Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Am J Physiol Renal Physiol ; 314(4): F501-F516, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29187371

ABSTRACT

The erythropoietin receptor (EpoR) is widely expressed but its renoprotective action is unexplored. To examine the role of EpoR in vivo in the kidney, we induced acute kidney injury (AKI) by ischemia-reperfusion in mice with different EpoR bioactivities in the kidney. EpoR bioactivity was reduced by knockin of wild-type human EpoR, which is hypofunctional relative to murine EpoR, and a renal tubule-specific EpoR knockout. These mice had lower EPO/EpoR activity and lower autophagy flux in renal tubules. Upon AKI induction, they exhibited worse renal function and structural damage, more apoptosis at the acute stage (<7 days), and slower recovery with more tubulointerstitial fibrosis at the subacute stage (14 days). In contrast, mice with hyperactive EpoR signaling from knockin of a constitutively active human EpoR had higher autophagic flux, milder kidney damage, and better renal function at the acute stage but, surprisingly, worse tubulointerstitial fibrosis and renal function at the subacute stage. Either excess or deficient EpoR activity in the kidney was associated with abnormal peritubular capillaries and tubular hypoxia, creating a "U-shaped" relationship. The direct effects of EpoR on tubular cells were confirmed in vitro by a hydrogen peroxide model using primary cultured proximal tubule cells with different EpoR activities. In summary, normal erythropoietin (EPO)/EpoR signaling in renal tubules provides defense against renal tubular injury maintains the autophagy-apoptosis balance and peritubular capillary integrity. High and low EPO/EpoR bioactivities both lead to vascular defect, and high EpoR activity overides the tubular protective effects in AKI recovery.


Subject(s)
Acute Kidney Injury/metabolism , Capillaries/metabolism , Erythropoietin/metabolism , Kidney Tubules, Proximal/blood supply , Kidney Tubules, Proximal/metabolism , Neovascularization, Physiologic , Receptors, Erythropoietin/metabolism , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Acute Kidney Injury/physiopathology , Animals , Apoptosis , Autophagy , Capillaries/pathology , Capillaries/physiopathology , Cell Hypoxia , Cells, Cultured , Disease Models, Animal , Fibrosis , Humans , Kidney Tubules, Proximal/pathology , Kidney Tubules, Proximal/physiopathology , Mice, 129 Strain , Mice, Transgenic , Receptors, Erythropoietin/deficiency , Receptors, Erythropoietin/genetics , Signal Transduction
2.
Am J Physiol Regul Integr Comp Physiol ; 308(4): R330-5, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25519735

ABSTRACT

Under conditions of accelerated erythropoiesis, elevated erythropoietin (Epo) levels are associated with inhibition of hepcidin synthesis, a response that ultimately increases iron availability to meet the enhanced iron needs of erythropoietic cells. In the search for erythroid regulators of hepcidin, many candidates have been proposed, including Epo itself. We aimed to test whether direct interaction between Epo and the liver is required to regulate hepcidin. We found that prolonged administration of high doses of Epo in mice leads to great inhibition of liver hepcidin mRNA levels, and concomitant induction of the hepcidin inhibitor erythroferrone (ERFE). Epo treatment also resulted in liver iron mobilization, mediated by increased ferroportin activity and accompanied by reduced ferritin levels and increased TfR1 expression. The same inhibitory effect was observed in mice that do not express the homodimeric Epo receptor (EpoR) in liver cells because EpoR expression is restricted to erythroid cells. Similarly, liver signaling pathways involved in hepcidin regulation were not influenced by the presence or absence of hepatic EpoR. Moreover, Epo analogs, possibly interacting with the postulated heterodimeric ß common EpoR, did not affect hepcidin expression. These findings were supported by the lack of inhibition on hepcidin found in hepatoma cells exposed to various concentrations of Epo for different periods of times. Our results demonstrate that hepcidin suppression does not require the direct binding of Epo to its liver receptors and rather suggest that the role of Epo is to stimulate the synthesis of the erythroid regulator ERFE in erythroblasts, which ultimately downregulates hepcidin.


Subject(s)
Erythropoietin/analogs & derivatives , Hepcidins/metabolism , Liver/drug effects , Oligopeptides/pharmacology , Animals , Cytokines/metabolism , Dose-Response Relationship, Drug , Down-Regulation , Erythropoietin/pharmacology , Hep G2 Cells , Hepcidins/genetics , Humans , Iron/metabolism , Liver/metabolism , Male , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Muscle Proteins/metabolism , RNA, Messenger/metabolism , Receptors, Erythropoietin/deficiency , Receptors, Erythropoietin/genetics , Time Factors
3.
PLoS One ; 8(7): e68083, 2013.
Article in English | MEDLINE | ID: mdl-23861852

