa N, Nonaka N, Nakamura M. The expression of embryonic globin mRNA in a severely anemic mouse model induced by treatment with nitrogen-containing bisphosphonate. BMC HEMATOLOGY 2016; 16:4. [PMID: 26877876 PMCID: PMC4751657 DOI: 10.1186/s12878-016-0041-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 01/17/2016] [Indexed: 12/21/2022]
Abstract
Background Mammalian erythropoiesis can be divided into two distinct types, primitive and definitive, in which new cells are derived from the yolk sac and hematopoietic stem cells, respectively. Primitive erythropoiesis occurs within a restricted period during embryogenesis. Primitive erythrocytes remain nucleated, and their hemoglobins are different from those in definitive erythrocytes. Embryonic type hemoglobin is expressed in adult animals under genetically abnormal condition, but its later expression has not been reported in genetically normal adult animals, even under anemic conditions. We previously reported that injecting animals with nitrogen-containing bisphosphonate (NBP) decreased erythropoiesis in bone marrow (BM). Here, we induced severe anemia in a mouse model by injecting NBP injection in combination with phenylhydrazine (PHZ), and then we analyzed erythropoiesis and the levels of different types of hemoglobin. Methods Splenectomized mice were treated with NBP to inhibit erythropoiesis in BM, and with PHZ to induce hemolytic anemia. We analyzed hematopoietic sites and peripheral blood using morphological and molecular biological methods. Results Combined treatment of splenectomized mice with NBP and PHZ induced critical anemia compared to treatment with PHZ alone, and numerous nucleated erythrocytes appeared in the peripheral blood. In the BM, immature CD71-positive erythroblasts were increased, and extramedullary erythropoiesis occurred in the liver. Furthermore, embryonic type globin mRNA was detected in both the BM and the liver. In peripheral blood, spots that did not correspond to control hemoglobin were observed in 2D electrophoresis. ChIP analyses showed that KLF1 and KLF2 bind to the promoter regions of β-like globin. Wine-colored capsuled structures were unexpectedly observed in the abdominal cavity, and active erythropoiesis was also observed in these structures. Conclusion These results indicate that primitive erythropoiesis occurs in adult mice to rescue critical anemia because primitive erythropoiesis does not require macrophages as stroma whereas macrophages play a pivotal role in definitive erythropoiesis even outside the medulla. The cells expressing embryonic hemoglobin in this study were similar to primitive erythrocytes, indicating the possibility that yolk sac-derived primitive erythroid cells may persist into adulthood in mice. Electronic supplementary material The online version of this article (doi:10.1186/s12878-016-0041-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hirotada Otsuka
- Department of Oral Anatomy and Developmental Biology, School of Dentistry, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555 Japan
| | - Jiro Takito
- Department of Oral Anatomy and Developmental Biology, School of Dentistry, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555 Japan
| | - Yasuo Endo
- Division of Molecular Regulation, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575 Japan
| | - Hideki Yagi
- Faculty of Pharmacy, International University of Health and Welfare, 2600-1 Kitakanamaru, Otawara-shi, Tochigi 324-8501 Japan
| | - Satoshi Soeta
- Department of Veterinary Anatomy, Nippon Veterinary and Animal Science University, 1-7-1 Kyonan-cho, Musashino-shi, Tokyo 180-8602 Japan
| | - Nobuaki Yanagisawa
- Department of Oral Anatomy and Developmental Biology, School of Dentistry, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555 Japan
| | - Naoko Nonaka
- Department of Oral Anatomy and Developmental Biology, School of Dentistry, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555 Japan
| | - Masanori Nakamura
- Department of Oral Anatomy and Developmental Biology, School of Dentistry, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555 Japan
| |
Collapse
|
3
|
Bischof C, Krishnan J. Exploiting the hypoxia sensitive non-coding genome for organ-specific physiologic reprogramming. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1782-90. [PMID: 26851074 DOI: 10.1016/j.bbamcr.2016.01.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 01/11/2016] [Accepted: 01/28/2016] [Indexed: 12/22/2022]
Abstract
In this review we highlight the role of non-coding RNAs in the development and progression of cardiac pathology and explore the possibility of disease-associated RNAs serving as targets for cardiac-directed therapeutics. Contextually, we focus on the role of stress-induced hypoxia as a driver of disease development and progression through activation of hypoxia inducible factor 1α (HIF1α) and explore mechanisms underlying HIFα function as an enforcer of cardiac pathology through direct transcriptional coupling with the non-coding transcriptome. In the interest of clarity, we will confine our analysis to cardiac pathology and focus on three defining features of the diseased state, namely metabolic, growth and functional reprogramming. It is the aim of this review to explore possible mechanisms through which HIF1α regulation of the non-coding transcriptome connects to spatiotemporal control of gene expression to drive establishment of the diseased state, and to propose strategies for the exploitation of these unique RNAs as targets for clinical therapy. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.
