Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Hypoxia (Auckl) ; 4: 29-39, 2016.
Article in English | MEDLINE | ID: mdl-27800506

ABSTRACT

Despite its well-known role in red blood cell production, it is now accepted that erythropoietin (Epo) has other physiological functions. Epo and its receptors are expressed in many tissues, such as the brain and heart. The presence of Epo/Epo receptors in these organs suggests other roles than those usually assigned to this protein. Thus, the aim of this review is to describe the effects of Epo deficiency on adaptation to normoxic and hypoxic environments and to suggest a key role of Epo on main physiological adaptive functions. Our original model of Epo-deficient (Epo-TAgh) mice allowed us to improve our knowledge of the possible role of Epo in O2 homeostasis. The use of anemic transgenic mice revealed Epo as a crucial component of adaptation to hypoxia. Epo-TAgh mice survive well in hypoxic conditions despite low hematocrit. Furthermore, Epo plays a key role in neural control of ventilatory acclimatization and response to hypoxia, in deformability of red blood cells, in cerebral and cardiac angiogenesis, and in neuro- and cardioprotection.

2.
Respir Physiol Neurobiol ; 186(2): 146-54, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23333855

ABSTRACT

The involvement of erythropoietin in cardiac adaptation to acute and chronic (CHx) hypoxia was investigated in erythropoietin deficient transgenic (Epo-TAg(h)) and wild-type (WT) mice. Left (LV) and right ventricular functions were assessed by echocardiography and hemodynamics. HIF-1α, VEGF and Epo pathways were explored through RT-PCR, ELISA, Western blot and immunocytochemistry. Epo gene and protein were expressed in cardiomyocytes of WT mice in normoxia and hypoxia. Increase in blood hemoglobin, angiogenesis and functional cardiac adaptation occurred in CHx in WT mice, allowing a normal oxygen delivery (O2T). Epo deficiency induced LV hypertrophy, increased cardiac output (CO) and angiogenesis, but O2T remained lower than in WT mice. In CHx Epo-TAg(h) mice, LV hypertrophy, CO and O2T decreased. HIF-1α and Epo receptor pathways were depressed, suggesting that Epo-TAg(h) mice could not adapt to CHx despite activation of cardioprotective pathways (increased P-STAT-5/STAT-5). HIF/Epo pathway is activated in the heart of WT mice in hypoxia. Chronic hypoxia induced cardiac adaptive responses that were altered with Epo deficiency, failing to maintain oxygen delivery to tissues.


Subject(s)
Adaptation, Physiological/physiology , Erythropoietin/metabolism , Hypoxia/metabolism , Myocytes, Cardiac/metabolism , Animals , Blotting, Western , Chronic Disease , Disease Models, Animal , Echocardiography , Enzyme-Linked Immunosorbent Assay , Heart/physiology , Hemodynamics , Male , Mice , Mice, Transgenic , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
3.
J Biomed Biotechnol ; 2010: 137817, 2010.
Article in English | MEDLINE | ID: mdl-20414335

ABSTRACT

Erythropoietin (Epo) and vascular growth factor (VEGF) are known to be involved in the regulation of cellular activity when oxygen transport is reduced as in anaemia or hypoxic conditions. Because it has been suggested that Epo could play a role in skeletal muscle development, regeneration, and angiogenesis, we aimed to assess Epo deficiency in both normoxia and hypoxia by using an Epo-deficient transgenic mouse model (Epo-TAg(h)). Histoimmunology, ELISA and real time RT-PCR did not show any muscle fiber atrophy or accumulation of active HIF-1alpha but an improvement of microvessel network and an upregulation of VEGFR2 mRNA in Epo-deficient gastrocnemius compared with Wild-Type one. In hypoxia, both models exhibit an upregulation of VEGF120 and VEGFR2 mRNA but no accumulation of Epo protein. EpoR mRNA is not up-regulated in both Epo-deficient and hypoxic gastrocnemius. These results suggest that muscle deconditioning observed in patients suffering from renal failure is not due to Epo deficiency.


Subject(s)
Erythropoietin/physiology , Hypoxia/metabolism , Muscle Fibers, Skeletal/physiology , Analysis of Variance , Animals , Erythropoietin/blood , Erythropoietin/genetics , Erythropoietin/metabolism , Histocytochemistry , Hypoxia/genetics , Hypoxia-Inducible Factor 1/metabolism , Male , Mice , Mice, Transgenic , Microvessels/growth & development , Microvessels/metabolism , Muscle Fibers, Skeletal/chemistry , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/blood supply , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , Muscular Atrophy , Neovascularization, Physiologic/physiology , Receptors, Erythropoietin/metabolism , Sarcomeres , Statistics, Nonparametric , Up-Regulation , Vascular Endothelial Growth Factors/genetics , Vascular Endothelial Growth Factors/metabolism
4.
Am J Physiol Regul Integr Comp Physiol ; 296(3): R801-11, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19109375

ABSTRACT

Anemia and hypoxia in rats result in an increase in factors potentially involved in cerebral angiogenesis. Therefore, the aim of this study was to assess the effect of chronic anemia and/or chronic hypoxia on cerebral cellular responses and angiogenesis in wild-type and anemic transgenic mice. These studies were done in erythropoietin-deficient mice (Epo-TAg(h)) in normoxia and following acute (one day) and chronic (14 days, barometric pressure = 420 mmHg) hypoxia. In normoxia, Epo-TAg(h) mice showed an increase in transcript and protein levels of hypoxia-inducible factor 1alpha (HIF-1alpha), vascular endothelial growth factor (VEGF), erythropoietin receptors (EpoR), phospho-STAT-5/STAT-5 ratio, and neuronal neuronal nitric oxide synthase (nNOS) along with a higher cerebral capillary density. In wild-type (WT) mice, acute hypoxia increased all of the studied factors, while in chronic hypoxia, HIF-1alpha, EpoR, phospho-STAT-5/STAT-5 ratio, nNOS, and inducible NOS remained elevated, with an increase in capillary density. Surprisingly, in Epo-TAg(h) mice, chronic hypoxia did not further increase any factor except the nitric oxide metabolites, while HIF-1alpha, EpoR, and phospho-STAT-5/STAT-5 ratio were reduced. Normoxic Epo-TAg(h) mice developed cerebral angiogenesis through the HIF-1alpha/VEGF pathway. In acute hypoxia, WT mice up-regulated all of the studied factors, including cerebral NO. Polycythemia and angiogenesis occurred with acclimatization to chronic hypoxia only in WT mice. In Epo-TAg(h), the decrease in HIF-1alpha, VEGF proteins, and phospho-STAT-5 ratio in chronic hypoxia suggest that neuroprotective and angiogenesis pathways are altered.


Subject(s)
Anemia/physiopathology , Brain/physiopathology , Erythropoietin/deficiency , Erythropoietin/genetics , Hypoxia/physiopathology , Animals , Body Weight/physiology , Cerebral Cortex/metabolism , Chronic Disease , Erythropoietin/metabolism , Hemoglobins/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunoassay , Immunohistochemistry , Male , Mice , Mice, Inbred CBA , Mice, Knockout , Nitric Oxide/metabolism , RNA/biosynthesis , RNA/isolation & purification , Receptors, Erythropoietin/biosynthesis , Receptors, Erythropoietin/genetics , Reverse Transcriptase Polymerase Chain Reaction , STAT5 Transcription Factor/metabolism , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...