ABSTRACT

Erythropoietin (Epo) is a cytokine that binds and activates an Epo receptor (EpoR) expressed on the surface of erythroid progenitor cells to promote erythropoiesis. While early studies suggested EpoR transcripts were expressed exclusively in the erythroid compartment, low-level EpoR transcripts were detected in nonhematopoietic tissues and tumor cell lines using sensitive RT-PCR methods. However due to the widespread use of nonspecific anti-EpoR antibodies there are conflicting data on EpoR protein expression. In tumor cell lines and normal human tissues examined with a specific and sensitive monoclonal antibody to human EpoR (A82), little/no EpoR protein was detected and it was not functional. In contrast, EpoR protein was reportedly detectable in a breast tumor cell line (MCF-7) and breast cancer tissues with an anti-EpoR polyclonal antibody (M-20), and functional responses to rHuEpo were reported with MCF-7 cells. In another study, a functional response was reported with the lung tumor cell line (NCI-H838) at physiological levels of rHuEpo. However, the specificity of M-20 is in question and the absence of appropriate negative controls raise questions about possible false-positive effects. Here we show that with A82, no EpoR protein was detectable in normal human and matching cancer tissues from breast, lung, colon, ovary and skin with little/no EpoR in MCF-7 and most other breast and lung tumor cell lines. We show further that M-20 provides false positive staining with tissues and it binds to a non-EpoR protein that migrates at the same size as EpoR with MCF-7 lysates. EpoR protein was detectable with NCI-H838 cells, but no rHuEpo-induced phosphorylation of AKT, STAT3, pS6RP or STAT5 was observed suggesting the EpoR was not functional. Taken together these results raise questions about the hypothesis that most tumors express high levels of functional EpoR protein.


Subject(s)
Neoplasm Proteins/genetics , Neoplasms/genetics , Receptors, Erythropoietin/analysis , Transcription Factors/genetics , Animals , Biopsy , Cell Line, Tumor , False Positive Reactions , Female , Gene Expression , Humans , Immunoassay , Male , Mice , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Phosphorylation , Receptors, Erythropoietin/deficiency , Receptors, Erythropoietin/genetics , Sensitivity and Specificity , Transcription Factors/metabolism
4.
FASEB J ; 26(7): 2847-58, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22490927

ABSTRACT

Erythropoietin acts by binding to its cell surface receptor on erythroid progenitor cells to stimulate erythrocyte production. Erythropoietin receptor expression in nonhematopoietic tissue, including skeletal muscle progenitor cells, raises the possibility of a role for erythropoietin beyond erythropoiesis. Mice with erythropoietin receptor restricted to hematopoietic tissue were used to assess contributions of endogenous erythropoietin to promote skeletal myoblast proliferation and survival and wound healing in a mouse model of cardiotoxin induced muscle injury. Compared with wild-type controls, these mice had fewer skeletal muscle Pax-7(+) satellite cells and myoblasts that do not proliferate in culture, were more susceptible to skeletal muscle injury and reduced maximum load tolerated by isolated muscle. In contrast, mice with chronic elevated circulating erythropoietin had more Pax-7(+) satellite cells and myoblasts with increased proliferation and survival in culture, decreased muscle injury, and accelerated recovery of maximum load tolerated by isolated muscle. Skeletal muscle myoblasts also produced endogenous erythropoietin that increased at low O(2). Erythropoietin promoted proliferation, survival, and wound recovery in myoblasts via the phosphoinositide 3-kinase/AKT pathway. Therefore, endogenous and exogenous erythropoietin contribute to increasing satellite cell number following muscle injury, improve myoblast proliferation and survival, and promote repair and regeneration in this mouse induced muscle injury model independent of its effect on erythrocyte production.


Subject(s)
Erythropoietin/physiology , Muscle, Skeletal/injuries , Muscle, Skeletal/physiopathology , Animals , Apoptosis , Cell Hypoxia , Cells, Cultured , Erythropoietin/administration & dosage , Erythropoietin/genetics , Female , GATA3 Transcription Factor/biosynthesis , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Muscle, Skeletal/drug effects , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/physiology , PAX7 Transcription Factor/physiology , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Erythropoietin/deficiency , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/physiology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/physiology , Signal Transduction/drug effects , Wound Healing/drug effects , Wound Healing/physiology
5.
Arch Pediatr ; 17 Suppl 3: S78-84, 2010 Sep.
Article in French | MEDLINE | ID: mdl-20728813

ABSTRACT

Erythropoietin (Epo) has long been recognised for its role in the control of erythropoiesis and therefore in the treatment of anemia including anemia of prematurity. The erythropoietin receptor (Epo-R) though is expressed in many other organs including the CNS. This review focuses on the role of erythropoietin during the development of the CNS and its potential role as a neuroprotective agent. Epo-R is expressed in many different cellules of the CNS during development including neural progenitor cells, neurons, astrocytes and oligodendrocytes. In the event of hypoxia CNS cells respond with increase of erythropoietin release with subsequent stimulation of neurogenesis through Epo-R on neural progenitor cells. In an Epo-R knock-out model therefore cerebral development is severely impaired. In models of hypoxia-ischemia exogenous Epo has been shown to reduce lesion size and improve structural and functional recovery. Human studies are emerging using Epo as a neuroprotective agent both for the term infant with hypoxia-ischemia as well as for the extremely preterm infant.