Collapse
Affiliation(s)
- Corinne Bischof
- MRC Clinical Sciences Centre, Imperial College London, London W12 0NN, United Kingdom; Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Jaya Krishnan
- MRC Clinical Sciences Centre, Imperial College London, London W12 0NN, United Kingdom; Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
| |
Collapse
|
4
|
Case AJ, Madsen JM, Motto DG, Meyerholz DK, Domann FE. Manganese superoxide dismutase depletion in murine hematopoietic stem cells perturbs iron homeostasis, globin switching, and epigenetic control in erythrocyte precursor cells. Free Radic Biol Med 2013; 56:17-27. [PMID: 23219873 PMCID: PMC3578015 DOI: 10.1016/j.freeradbiomed.2012.11.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 11/16/2012] [Accepted: 11/28/2012] [Indexed: 11/20/2022]
Abstract
Heme synthesis partially occurs in the mitochondrial matrix; thus there is a high probability that enzymes and intermediates important in the production of heme will be exposed to metabolic by-products including reactive oxygen species. In addition, the need for ferrous iron for heme production, Fe/S coordination, and other processes occurring in the mitochondrial matrix suggests that aberrant fluxes of reactive oxygen species in this compartment might perturb normal iron homeostasis. Manganese superoxide dismutase (Sod2) is an antioxidant enzyme that governs steady-state levels of the superoxide in the mitochondrial matrix. Using hematopoietic stem cell-specific conditional Sod2 knockout mice we observed increased superoxide concentrations in red cell progeny, which caused significant pathologies including impaired erythrocytes and decreased ferrochelatase activity. Animals lacking Sod2 expression in erythroid precursors also displayed extramedullary hematopoiesis and systemic iron redistribution. Additionally, the increase in superoxide flux in erythroid precursors caused abnormal gene regulation of hematopoietic transcription factors, globins, and iron-response genes. Moreover, the erythroid precursors also displayed evidence of global changes in histone posttranslational modifications, a likely cause of at least some of the aberrant gene expression noted. From a therapeutic translational perspective, mitochondrially targeted superoxide-scavenging antioxidants partially rescued the observed phenotype. Taken together, our findings illuminate the superoxide sensitivity of normal iron homeostasis in erythrocyte precursors and suggest a probable link between mitochondrial redox metabolism and epigenetic control of nuclear gene regulation during mammalian erythropoiesis.
Collapse
Affiliation(s)
- Adam J. Case
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Joshua M. Madsen
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - David G. Motto
- Division of Hematology Oncology, Department of Internal Medicine, The University of Iowa, Iowa City, IA 52242, USA
| | - David K. Meyerholz
- Department of Pathology, The University of Iowa, Iowa City, IA 52242, USA
| | - Frederick E. Domann
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
5
|
Ramírez MÁ, Pericuesta E, Yáñez-Mó M, Palasz A, Gutiérrez-Adán A. Effect of long-term culture of mouse embryonic stem cells under low oxygen concentration as well as on glycosaminoglycan hyaluronan on cell proliferation and differentiation. Cell Prolif 2011; 44:75-85. [PMID: 21199012 DOI: 10.1111/j.1365-2184.2010.00732.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
OBJECTIVES Maintaining undifferentiated stem cells in defined conditions is of critical importance to improve their in vitro culture. We have evaluated the effects of culturing mouse stem (mES) cells under physiological oxygen concentration as well as by replacing fibroblast feeder layer (mEF) with gelatin or glycosaminoglycan hyaluronan (HA), on cell proliferation and differentiation. MATERIALS AND METHODS After 3 days culture or after long-term cell culture under different conditions, levels of apoptotic cell death were determined by cell cycle and TUNEL (TdT-mediated dUTP nick end labelling) assays and levels of cell proliferation by CFSE (5-(and-6)-carboxyfluorescein diacetate succinimidyl ester) labelling. We assessed spontaneous differentiation into cardiomyocytes and mRNA expression of pluripotency and differentiation biomarkers. RESULTS After 3 days culture under hypoxic conditions, levels of proliferation and apoptosis of mES cells were higher, in correlation with increase in intracellular reactive oxygen species. However, when cells were continuously grown for 1 month under those conditions, the level of apoptosis was, in all cases, under 4%. Hypoxia reduced spontaneous differentiation of mES into cardiomyocytes. Long-term culture on HA was more effective in maintaining the pluripotent state of the mES cells when compared to that on gelatin. Level of terminal differentiation was highest on mEF, intermediate on HA and lowest on gelatin. CONCLUSIONS Our data suggest that hypoxia is not necessary for maintaining pluripotency of mES cells and appeared to be detrimental during ES differentiation. Moreover, HA may offer a valuable alternative for long-term culture of mES cells in vitro.
