Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 332
Filter
1.
Front Immunol ; 12: 768435, 2021.
Article in English | MEDLINE | ID: mdl-34925338

ABSTRACT

Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is a condition with an imbalanced inflammatory response and delayed resolution of inflammation. Macrophage polarization plays an important role in inflammation and resolution. However, the mechanism of macrophage polarization in ALI/ARDS is not fully understood. We found that mice with lipopolysaccharide administration developed lung injury with the accumulation of extracellular cold-inducible RNA-binding protein (eCIRP) in the lungs. eCIRP, as a damage-associated molecular pattern (DAMP), inhibited M2 macrophage polarization, thereby tipping the balance toward inflammation rather than resolution. Anti-CIRP antibodies reversed such phenotypes. The levels of macrophage erythropoietin (EPO) receptor (EPOR) were reduced after eCIRP treatment. Myeloid-specific EPOR-deficient mice displayed restrained M2 macrophage polarization and impaired inflammation resolution. Mechanistically, eCIRP impaired Rab26, a member of Ras superfamilies of small G proteins, and reduced the transportation of surface EPOR, which resulted in macrophage polarization toward the M1 phenotype. Moreover, EPO treatment hardly promotes M2 polarization in Rab26 knockout (KO) macrophages through EPOR. Collectively, macrophage EPOR signaling is impaired by eCIRP through Rab26 during ALI/ARDS, leading to the restrained M2 macrophage polarization and delayed inflammation resolution. These findings identify a mechanism of persistent inflammation and a potential therapy during ALI/ARDS.


Subject(s)
Acute Lung Injury/immunology , Macrophages/physiology , RNA-Binding Proteins/physiology , Receptors, Erythropoietin/physiology , rab GTP-Binding Proteins/physiology , Animals , Cell Polarity , Cells, Cultured , Inflammation/etiology , Mice , Mice, Inbred C57BL , PPAR gamma/physiology
2.
Int J Mol Sci ; 22(13)2021 Jul 01.
Article in English | MEDLINE | ID: mdl-34281163

ABSTRACT

Erythropoietin (EPO) acts on multiple tissues through its receptor EPOR, a member of a cytokine class I receptor superfamily with pleiotropic effects. The interaction of EPO and EPOR triggers the activation of several signaling pathways that induce erythropoiesis, including JAK2/STAT5, PI3K/AKT, and MAPK. The canonical EPOR/JAK2/STAT5 pathway is a known regulator of differentiation, proliferation, and cell survival of erythroid progenitors. In addition, its role in the protection of other cells, including cancer cells, is under intense investigation. The involvement of EPOR/JAK2/STAT5 in other processes such as mRNA splicing, cytoskeleton reorganization, and cell metabolism has been recently described. The transcriptomics, proteomics, and epigenetic studies reviewed in this article provide a detailed understanding of EPO signalization. Advances in this area of research may be useful for improving the efficacy of EPO therapy in hematologic disorders, as well as in cancer treatment.


Subject(s)
Erythropoietin/metabolism , STAT5 Transcription Factor/metabolism , STAT5 Transcription Factor/physiology , Animals , Cell Differentiation/drug effects , Epigenomics/methods , Erythropoiesis/drug effects , Erythropoietin/physiology , Humans , Janus Kinase 2/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proteomics/methods , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Erythropoietin/metabolism , Receptors, Erythropoietin/physiology , STAT5 Transcription Factor/genetics , Signal Transduction/drug effects , Trans-Activators/metabolism , Transcriptome/genetics
3.
Int J Mol Sci ; 22(14)2021 Jul 19.
Article in English | MEDLINE | ID: mdl-34299300

ABSTRACT

Erythropoietin (EPO) is a glycoprotein cytokine known for its pleiotropic effects on various types of cells and tissues. EPO and its receptor EPOR trigger signaling cascades JAK2/STAT5, MAPK, and PI3K/AKT that are interconnected and irreplaceable for cell survival. In this article, we describe the role of the MAPK and PI3K/AKT signaling pathways during red blood cell formation as well as in non-hematopoietic tissues and tumor cells. Although the central framework of these pathways is similar for most of cell types, there are some stage-specific, tissue, and cell-lineage differences. We summarize the current state of research in this field, highlight the novel members of EPO-induced PI3K and MAPK signaling, and in this respect also the differences between erythroid and non-erythroid cells.