Subject(s)
Erythropoietin/therapeutic use , Infant, Premature, Diseases/drug therapy , Brain/growth & development , Gene Knockout Techniques , Humans , Infant, Newborn , Infant, Premature, Diseases/genetics , Neuroprotective Agents/therapeutic use , Receptors, Erythropoietin/deficiency , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/physiology
6.
Br J Pharmacol ; 160(8): 2085-96, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20649603

ABSTRACT

BACKGROUND AND PURPOSE: Darbepoetin, a long-acting erythropoietin derivative, attenuates cardiomyocyte apoptosis and improves short-term (3 days) cardiac function, but the mechanisms responsible are unknown. We investigated potential mechanisms by which darbepoetin exerts cardioprotection following myocardial infarction in mice and the significance of the erythropoietin receptor (EPOR)-common beta-chain (c-beta-chain) heteroreceptor. EXPERIMENTAL APPROACH: Mice underwent 60 min coronary occlusion followed by treatment with vehicle or a single dose of darbepoetin. Effects on gene expression, apoptosis and neutrophil accumulation in infarcted left ventricle were assessed 24 h later. Cardiac function, effects on vascularization and fibrosis were assessed 28 days later. The significance of EPOR-c-beta-chain heteroreceptor was examined 28 days after infarction using mice deficient in c-beta-chain. KEY RESULTS: Twenty-four hours after darbepoetin, mRNAs encoding haeme oxygenase-1 (HO-1), iNOS and brain natriuretic peptide (BNP) were markedly elevated only in infarcted regions, and the frequency of apoptotic cells attenuated. Inflammation was also attenuated with reductions in neutrophil numbers. Darbepoetin also elevated mRNAs encoding angiogenic factors: placental growth factor, monocyte chemoattractant protein-1 and interleukin-1beta. Twenty-eight days after treatment, CD31+ vessels in the infarct zone doubled and fibrosis reduced. Cardiac haemodynamics were improved. Darbepoetin also improved cardiac haemodynamics in c-beta-chain-deficient mice, increased HO-1 and iNOS expression and vessel numbers and attenuated fibrosis. CONCLUSIONS AND IMPLICATIONS: Darbepoetin stimulates expression of haeme oxygenase, iNOS, BNP and angiogenic factors specifically in infarcted left ventricle that attenuates inflammation, apoptosis and fibrosis; elevate vessel numbers; and improve cardiac function. The EPOR-c-beta-chain heteroreceptor is not essential for these effects.


Subject(s)
Cardiotonic Agents/pharmacology , Erythropoietin/analogs & derivatives , Hemodynamics/drug effects , Myocardial Infarction/diet therapy , Myocardium/pathology , Receptors, Erythropoietin/drug effects , Ventricular Function, Left/drug effects , Animals , Apoptosis/drug effects , Darbepoetin alfa , Disease Models, Animal , Dose-Response Relationship, Drug , Erythropoietin/pharmacology , Fibrosis , Gene Expression Regulation/drug effects , Heme Oxygenase-1/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/metabolism , Natriuretic Peptide, Brain/genetics , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/genetics , Neutrophil Infiltration/drug effects , Nitric Oxide Synthase Type II/genetics , Receptors, Erythropoietin/deficiency , Receptors, Erythropoietin/genetics , Time Factors
7.
Mol Cancer Res ; 7(7): 1150-7, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19567780

ABSTRACT

Erythropoietin receptors have been identified on a variety of cancer-derived cell lines and primary cancer cells, including those of prostate cancer. The functional status of these extrahematopoietic erythropoietin receptors remains a matter of some dispute. The publication of several important clinical trials suggesting a direct effect of erythropoietin on the growth and survival of primary tumors adds further importance to the question of whether erythropoietin receptors on cancer cells are functional. We have reported previously that human prostate cancer cell lines and primary prostate cancer cells express functional erythropoietin receptors that respond to exogenous erythropoietin by increased cell proliferation and STAT5 phosphorylation. We now show that prostate cancer cell lines express both the EPO gene and the biologically active erythropoietin. The coexpression of functional receptor and biologically active ligand in the cells has led us to hypothesize an autocrine/paracrine mechanism, driven by endogenous erythropoietin, which may modulate the growth and progression of prostate cancer. To test our hypothesis, we have knocked down, independently, erythropoietin receptor and erythropoietin on prostate cancer cells by transfection with short hairpin RNAs. Erythropoietin receptor knockdown cells grow significantly more slowly than their erythropoietin receptor-bearing counterparts in monolayer culture, produce fewer, smaller colonies in soft agar, and do not exhibit erythropoietin-induced signaling. Erythropoietin knockdown cells exhibit dramatically slower rates of growth, which could be restored by transfecting the cells with a murine erythropoietin gene. Taken together, our data suggest that the coordinated regulation of a functional erythropoietin/erythropoietin receptor axis in prostate cancer cells may be integral to the growth and progression of prostate cancer.


Subject(s)
Erythropoietin/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Erythropoietin/metabolism , Cell Adhesion/physiology , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Survival/physiology , Down-Regulation , Gene Knockdown Techniques , Humans , Male , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Erythropoietin/deficiency , Receptors, Erythropoietin/genetics , Signal Transduction
8.
FASEB J ; 22(8): 2690-701, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18424770