Collapse
Affiliation(s)
- M Á Ramírez
- Departamento de Reproducción Animal INIA, Madrid, Spain.
| | | | | | | | | |
Collapse
|
6
|
Vlaski M, Lafarge X, Chevaleyre J, Duchez P, Boiron JM, Ivanovic Z. Low oxygen concentration as a general physiologic regulator of erythropoiesis beyond the EPO-related downstream tuning and a tool for the optimization of red blood cell production ex vivo. Exp Hematol 2009; 37:573-84. [PMID: 19375648 DOI: 10.1016/j.exphem.2009.01.007] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2008] [Revised: 01/19/2009] [Accepted: 01/20/2009] [Indexed: 01/30/2023]
Abstract
OBJECTIVE The control of mature erythroid progenitors and precursors' production via erythropoietin (EPO) is the major systemic regulatory mechanism in erythropoiesis. However, hypoxia seems to influence erythropoiesis beyond this well-known mechanism. The aim of our study is to test this hypothesis adapting the oxygenation level to each stage of erythropoiesis. MATERIALS AND METHODS We exploited the newly developed ex vivo three-phase protocol for red blood cell (RBC) production starting from the steady-state peripheral blood and cord blood CD34(+) cells exposed to adapted O(2) concentrations. Differentiation and maturation were followed by functional tests, morphology, immunophenotype, and analysis of molecular markers' expression. RESULTS We report here an enhancement of total RBC production if low O(2) concentrations (1.5-5%) were applied, instead of 20% O(2), during the first phase of culture. This results from a comprehensive action of low-O(2) concentration on: 1) amplification of erythroid progenitors, 2) acceleration of their proliferation, 3) differentiation, and 4) maturation of erythroid precursors. In addition, arterial blood O(2) concentration (13%) is critical for stromal cells to fully sustain the differentiation of erythroid precursors. These effects were associated with upregulation of erythroid 5-aminolevulinate synthase and gamma-globin gene expression. CONCLUSION These results imply that integral regulation of erythropoiesis is operated by low O(2) concentrations, beyond the EPO/EPO-responsive cells loop and provide a tool to optimize the technology for ex vivo production of RBC.
Collapse
Affiliation(s)
- Marija Vlaski
- Aquitaine-Limousin Branch of French Blood Institute, Bordeaux, France
| | | | | | | | | | | |
Collapse
|
7
|
Evolution of Factors Affecting Placental Oxygen Transfer. Placenta 2009; 30 Suppl A:S19-25. [DOI: 10.1016/j.placenta.2008.11.006] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2008] [Revised: 11/10/2008] [Accepted: 11/10/2008] [Indexed: 01/07/2023]
|
8
|
Abstract
This study examined whether neuronal hemoglobin (Hb) is present in rats. It then examined whether cerebral ischemia or ischemic preconditioning (IPC) affects neuronal Hb levels in vivo and in vitro. In vivo, male Sprague-Dawley rats were subjected to either 15 mins of transient middle cerebral artery occlusion (MCAO) with 24 h of reperfusion, an IPC stimulus, or 24 h of permanent MCAO (pMCAO), or IPC followed 3 days later by 24 h of pMCAO. In vitro, primary cultured neurons were exposed to 2 h of oxygen-glucose deprivation (OGD) with 22 h of reoxygenation. Results showed that Hb is widely expressed in rat cerebral neurons but not astrocytes. Hemoglobin expression was significantly upregulated in the ipsilateral caudate and the cortical core of the middle cerebral artery territory after IPC. Hemoglobin levels also increased more in the penumbral cortex and the contralateral hemisphere 24 h after pMCAO, but expressions in the ipsilateral caudate and the cortical core area were decreased. Ischemic preconditioning modified pMCAO-induced brain Hb changes. Neuronal Hb levels in vitro were increased by 2 h of OGD and 22 h of reoxygenation. These results indicate that Hb is synthesized in neurons and can be upregulated by ischemia.
Collapse
Affiliation(s)
- Yangdong He
- Department of Neurosurgery, University of Michigan, Ann Arbor, Michigan 48109-2200, USA
| | | | | | | | | | | |
Collapse
|
9
|
Komita H, Zhao X, Taylor JL, Sparvero LJ, Amoscato AA, Alber S, Watkins SC, Pardee AD, Wesa AK, Storkus WJ. CD8+ T-cell responses against hemoglobin-beta prevent solid tumor growth. Cancer Res 2008; 68:8076-84. [PMID: 18829566 PMCID: PMC2597529 DOI: 10.1158/0008-5472.can-08-0387] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Bone marrow-derived dendritic cells engineered using recombinant adenovirus to secrete high levels of IL-12p70 dramatically inhibited the growth of established CMS4 sarcomas in BALB/c mice after intratumoral administration. An analysis of splenic CD8(+) T cells in regressor mice revealed a strong, complex reactivity pattern against high-performance liquid chromatography (HPLC)-resolved peptides isolated by acid elution from single-cell suspensions of surgically resected CMS4 lesions. Mass spectrometry analyses defined two major overlapping peptide species that derive from the murine hemoglobin-beta (HBB) protein within the most stimulatory HPLC fractions. Although cultured CMS4 tumor cells failed to express HBB mRNA based on reverse transcription-PCR analyses, prophylactic vaccination of BALB/c mice with vaccines containing HBB peptides promoted specific CD8(+) T-cell responses that protected mice against a subsequent challenge with CMS4 or unrelated syngeneic (HBB(neg)) tumors of divergent histology (sarcoma, carcinomas of the breast or colon). In situ imaging suggested that vaccines limit or destabilize tumor-associated vascular structures, potentially by promoting immunity against HBB+ vascular pericytes. Importantly, there were no untoward effects of vaccination with the HBB peptide on peripheral RBC numbers, RBC hemoglobin content, or vascular structures in the brain or eye.