ABSTRACT

The Wilms' tumor protein Wt1 is required for embryonic development and has been implicated in hematologic disorders. Since Wt1 deficiency may compromise the proliferation and differentiation of erythroid progenitor cells, we analyzed the possible role of the transcriptionally active Wt1 isoform, Wt1(-KTS), in regulating the expression of the erythropoietin receptor (EpoR). Wt1 and EpoR were coexpressed in CD117(+) hematopoietic progenitor cells and in several hematopoietic cell lines. CD117(+) cells of Wt1-deficient murine embryos (Wt1(-/-)) exhibited a significantly lower proliferation response to recombinant erythropoietin than CD117(+) cells of heterozygous (Wt1(+/-)) and wild-type littermates (Wt1(+/+)). EpoR expression was significantly diminished in hematopoietic progenitors (CD117(+)) that lacked Wt1, and the erythroid colony-forming capacity was reduced by more than 50% in fetal liver cells of Wt1-deficient embryonic mice. Wt1(-KTS) significantly increased endogenous EpoR transcripts in transfected cells. The proximal EpoR promoter of human and mouse was stimulated more than 10-fold by Wt1(-KTS) in transiently cotransfeced K562 erythroleukemia cells. A responsible cis-element, which is highly conserved in the EpoR promoter of human and mouse, was identified by mutation analysis, electrophoretic mobility shift assay, and chromatin immunoprecipitation assay. In conclusion, activation of the EpoR gene by Wt1 may represent an important mechanism in normal hematopoiesis.


Subject(s)
Genes, Wilms Tumor , Hematopoietic Stem Cells/metabolism , Receptors, Erythropoietin/genetics , Animals , Base Sequence , Binding Sites/genetics , Cell Line , DNA/genetics , DNA/metabolism , DNA Primers/genetics , Embryonic Stem Cells/metabolism , Erythropoiesis/genetics , Erythropoietin/pharmacology , Genes, Reporter , Hematopoiesis/drug effects , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Heterozygote , Humans , Mice , Mice, Knockout , Mutagenesis, Site-Directed , Promoter Regions, Genetic , Proto-Oncogene Proteins c-kit/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Erythropoietin/deficiency , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transcriptional Activation , Transfection , WT1 Proteins/deficiency , WT1 Proteins/genetics , WT1 Proteins/metabolism
9.
Neonatology ; 93(1): 56-63, 2008.
Article in English | MEDLINE | ID: mdl-17664892

ABSTRACT

BACKGROUND: Erythropoietin (Epo) receptors (EpoR) are present in embryonic and postnatal mammalian bowel, and activation of EpoR signaling with recombinant Epo (rEpo) has trophic effects. Transgenic mice with absent Epo function are embryonic lethal, so it is not known whether Epo function is required for bowel development. OBJECTIVE: To characterize bowel structure in the absence of EpoR signaling. METHODS: Heterozygous EpoR knockout mice were mated. Bowel segments from their embryos were dissected and transplanted beneath the renal capsule of adult wild-type mice and residual embryo tissue was excised for genotyping. Transplants were harvested at 7, 14 or 21 days. The transplanted bowel segments were immunostained to identify proliferation (BrdU+), as well as neuronal (PGP9.5+), endothelial (vWF+), and neuroendocrine (synaptophysin+) cells. Gross and microscopic characteristics of intestinal differentiation were evaluated. RESULTS: 50 transplants were performed: bowel from 49 embryos survived to harvest and 43 showed evidence of bowel development with appropriate small or large intestinal features. No differences in morphology, immunolabeling, or BrdU incorporation were observed between homozygous-null, heterozygote or wild-type bowel. Smooth muscle and mucosal cells were present, along with neuronal, endothelial, and neuroendocrine cells in all genotypes. CONCLUSIONS: Enteric EpoR signaling is not essential for intestinal morphogenesis.


Subject(s)
Intestines/embryology , Intestines/transplantation , Receptors, Erythropoietin/deficiency , Animals , Intestines/anatomy & histology , Mice , Mice, Knockout , Organogenesis/physiology , Receptors, Erythropoietin/genetics , Transplants
10.
Blood ; 110(7): 2408-13, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17620453

ABSTRACT

Recombinant human erythropoietin (rHu-EPO) is used to treat anemia by activating the erythropoietin receptor (EPOR) in erythroid progenitor cells, leading to proliferation and differentiation into mature red blood cells. To allow less frequent dosing, a hyperglycosylated version of EPO has been developed with a longer half-life. In principle, an agonistic antibody targeting EPOR would offer an even longer half-life, support robust monthly dosing, and, unlike EPO products, reduce the risk of pure red cell aplasia. The efficiency of signaling and corresponding potency of previously reported antibody mimics are generally suboptimal compared with EPO and not suitable for clinical use. Here we describe a potent, fully human, agonistic antibody (ABT007) targeting EPOR that supports potent, more sustained, and less pulsatile elevation of hematocrit in a human EPOR-expressing transgenic mouse model compared with standard doses of rHu-EPO while requiring less frequent dosing. Resolution of the crystal structure of the EPOR extracellular domain (ECD) complexed to the ABT007 Fab fragment, determined at 0.32 nm, identifies a binding site that is consistent with a novel mechanism of receptor activation based on a unique antibody-imposed conformational change. These results demonstrate that a symmetric molecule can serve as a potent activator of the EPOR.


Subject(s)
Antibodies/immunology , Erythropoietin/metabolism , Molecular Mimicry , Animals , Binding Sites , Cell Line , Crystallography, X-Ray , Erythropoiesis , Hematocrit , Humans , Mice , Mice, Knockout , Models, Molecular , Protein Structure, Quaternary , Protein Structure, Tertiary , Receptors, Erythropoietin/chemistry , Receptors, Erythropoietin/deficiency , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/metabolism , Structural Homology, Protein
11.
J Biol Chem ; 282(35): 25875-83, 2007 Aug 31.
Article in English | MEDLINE | ID: mdl-17604282

ABSTRACT

Erythropoietin (Epo) and its receptor (EpoR), critical for erythropoiesis, are expressed in the nervous system. Prior to death in utero because of severe anemia EpoR-null mice have fewer neural progenitor cells, and differentiated neurons are markedly sensitive to hypoxia, suggesting that during development Epo stimulates neural cell proliferation and prevents neuron apoptosis by promoting oxygen delivery to brain or by direct interaction with neural cells. Here we present evidence that neural progenitor cells express EpoR at higher levels compared with mature neurons; that Epo stimulates proliferation of embryonic neural progenitor cells; and that endogenous Epo contributes to neural progenitor cell proliferation and maintenance. EpoR-null mice were rescued with selective EpoR expression driven by the endogenous EpoR promoter in hematopoietic tissue but not in brain. Although these mice exhibited normal hematopoiesis and erythrocyte production and survived to adulthood, neural cell proliferation and viability were affected. Embryonic brain exhibited increased neural cell apoptosis, and neural cell proliferation was reduced in the adult hippocampus and subventricular zone. Neural cells from these animals were more sensitive to hypoxia/glutamate neurotoxicity than normal neurons in culture and in vivo. These observations demonstrate that endogenous Epo/EpoR signaling promotes cell survival in embryonic brain and contributes to neural cell proliferation in adult brain in regions associated with neurogenesis. Therefore, Epo exerts extra-hematopoietic function and contributes directly to brain development, maintenance, and repair by promoting cell survival and proliferation independent of insult, injury, or ischemia.


Subject(s)
Apoptosis/physiology , Brain/metabolism , Cell Differentiation/physiology , Cell Proliferation , Neurons/metabolism , Receptors, Erythropoietin/metabolism , Stem Cells/metabolism , Animals , Brain/cytology , Brain/embryology , Cell Hypoxia , Cell Survival/physiology , Erythropoiesis/physiology , Erythropoietin , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/pathology , Mice , Mice, Mutant Strains , Neurons/cytology , Organ Specificity/physiology , Promoter Regions, Genetic/physiology , Receptors, Erythropoietin/deficiency , Signal Transduction/physiology , Stem Cells/cytology
12.
Cardiovasc Res ; 71(3): 466-77, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16781691

ABSTRACT

OBJECTIVE: Recent studies suggested that erythropoietin (Epo) receptors (EpoR) are expressed not only in the hematopoietic lineage cells but also in the heart and that the administration of recombinant human Epo elicits protective effects in myocardial ischemia and reperfusion (I/R). We tested our hypothesis that endogenous Epo signals mediated by EpoR expressed in the non-hematopoietic lineage cells play a protective role against myocardial I/R injury. METHODS: Transgene-rescued EpoR null mutant mice (RES), which express EpoR exclusively in the hematopoietic lineage cells, were subjected to 30 min left coronary artery occlusion followed by reperfusion. RESULTS: Hematocrit, heart rate, blood pressure, heart weight, and echocardiographic parameters were comparable between wild-type mice (WT) and RES under the baseline condition. After 24 h of reperfusion, the infarct size in RES with I/R (RES/MI) was larger than that in WT/MI. Caspase-3 activity and number of TUNEL-positive cardiomyocytes in the ischemic area were increased in RES/MI compared with WT/MI. The extents of p38 and JNK phosphorylations in the ischemic area were significantly increased in WT/MI, but not in RES/MI as compared with corresponding sham-operated mice. Plasma Epo concentration in RES/MI did not differ from that in sham-operated RES, while that in WT/MI was peaked at 24 h post I/R. Additionally, left ventricular (LV) end-diastolic diameter was increased and LV fractional shortening tended to be reduced in the RES/MI compared with WT/MI at 21 days after I/R. CONCLUSIONS: These results suggest that the endogenous Epo-EpoR system in the non-hematopoietic lineage cells plays an important protective role against myocardial I/R injury.


Subject(s)
Erythropoietin/physiology , Myocardial Reperfusion Injury/pathology , Animals , Apoptosis , Blood Pressure , Blotting, Western , Erythropoietin/blood , Hematocrit , Mice , Mice, Transgenic , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/physiopathology , Myocardium/pathology , Myocytes, Cardiac/pathology , Organ Size , Receptors, Erythropoietin/deficiency , Receptors, Erythropoietin/metabolism , Signal Transduction , Ventricular Remodeling
13.
Circulation ; 113(11): 1442-50, 2006 Mar 21.
Article in English | MEDLINE | ID: mdl-16534010