Collapse
Affiliation(s)
- Hideo Komita
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Xi Zhao
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Jennifer L. Taylor
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Louis J. Sparvero
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, 15213, USA
| | - Andrew A. Amoscato
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, 15213, USA
| | - Sean Alber
- Departments of Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Simon C. Watkins
- Departments of Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Angela D. Pardee
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Amy K. Wesa
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Walter J. Storkus
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, 15213, USA
| |
Collapse
|
10
|
Yoon D, Pastore YD, Divoky V, Liu E, Mlodnicka AE, Rainey K, Ponka P, Semenza GL, Schumacher A, Prchal JT. Hypoxia-inducible factor-1 deficiency results in dysregulated erythropoiesis signaling and iron homeostasis in mouse development. J Biol Chem 2006; 281:25703-11. [PMID: 16787915 DOI: 10.1074/jbc.m602329200] [Citation(s) in RCA: 170] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Hypoxia-inducible factor-1 (HIF-1) regulates the transcription of genes whose products play critical roles in energy metabolism, erythropoiesis, angiogenesis, and cell survival. Limited information is available concerning its function in mammalian hematopoiesis. Previous studies have demonstrated that homozygosity for a targeted null mutation in the Hif1alpha gene, which encodes the hypoxia-responsive alpha subunit of HIF-1, causes cardiac, vascular, and neural malformations resulting in lethality by embryonic day 10.5 (E10.5). This study revealed reduced myeloid multilineage and committed erythroid progenitors in HIF-1alpha-deficient embryos, as well as decreased hemoglobin content in erythroid colonies from HIF-1alpha-deficient yolk sacs at E9.5. Dysregulation of erythropoietin (Epo) signaling was evident from a significant decrease in mRNA levels of Epo receptor (EpoR) in Hif1alpha-/- yolk sac as well as Epo and EpoR mRNA in Hif1alpha-/- embryos. The erythropoietic defects in HIF-1alpha-deficient erythroid colonies could not be corrected by cytokines, such as vascular endothelial growth factor and Epo, but were ameliorated by Fe-SIH, a compound delivering iron into cells independently of iron transport proteins. Consistent with profound defects in iron homeostasis, Hif1alpha-/- yolk sac and/or embryos demonstrated aberrant mRNA levels of hepcidin, Fpn1, Irp1, and frascati. We conclude that dysregulated expression of genes encoding Epo, EpoR, and iron regulatory proteins contributes to defective erythropoiesis in Hif1alpha-/- yolk sacs. These results identify a novel role for HIF-1 in the regulation of iron homeostasis and reveal unexpected regulatory differences in Epo/EpoR signaling in yolk sac and embryonic erythropoiesis.
Collapse
Affiliation(s)
- Donghoon Yoon
- Hematology Section, University of Utah, Salt Lake City, Utah 84132, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Abu-Farha M, Niles J, Willmore WG. Erythroid-specific 5-aminolevulinate synthase protein is stabilized by low oxygen and proteasomal inhibition. Biochem Cell Biol 2006; 83:620-30. [PMID: 16234850 DOI: 10.1139/o05-045] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
5-Aminolevulinate synthase (ALAS; E.C. 2.3.1.37) catalyzes the first and rate-limiting step of heme synthesis within the mitochondria. Two isozymes of ALAS, encoded by separate genes, exist. ALAS1 is ubiquitously expressed and provides heme for cytochromes and other hemoproteins. ALAS2 is expressed exclusively in erythroid cells and synthesizes heme specifically for haemoglobin. A database search for proteins potentially regulated by oxygen tension revealed that ALAS2 contained a sequence of amino acids (LXXLAP where L is leucine, X is any amino acid, A is alanine, and P is proline) not occurring in ALAS1, which may be hydroxylated under normoxic conditions (21% O2) and target the enzyme for ubiquitination and degradation by the proteasome. We examined protein turnover of ALAS2 in the presence of cycloheximide in K562 cells. Normoxic ALAS2 had a turnover time of approximately 36 h. Hypoxia (1% O2) and inhibition of the proteasome increased both the stability and the specific activity of ALAS2 (greater than 2- and 7-fold, respectively, over 72 h of treatment). Mutation of a key proline within the LXXLAP sequence of ALAS2 also stabilized the protein beyond 36 h under normoxic conditions. The von Hippel-Lindau (vHL) protein was immunoprecipitated with FLAG epitope-tagged ALAS2 produced in normoxic cells but not in hypoxic cells, suggesting that the ALAS2 is hydroxylated under normoxic conditions and targeted for ubiquitination by the E3 ubiquitin ligase system. ALAS2 could also be ubiquitinated under normoxia using an in vitro ubiquitination assay. The present study provides evidence that ALAS2 is broken down under normoxic conditions by the proteasome and that the prolyl-4-hydroxylase/vHL E3 ubiquitin ligase pathway may be involved.