ABSTRACT

BACKGROUND: Recent studies have suggested that endogenous erythropoietin (Epo) plays an important role in the mobilization of bone marrow-derived endothelial progenitor cells (EPCs). However, it remains to be elucidated whether the Epo system exerts protective effects on pulmonary hypertension (PH), a fatal disorder encountered in cardiovascular medicine. METHODS AND RESULTS: A mouse model of hypoxia-induced PH was used for study. We evaluated right ventricular systolic pressure, right ventricular hypertrophy, and pulmonary vascular remodeling in mice lacking the Epo receptor (EpoR) in nonerythroid lineages (EpoR(-/-) rescued mice) after 3 weeks of exposure to hypoxia. Those mice lack EpoR in the cardiovascular system but not in the hematopoietic system. The development of PH and pulmonary vascular remodeling were accelerated in EpoR(-/-) rescued mice compared with wild-type mice. The mobilization of EPCs and their recruitment to the pulmonary endothelium were significantly impaired in EpoR(-/-) rescued mice. By contrast, reconstitution of the bone marrow with wild-type bone marrow cells ameliorated PH in the EpoR(-/-) rescued mice. Hypoxia enhanced the expression of EpoR on pulmonary endothelial cells in wild-type but not EpoR(-/-) rescued mice. Finally, hypoxia activated endothelial nitric oxide synthase in the lungs in wild-type mice but not in EpoR(-/-) rescued mice. CONCLUSIONS: These results indicate that the endogenous Epo/EpoR system plays an important role in the recruitment of EPCs and prevents the development of PH during chronic hypoxia in mice in vivo, suggesting the therapeutic importance of the system for the treatment of PH.


Subject(s)
Endothelium, Vascular/pathology , Erythropoietin/physiology , Hematopoietic Stem Cells/physiology , Hypertension, Pulmonary/prevention & control , Hypoxia/physiopathology , Lung/pathology , Receptors, Erythropoietin/physiology , Animals , Bone Marrow Transplantation , Cell Movement , Cells, Cultured/cytology , Chronic Disease , Endothelial Cells/pathology , Endothelium/pathology , Enzyme Activation , Erythroid Precursor Cells/metabolism , GATA1 Transcription Factor/physiology , Heart Failure/etiology , Hypertension, Pulmonary/etiology , Hypertrophy, Right Ventricular/etiology , Hypertrophy, Right Ventricular/physiopathology , Hypoxia/complications , Lung/blood supply , Male , Mice , Mice, Knockout , Mice, Transgenic , Muscle, Smooth, Vascular/pathology , Nitric Oxide Synthase Type III/metabolism , Organ Specificity , Radiation Chimera , Receptor, TIE-2/genetics , Receptors, Erythropoietin/deficiency , Receptors, Erythropoietin/genetics , Systole , Ventricular Dysfunction, Right/etiology , Ventricular Dysfunction, Right/physiopathology
14.
Blood ; 107(7): 2662-72, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16332976

ABSTRACT

Critical signals for erythroblast formation are transduced by activated, tyrosine-phosphorylated erythropoietin receptor (EpoR) complexes. Nonetheless, steady-state erythropoiesis is supported effectively by EpoR alleles that are deficient in cytoplasmic phosphotyrosine sites. To better define core EpoR action mechanisms, signaling capacities of minimal PY-null (EpoR-HM) and PY343-retaining (EpoR-H) alleles were analyzed for the first time in bone marrow-derived erythroblasts. Jak2 activation via each allele was comparable. Stat5 (and several Stat5-response genes) were induced via EpoR-H but not via EpoR-HM. Stat1 and Stat3 activation was nominal for all EpoR forms. For both EpoR-HM and EpoR-H, Akt and p70S6-kinase activation was decreased multifold, and JNK activation was minimal. ERKs, however, were hyperactivated uniquely via EpoR-HM. In vivo, Epo expression in EpoR-HM mice was elevated, while Epo-induced reticulocyte production was diminished. In vitro, EpoR-HM erythroblast maturation also was attenuated (based on DNA content, forward-angle light scatter, and hemoglobinization). These EpoR-HM-specific defects were corrected not only upon PY343 site restoration in EpoR-H, but also upon MEK1,2 inhibition. Core EpoR PY site-independent signals for erythroblast formation therefore appear to be Stat5, Stat1, Stat3, p70S6-kinase, and JNK independent, but ERK dependent. Wild-type signaling capacities, however, depend further upon signals provided via an EpoR/PY343/Stat5 axis.


Subject(s)
Erythroblasts/physiology , Receptors, Erythropoietin/physiology , Reticulocytes/physiology , Stem Cells/cytology , Animals , Bone Marrow Cells/cytology , Cells, Cultured , Erythroblasts/cytology , Flow Cytometry , Gene Expression Regulation , Mice , Mice, Knockout , Receptors, Erythropoietin/deficiency , Receptors, Erythropoietin/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
15.
Proc Natl Acad Sci U S A ; 101(41): 14907-12, 2004 Oct 12.
Article in English | MEDLINE | ID: mdl-15456912

ABSTRACT

The cytokine erythropoietin (Epo) is tissue-protective in preclinical models of ischemic, traumatic, toxic, and inflammatory injuries. We have recently characterized Epo derivatives that do not bind to the Epo receptor (EpoR) yet are tissue-protective. For example, carbamylated Epo (CEpo) does not stimulate erythropoiesis, yet it prevents tissue injury in a wide variety of in vivo and in vitro models. These observations suggest that another receptor is responsible for the tissue-protective actions of Epo. Notably, prior investigation suggests that EpoR physically interacts with the common beta receptor (betacR), the signal-transducing subunit shared by the granulocyte-macrophage colony stimulating factor, and the IL-3 and IL-5 receptors. However, because betacR knockout mice exhibit normal erythrocyte maturation, betacR is not required for erythropoiesis. We hypothesized that betacR in combination with the EpoR expressed by nonhematopoietic cells constitutes a tissue-protective receptor. In support of this hypothesis, membrane proteins prepared from rat brain, heart, liver, or kidney were greatly enriched in EpoR after passage over either Epo or CEpo columns but covalently bound in a complex with betacR. Further, antibodies against EpoR coimmunoprecipitated betacR from membranes prepared from neuronal-like P-19 cells that respond to Epo-induced tissue protection. Immunocytochemical studies of spinal cord neurons and cardiomyocytes protected by Epo demonstrated cellular colocalization of Epo betacR and EpoR. Finally, as predicted by the hypothesis, neither Epo nor CEpo was active in cardiomyocyte or spinal cord injury models performed in the betacR knockout mouse. These data support the concept that EpoR and betacR comprise a tissue-protective heteroreceptor.


Subject(s)
Erythropoietin/therapeutic use , Receptors, Erythropoietin/physiology , Spinal Cord Injuries/physiopathology , Animals , Aorta , Cell Line , Cell Membrane/ultrastructure , Cells, Cultured , Erythropoietin/deficiency , Erythropoietin/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Protein Subunits , Receptors, Erythropoietin/deficiency , Receptors, Erythropoietin/genetics , Spinal Cord Injuries/drug therapy , Time Factors , Ventricular Function/physiology
16.
Blood ; 100(7): 2279-88, 2002 Oct 01.
Article in English | MEDLINE | ID: mdl-12239135

ABSTRACT

Erythropoietin (Epo) and its receptor (EpoR) are indispensable to erythropoiesis. Although roles besides angiogenesis, such as neuroprotection and heart development, have been reported for the Epo-EpoR system, the precise contribution of Epo-EpoR to these nonhematopoietic tissues requires clarification. Exploiting a GATA-1 minigene cassette with hematopoietic regulatory domains, we established 2 lines of transgene-rescued EpoR-null mutant mice expressing EpoR exclusively in the hematopoietic lineage. Surprisingly, despite the lack of EpoR expression in nonhematopoietic tissues, these mice develop normally and are fertile. As such, we could exploit them for analyzing the roles of the Epo-EpoR system in adult hematopoiesis and in nonhematopoietic tissues. These rescued lines showed a differential level of EpoR expression in erythroid cells; one expressed approximately 40%, and the other expressed 120% of the wild-type EpoR level. A colony formation assay showed that erythroid progenitors in the 2 mutant lines exhibit distinct sensitivity to Epo. The circulating Epo level was much higher in the transgenic line with a lower EpoR expression. In response to induced anemia, the plasma Epo concentrations increased in both lines. Notably, the timing of the peak of plasma Epo concentration was delayed in both lines of rescued mice compared with wild type, suggesting that, in wild-type mice, nonhematopoietic EpoR contributes to the regulation of plasma Epo concentration. We thus conclude that nonhematopoietic expression of EpoR is dispensable to normal mouse development and that the expression level of EpoR regulates erythropoiesis by controlling the sensitivity of erythroid progenitors to Epo.


Subject(s)
Hematopoietic Stem Cells/physiology , Receptors, Erythropoietin/genetics , Anemia/genetics , Anemia/pathology , Anemia/therapy , Animals , Apoptosis , DNA Primers , Embryonic and Fetal Development , Female , Fetal Death , Genes, Lethal , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/pathology , In Situ Nick-End Labeling , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Polymerase Chain Reaction , Pregnancy , Receptors, Erythropoietin/deficiency , Reverse Transcriptase Polymerase Chain Reaction
17.
Blood ; 99(7): 2603-5, 2002 Apr 01.
Article in English | MEDLINE | ID: mdl-11895800

ABSTRACT

Erythropoietin (EPO) and its receptor (EPOR) are critical for definitive erythropoiesis, as mice lacking either gene product die during embryogenesis with severe anemia. Here we demonstrate that mice expressing just one functional allele of the EpoR have lower hematocrits and die more frequently than do wild-type littermates on anemia induction. Furthermore, EpoR(+/-) erythroid colony-forming unit (CFU-E) progenitors are reduced both in frequency and in responsiveness to EPO stimulation. To evaluate the interaction between EPO and granulocyte-macrophage colony-stimulating factor (GM-CSF) or interleukin 3 (IL-3), GM-CSF(-/-) or IL-3(-/-) mice were interbred with EpoR(+/)(-) mice. Deletion of either GM-CSF or IL-3 also leads to reduction in CFU-E numbers and hematocrits but does not significantly alter steady-state erythroid burst-forming unit numbers. These results suggest EpoR haploinsufficiency and promotion of in vivo erythropoiesis by GM-CSF and IL-3.


Subject(s)
Anemia/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Interleukin-3/physiology , Receptors, Erythropoietin/physiology , Alleles , Animals , Crosses, Genetic , Erythropoiesis , Granulocyte-Macrophage Colony-Stimulating Factor/deficiency , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Haplotypes , Hematocrit , Hematopoietic Stem Cells/cytology , Interleukin-3/deficiency , Interleukin-3/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptors, Erythropoietin/deficiency , Receptors, Erythropoietin/genetics
18.
Blood ; 98(5): 1408-15, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11520789