Collapse
Affiliation(s)
- Mohamed Abu-Farha
- Institute of Biochemistry, Carleton University, Ottawa, ON K1S 5B6, Canada
| | | | | |
Collapse
|
12
|
Ateghang B, Wartenberg M, Gassmann M, Sauer H. Regulation of cardiotrophin-1 expression in mouse embryonic stem cells by HIF-1alpha and intracellular reactive oxygen species. J Cell Sci 2006; 119:1043-52. [PMID: 16507596 DOI: 10.1242/jcs.02798] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Cardiomyogenesis in differentiating mouse embryonic stem (ES) cells is promoted by cardiotrophin-1 (CT-1), a member of the IL-6 interleukin superfamily that acts through the tall gp130 cytokine receptor. We show that prooxidants (menadione, hydrogen peroxide) as well as chemical (CoCl2) and physiological (1% O2) hypoxia increased CT-1 as well as HIF-1alpha protein and mRNA expression in embryoid bodies, indicating that CT-1 expression is regulated by reactive oxygen species (ROS) and hypoxia. Treatment with either prooxidants or chemical hypoxia increased gp130 phosphorylation and protein expression of NADPH oxidase subunits p22-phox, p47-phox, p67-phox, as well as Nox1 and Nox4 mRNA. Consequently, inhibition of NADPH oxidase activity by diphenylen iodonium chloride (DPI) and apocynin abolished prooxidant- and chemical hypoxia-induced upregulation of CT-1. Prooxidants and chemical hypoxia activated ERK1,2, JNK and p38 as well as PI3-kinase. The proxidant- and CoCl2-mediated upregulation of CT-1 was significantly inhibited in the presence of the ERK1,2 antagonist UO126, the JNK antagonist SP600125, the p38 antagonist SKF86002, the PI3-kinase antagonist LY294002, the Jak-2 antagonist AG490 as well as in the presence of free radical scavengers. Moreover, developing embryoid bodies derived from HIF-1alpha-/- ES cells lack cardiomyogenesis, and prooxidants as well as chemical hypoxia failed to upregulate CT-1 expression. Our results demonstrate that CT-1 expression in ES cells is regulated by ROS and HIF-1alpha and imply a crucial role of CT-1 in the survival and proliferation of ES-cell-derived cardiac cells.
Collapse
|
13
|
Scholz D, Schaper W. Enhanced arteriogenesis in mice overexpressing erythropoietin. Cell Tissue Res 2006; 324:395-401. [PMID: 16485134 DOI: 10.1007/s00441-005-0072-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2005] [Accepted: 07/27/2005] [Indexed: 11/28/2022]
Abstract
After permanent occlusion of the femoral artery, the survival of ischemic limb tissue depends on collateral artery growth (arteriogenesis). In previous work, we have shown that shear stress triggers arteriogenesis. To test whether increased shear stress results in enhanced arteriogenesis, we compared arteriogenesis in transgenic mice overexpressing erythropoietin (EPO), which possessed increased blood viscosity through the higher hematocrit (thereby providing increased shear stress), with wild-type mice. The right femoral artery was occluded proximal to the origin of the arteria poplitea. Distal blood flow was assessed by laser Doppler imaging, and the growth and remodeling of collateral arteries was examined by light and electron microscopy and morphometry. After occlusion of the femoral artery, EPO mice demonstrated enhanced arteriogenesis: their collateral arteries developed a 1.7-fold diameter and a 2-fold wall thickness compared with wild-type. However, the blood flow recovery in EPO mice was markedly retarded. Structural remodeling and growth of collateral arteries was markedly enhanced in EPO mice, presumably as a result of increased blood viscosity and shear stress.
Collapse
Affiliation(s)
- Dimitri Scholz
- Department of Exp. Cardiology, Max Planck Institute, Benekestrasse 2, 61231 Bad Nauheim, Germany.
| | | |
Collapse
|
14
|
Land SC. Hochachka's "Hypoxia Defense Strategies" and the development of the pathway for oxygen. Comp Biochem Physiol B Biochem Mol Biol 2005; 139:415-33. [PMID: 15544965 DOI: 10.1016/j.cbpc.2004.02.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 02/26/2004] [Accepted: 02/29/2004] [Indexed: 12/17/2022]
Abstract
Hochachka's "Hypoxia Defense Strategies" identify oxygen signalling, metabolic arrest, channel arrest and coordinated suppression of ATP turnover rates as key factors that determine the ability of organisms to survive exposure to chronic hypoxia. In this review, I assess the developmental role played by these phenomena in the morphogenesis of the gas exchange tissues that define the pathway for oxygen transport to cytochrome c oxidase. Key areas of regulation lie in: (I) the suppression of fetal mitochondrial oxidative function in hand with mitochondrial biogenesis (metabolic arrest), (II) the role of hypoxia-driven oxygen signalling pathways in directing the scope of non-differentiated stem cell proliferation in placenta and lung development and (III) the regulation of epithelial fluid secretion/absorption in the lung through the oxygen-dependent modulation of Na+ conductance pathways. The identification of developmental roles for Hochachka's "Hypoxia Defense Strategies" in directing the morphogenesis of gas exchange structures bears with it the implication that these strategies are fundamental to establishing the scope for aerobic metabolic performance throughout life.
Collapse
Affiliation(s)
- Stephen C Land
- Division of Maternal and Child Health Sciences, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, Scotland, UK.
| |
Collapse
|
15
|
Narayan AD, Ersek A, Campbell TA, Colón DM, Pixley JS, Zanjani ED. The effect of hypoxia and stem cell source on haemoglobin switching. Br J Haematol 2005; 128:562-70. [PMID: 15686468 DOI: 10.1111/j.1365-2141.2004.05336.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
This study investigated whether relative changes that accompany the naturally occurring shifts in haematopoietic sites during human development play a role in haemoglobin (Hb) switching or whether Hb switching is innately programmed into cells. CD34(+)/Lineage(-) haematopoietic stem/progenitor cells (HSCs) were isolated from human fetal liver (F-LVR), cord blood (CB), and adult bone marrow (ABM), and the Hb was characterized by flow cytometry on cultures that generated enucleated red cells. All feeder layers (stroma from F-LVR, ABM, and human fetal aorta) enhanced cell proliferation and erythropoiesis but did not affect Hb type. HSCs from CB and F-LVR generated the same Hb profile under normoxia and hypoxia. HSCs from ABM had single-positive HbA and double-positive HbA and HbF cells at normoxia and almost entirely double-positive cells at hypoxia. Further characterization of these ABM cultures was determined by following mRNA expression for the transcription factors erythroid Kruppel-like factor (EKLF) and fetal Kruppel-like factor (FKLF) as a function of time in cultures under hypoxia and normoxia. The erythroid-specific isoform of 5-amino-levulinate synthase (ALAS2) was also expressed under hypoxic conditions. We conclude that Hb switching is affected by the environment but not all HSCs are preprogrammed to respond.
Collapse
Affiliation(s)
- A Daisy Narayan
- Department of Animal Biotechnology, University of Nevada, Reno, NV 89557, USA.
| | | | | | | | | | | |
Collapse
|
16
|
Abstract
Throughout gestation, low oxygen tensions are a dominant feature of the fetal environment and so may be important in sustaining a normal pattern of lung morphogenesis until the moment of birth. As breathing begins, the equilibration of the lung lumen to postnatal PO2 evokes a series of physiologic and morphogenic maturation events that are partially reversible by hypoxia. In this review, we discuss the experimental evidence that fetal and perinatal oxygen tensions differently influence lung morphogenesis through oxygen- and redox-responsive signaling pathways and identify five loci at which this regulation may occur: (I) proliferation of undifferentiated lung mesenchyme as governed by hypoxia-regulated transcription factors (HIF-1alpha, C/EBPbeta); (II) transient production of reactive oxygen species (ROS) and nuclear oxidation of the perinatal lung epithelium; (III) nuclear transport and oxidation of thioredoxin in hand with the acute activation of nuclear factor- kappaB (NF-kappaB); (IV) ROS-evoked chronic rise in intracellular glutathione and thioredoxin redox buffering capacity; and (V) NF-kappaB-dependent increase in transepithelial Na+ transport and lung lumenal fluid clearance. Although not exhaustive, this analysis leads us to the conclusion that redox events that occur in the lung during gestation, parturition, and the early neonatal period may dramatically influence the expression of genes and physiological events that are crucial to the successful transition from fetal to postnatal lung maturation.
Collapse
Affiliation(s)
- Stephen C Land
- Maternal and Child Health Sciences, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland, UK.
| | | |
Collapse
|
17
|
Greenlee AR, Kronenwetter-Koepel TA, Kaiser SJ, Ellis TM, Liu K. Combined effects of MatrigelTM and growth factors on maintaining undifferentiated murine embryonic stem cells for embryotoxicity testing. Toxicol In Vitro 2004; 18:543-53. [PMID: 15130612 DOI: 10.1016/j.tiv.2004.01.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2003] [Accepted: 01/28/2004] [Indexed: 10/26/2022]
Abstract
Undifferentiated, murine embryonic stem (mES) cells have shown promise as a substrate for identifying embryotoxic chemicals and for studying mechanisms of early developmental injury. However, long-term maintenance of mES cells in an undifferentiated state is problematic. The present study evaluates the combination of Matrigel matrix and three growth factors for this purpose. Biomarkers of mES cell pluripotency, apoptosis, chromosome number and cardiomyocyte differentiation were monitored over 119 population doublings. D3 mES cells retained undifferentiated characteristics, including sustained expression of alkaline phosphatase and stage specific embryonic antigen-1 (SSEA-1) and continued transcription of Pou5f1 (Oct-4). Cell viability remained at > or=95% and population-doubling times averaged 14.3 h over 10 weeks of observation. Caspase-3 activation, a marker of cellular death by apoptosis, was measured in early- and late-passage mES cells. Early-passage cells showed dose-responsive caspase-3 activation following exposure to sodium arsenite, whereas caspase-3 activation of late-passage cells dropped to background levels at toxicant dosages above 50 ppb. Aneuploidy and impaired differentiation into beating cardiomyocytes were noted for late-passage mES cells. Matrigel, combined with growth factors, may sustain undifferentiated mES cells. However, aneuploidy, reduced caspase-3 activation, and inability to differentiate suggests further modifications to the culture system may be needed for long-term propagation of cells for embryotoxicity endpoints.