ABSTRACT

Erythropoiesis occurs in 2 distinct waves during embryogenesis: the primitive wave in the extra-embryonic yolk sac (YS) followed by the definitive wave in the fetal liver and spleen. Even though progenitors for both cell types are present in the YS blood islands, only primitive cells are formed in the YS during early embryogenesis. In this study, it is proposed that erythropoietin (Epo) expression and the resultant EpoR activation regulate the timing of the definitive wave. First, it was demonstrated that Epo and EpoR gene expressions are temporally and spatially segregated: though EpoR is expressed early (embryonic days 8.0-9.5) in the yolk sac blood islands, no Epo expression can be detected in this extra-embryonic tissue. Only at a later stage can Epo expression be detected intra-embryonically, and the onset of Epo expression correlates with the initiation of definitive erythropoiesis. It was further demonstrated that the activation of the EpoR signaling pathway by knocking-in a constitutively active form of EpoR (R129C EpoR) can lead to earlier onset of definitive erythropoiesis in the YS. Thus, these results provide the first in vivo mechanism as to how 2 erythroid progenitor populations can coexist concurrently in the YS yet always differentiate successively during embryogenesis.


Subject(s)
Erythropoiesis/physiology , Erythropoietin/physiology , Gene Expression Regulation, Developmental , Receptors, Erythropoietin/physiology , Animals , Chimera , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/drug effects , Erythroid Precursor Cells/metabolism , Erythropoiesis/genetics , Erythropoietin/biosynthesis , Erythropoietin/deficiency , Erythropoietin/genetics , Globins/biosynthesis , Globins/genetics , In Situ Hybridization , Mice , Mice, Knockout , Organ Culture Techniques , Receptors, Erythropoietin/biosynthesis , Receptors, Erythropoietin/deficiency , Receptors, Erythropoietin/genetics , Signal Transduction , Yolk Sac/metabolism
19.
Blood ; 98(2): 475-7, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11435319

ABSTRACT

Erythropoietin and its receptor are required for definitive erythropoiesis and maturation of erythroid progenitor cells. Mice lacking the erythropoietin receptor exhibit severe anemia and die at about embryonic day 13.5. This phenotype can be rescued by the human erythropoietin receptor transgene. Animals expressing only the human erythropoietin receptor survived through adulthood with normal hematologic parameters and appeared to respond appropriately to induced anemic stress. In addition to restoration of erythropoiesis during development, the cardiac defect associated with embryos lacking the erythropoietin receptor was corrected and the increased apoptosis in fetal liver, heart, and brain in the erythropoietin receptor null phenotype was markedly reduced. These studies indicate that no species barrier exists between mouse and human erythropoietin receptor and that the human erythropoietin receptor transgene is able to provide specific expression in hematopoietic and other selected tissues to rescue erythropoiesis and other organ defects observed in the erythropoietin receptor null mouse.


Subject(s)
Erythropoiesis/genetics , Receptors, Erythropoietin/deficiency , Receptors, Erythropoietin/genetics , Anemia/chemically induced , Anemia/genetics , Anemia/therapy , Animals , Apoptosis , Bone Marrow/metabolism , Bone Marrow Cells/cytology , Colony-Forming Units Assay , Crosses, Genetic , Erythropoietin/physiology , Female , Gene Expression , Hematopoietic Stem Cells/chemistry , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , In Situ Nick-End Labeling , Male , Mice , Mice, Knockout , Mice, Transgenic , RNA, Messenger/analysis , Spleen/metabolism
20.
Pathol Biol (Paris) ; 49(2): 170-7, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11317965

ABSTRACT

By definition, idiopathic erythrocytosis (IE) applies to a group of patients characterised by having a measured RCM above their predicted normal range (an absolute erythrocytosis) and following investigation do not have a form of primary or secondary erythrocytosis. Patients with IE are heterogenous. The possibilities include physiological variation, 'early' polycythaemia vera (10-15% develop clear features of PV over a few years), unrecognized congenital erythrocytosis, unrecognized or unrecognizable secondary acquired erythrocytosis or a currently undescribed form of primary or secondary erythrocytosis. Patients are more commonly male with a median age at presentation of 55-60 years. Approximately half of the patients present with vascular occlusive complications. Retrospective evidence indicates that vascular occlusion occurs less frequently when the PCV is controlled at normal levels. Venesection is the treatment of choice to lower the PCV. As a general approach to management, all patients with a PCV above 0.54 should be venesected to a PCV less than 0.45. This target PCV should also apply to patients with lesser degrees of raised PCV who have additional other risk factors for vascular occlusion.


Subject(s)
Polycythemia , Aged , Arterial Occlusive Diseases/etiology , Bone Marrow/pathology , Chlorambucil/adverse effects , Chlorambucil/therapeutic use , Diagnosis, Differential , Endocrine System Diseases/complications , Erythrocyte Volume , Erythroid Precursor Cells/pathology , Erythropoietin/blood , Genetic Predisposition to Disease , Humans , Hypoxia/complications , Kidney Diseases/complications , Leukemia/chemically induced , Leukemia, Radiation-Induced/etiology , Middle Aged , Phosphorus Radioisotopes/adverse effects , Phosphorus Radioisotopes/therapeutic use , Polycythemia/classification , Polycythemia/congenital , Polycythemia/diagnosis , Polycythemia/etiology , Polycythemia/therapy , Polycythemia Vera/diagnosis , Receptors, Erythropoietin/deficiency , Receptors, Erythropoietin/genetics , Sequence Deletion , Smoking/blood , Stroke/etiology
SELECTION OF CITATIONS
SEARCH DETAIL
...