Collapse
Affiliation(s)
- A R Greenlee
- National Farm Medicine Center, Marshfield Clinic Research Foundation, 1000 North Oak Avenue, Marshfield, WI 54449, USA.
| | | | | | | | | |
Collapse
|
18
|
Quaschning T, Ruschitzka F, Stallmach T, Shaw S, Morawietz H, Goettsch W, Hermann M, Slowinski T, Theuring F, Hocher B, Lüscher TF, Gassmann M. Erythropoietin-induced excessive erythrocytosis activates the tissue endothelin system in mice. FASEB J 2003; 17:259-61. [PMID: 12490547 DOI: 10.1096/fj.02-0296fje] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The endothelium controls blood flow and pressure by releasing several vasoactive factors, among them the vasodilator nitric oxide (NO) and the potent vasoconstrictor endothelin-1 (ET-1). Although increased NO levels have been found in excessive erythrocytosis, little is known concerning ET-1 expression in this condition. Thus, we examined the endothelin system in transgenic mice that due to constitutive overexpression of erythropoietin (Epo) reached hematocrit levels of approximately 80%. Surprisingly, despite generalized vasodilatation, polycythemic mice exhibited a two- to fivefold elevation in ET-1 mRNA levels in aorta, liver, heart, and kidney. In line with this, increased expression of ET-1 protein was detected in the pulmonary artery by immunohistochemical analysis. Compared with their wild-type littermates, aortic rings of Epo transgenic animals exhibited a marked reduction in vascular reactivity to ET-1 and big ET-1, but this effect was abrogated upon preincubation with the NO synthase inhibitor N-nitro-L-arginine methyl ester (L-NAME). Pretreatment of polycythemic mice with the ET(A) receptor antagonist darusentan for 3 wk significantly prolonged their survival upon acute exposure to L-NAME. Taken together, these results demonstrate for the first time that excessive erythrocytosis induces a marked activation of the tissue endothelin system that results in increased mortality upon blockade of NO-mediated vasodilatation. Because ETA antagonism prolonged survival after acute blockade of NO synthesis, endothelin may be regarded as a contributor to the adverse cardiovascular effects of erythrocytosis and may thus represent a new target in the treatment of cardiovascular disease associated with erythrocytosis.
Collapse
Affiliation(s)
- Thomas Quaschning
- Cardiology and Cardiovascular Research and Institute of Physiology, University of Zürich, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Hofer T, Wenger RH, Kramer MF, Ferreira GC, Gassmann M. Hypoxic up-regulation of erythroid 5-aminolevulinate synthase. Blood 2003; 101:348-50. [PMID: 12393745 DOI: 10.1182/blood-2002-03-0773] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The erythroid-specific isoform of 5-aminolevulinate synthase (ALAS2) catalyzes the rate-limiting step in heme biosynthesis. The hypoxia-inducible factor-1 (HIF-1) transcriptionally up-regulates erythropoietin, transferrin, and transferrin receptor, leading to increased erythropoiesis and hematopoietic iron supply. To test the hypothesis that ALAS2 expression might be regulated by a similar mechanism, we exposed murine erythroleukemia cells to hypoxia (1% O(2)) and found an up to 3-fold up-regulation of ALAS2 mRNA levels and an increase in cellular heme content. A fragment of the ALAS2 promoter ranging from -716 to +1 conveyed hypoxia responsiveness to a heterologous luciferase reporter gene construct in transiently transfected HeLa cells. In contrast, iron depletion, known to induce HIF-1 activity but inhibit ALAS2 translation, did not increase ALAS2 promoter activity. Mutation of a previously predicted HIF-1-binding site (-323/-318) within this promoter fragment and DNA-binding assays revealed that hypoxic up-regulation is independent of this putative HIF-1 DNA-binding site.
Collapse
Affiliation(s)
- Thomas Hofer
- Institute of Veterinary Physiology, University of Zurich, Switzerland
| | | | | | | | | |
Collapse
|
20
|
Scheid A, Wenger RH, Schäffer L, Camenisch I, Distler O, Ferenc A, Cristina H, Ryan HE, Johnson RS, Wagner KF, Stauffer UG, Bauer C, Gassmann M, Meuli M. Physiologically low oxygen concentrations in fetal skin regulate hypoxia-inducible factor 1 and transforming growth factor-beta3. FASEB J 2002; 16:411-3. [PMID: 11790723 DOI: 10.1096/fj.01-0496fje] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
In the first-trimester mammalian fetus, skin wounds heal with perfect reconstitution of the dermal architecture without scar formation. Understanding environmental molecular regulation in fetal wound healing may reveal scar-limiting therapeutical strategies for the prevention of postnatal scarring wound repair. Therefore, we performed studies on fetal skin oxygenation and skin and wound expression of hypoxia-inducible factor 1alpha (HIF-1alpha) in the sheep model in vivo and performed studies on the potential relevance of HIF-1alpha during wound healing in vitro. Skin oxygen partial pressure levels were hypoxic throughout normal development. In nonscarring fetal skin at gestation day (GD)60, HIF-1alpha could be detected neither in healthy nor in wounded tissue. At GD100, in wounds with minimal scar formation, HIF-1alpha was expressed in fibroblasts and was markedly up-regulated at the wound edge. In scarring fetal wounds at GD120, HIF-1alpha was predominantly expressed in inflammatory cells. Expression of transforming growth factor beta3 (TGF-beta3), a potent antiscarring cytokine, overlapped with HIF-1a expression at GD100. HIF-1alpha-deficient mouse embryonic fibroblasts showed impaired migratory capabilities and demonstrated that TGF-beta3, but not proscarring TGF-beta1, manifests hypoxia- and HIF-1alpha-dependent regulation. In conclusion, HIF-1alpha-dependent regulation of a potent antiscarring cytokine may provide new strategies for antiscarring manipulation of wound healing.
Collapse
Affiliation(s)
- Annette Scheid
- Department of Surgery, University Children's Hospital of Zürich, Switzerland.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Ramírez-Bergeron DL, Simon MC. Hypoxia-inducible factor and the development of stem cells of the cardiovascular system. Stem Cells 2002; 19:279-86. [PMID: 11463947 DOI: 10.1634/stemcells.19-4-279] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Decreased oxygen (O2) levels activate hypoxia-inducible factor (HIF-1) to induce genes involved in glycolysis, glucose transport, erythropoiesis, and angiogenesis. Mutations in various HIF-1 subunits have contributed to our understanding of the role hypoxia plays during early embryonic development in general and the cardiovascular system in particular. We propose that HIF-1 is important for the generation, proliferation, maintenance, and differentiation of the early cardiovascular system. Understanding aberrations in these hypoxic responses is important since they contribute to serious human disease such as ischemia and tumorigenesis. In this review we will focus on the critical role of O2 in regulating cardiovascular events during early embryonic development.
Collapse
Affiliation(s)
- D L Ramírez-Bergeron
- Abramson Family Cancer Research Institute, Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | | |
Collapse
|
22
|
Sun Y, Jin K, Mao XO, Zhu Y, Greenberg DA. Neuroglobin is up-regulated by and protects neurons from hypoxic-ischemic injury. Proc Natl Acad Sci U S A 2001; 98:15306-11. [PMID: 11742077 PMCID: PMC65025 DOI: 10.1073/pnas.251466698] [Citation(s) in RCA: 380] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Globins are oxygen-binding heme proteins present in bacteria, protists, fungi, plants, and animals. Their functions have diverged widely in evolution, and include binding, transport, scavenging, detoxification, and sensing of gases like oxygen, nitric oxide, and carbon monoxide. Neuroglobin (Ngb) is a recently discovered monomeric globin with high affinity for oxygen and preferential localization to vertebrate brain. No function for Ngb is known, but its affinity for oxygen and its expression in cerebral neurons suggest a role in neuronal responses to hypoxia or ischemia. Here we report that Ngb expression is increased by neuronal hypoxia in vitro and focal cerebral ischemia in vivo, and that neuronal survival after hypoxia is reduced by inhibiting Ngb expression with an antisense oligodeoxynucleotide and enhanced by Ngb overexpression. Both induction of Ngb and its protective effect show specificity for hypoxia over other stressors. We conclude that hypoxia-inducible Ngb expression helps promote neuronal survival from hypoxic-ischemic insults.
Collapse
Affiliation(s)
- Y Sun
- Buck Institute for Age Research, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | | | | | | | | |
Collapse
|
23
|
Ruschitzka FT, Wenger RH, Stallmach T, Quaschning T, de Wit C, Wagner K, Labugger R, Kelm M, Noll G, Rülicke T, Shaw S, Lindberg RL, Rodenwaldt B, Lutz H, Bauer C, Lüscher TF, Gassmann M. Nitric oxide prevents cardiovascular disease and determines survival in polyglobulic mice overexpressing erythropoietin. Proc Natl Acad Sci U S A 2000; 97:11609-13. [PMID: 11027359 PMCID: PMC17248 DOI: 10.1073/pnas.97.21.11609] [Citation(s) in RCA: 198] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Nitric oxide (NO) induces vasodilatatory, antiaggregatory, and antiproliferative effects in vitro. To delineate potential beneficial effects of NO in preventing vascular disease in vivo, we generated transgenic mice overexpressing human erythropoietin. These animals induce polyglobulia known to be associated with a high incidence of vascular disease. Despite hematocrit levels of 80%, adult transgenic mice did not develop hypertension or thromboembolism. Endothelial NO synthase levels, NO-mediated endothelium-dependent relaxation and circulating and vascular tissue NO levels were markedly increased. Administration of the NO synthase inhibitor N(G)-nitro-L-arginine methyl ester (L-NAME) led to vasoconstriction of peripheral resistance vessels, hypertension, and death of transgenic mice, whereas wild-type siblings developed hypertension but did not show increased mortality. L-NAME-treated polyglobulic mice revealed acute left ventricular dilatation and vascular engorgement associated with pulmonary congestion and hemorrhage. In conclusion, we here unequivocally demonstrate that endothelial NO maintains normotension, prevents cardiovascular dysfunction, and critically determines survival in vivo under conditions of increased hematocrit.
Collapse
Affiliation(s)
- F T Ruschitzka
- Institute of Physiology, University of Zürich-Irchel, CH-8057 Zürich, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|