Subject(s)
Erythropoiesis/physiology , Erythropoietin/physiology , Animals , Cell Differentiation/physiology , Cell Proliferation/physiology , Cell Survival/physiology , Humans , MAP Kinase Signaling System , Models, Biological , Neoplasms/physiopathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Erythropoietin/physiology , Signal Transduction
4.
F1000Res ; 92020.
Article in English | MEDLINE | ID: mdl-32983414

ABSTRACT

More than 50 years of efforts to identify the major cytokine responsible for red blood cell (RBC) production (erythropoiesis) led to the identification of erythropoietin (EPO) in 1977 and its receptor (EPOR) in 1989, followed by three decades of rich scientific discovery. We now know that an elaborate oxygen-sensing mechanism regulates the production of EPO, which in turn promotes the maturation and survival of erythroid progenitors. Engagement of the EPOR by EPO activates three interconnected signaling pathways that drive RBC production via diverse downstream effectors and simultaneously trigger negative feedback loops to suppress signaling activity. Together, the finely tuned mechanisms that drive endogenous EPO production and facilitate its downstream activities have evolved to maintain RBC levels in a narrow physiological range and to respond rapidly to erythropoietic stresses such as hypoxia or blood loss. Examination of these pathways has elucidated the genetics of numerous inherited and acquired disorders associated with deficient or excessive RBC production and generated valuable drugs to treat anemia, including recombinant human EPO and more recently the prolyl hydroxylase inhibitors, which act partly by stimulating endogenous EPO synthesis. Ongoing structure-function studies of the EPOR and its essential partner, tyrosine kinase JAK2, suggest that it may be possible to generate new "designer" drugs that control selected subsets of cytokine receptor activities for therapeutic manipulation of hematopoiesis and treatment of blood cancers.


Subject(s)
Erythrocytes/cytology , Erythropoiesis , Erythropoietin/physiology , Receptors, Erythropoietin/physiology , Humans , Janus Kinase 2/physiology , Signal Transduction
5.
G Ital Nefrol ; 36(5)2019 Sep 24.
Article in Italian | MEDLINE | ID: mdl-31580542

ABSTRACT

Over the past two decades it has emerged that, in addition to erythropoietic activity, erythropoietin (EPO) has numerous other functions, including neuro-protective, anti-apoptotic, antioxidant, angiogenetic and immunomodulatory ones. EPO interacts with two different forms of its receptor (EPOR): a homodimer receptor, responsible for the erythropoietic effects, and a heterodimer receptor, responsible for the non-erythropoietic effects. The effects on the heterodimer receptor are responsible for EPO-induced prolongation of organ transplant survival in mice and humans. The development of new molecules that selectively target the heterodimer EPOR is allowing to test the effect of long-term treatments, without the possible complications related to the increased hematocrit.


Subject(s)
Erythropoiesis/physiology , Erythropoietin/physiology , Graft Survival/physiology , Receptors, Erythropoietin/physiology , Adaptive Immunity , Anemia/drug therapy , Anemia/etiology , Animals , Cell Hypoxia/physiology , Erythropoietin/genetics , Erythropoietin/pharmacology , Graft Survival/drug effects , Heart/drug effects , Humans , Immunity, Cellular , Immunity, Innate , Kidney/drug effects , Kidney/metabolism , Mice , Nervous System/metabolism , Organ Transplantation , Rats , Receptors, Colony-Stimulating Factor/physiology , Recombinant Proteins/therapeutic use , Renal Insufficiency, Chronic/complications , Retina/metabolism
6.
World Neurosurg ; 131: 346-355, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31658577

ABSTRACT

BACKGROUND: Erythropoietin (EPO) is a cytokine primarily involved in the regulation of erythropoiesis. In response to hypoxia-ischemia, hypoxia-inducible factor 1 induces EPO production, which, in turn, inhibits apoptosis of erythroid progenitor cells. By the same mechanism and acting through other signaling pathways, EPO exerts neuroprotective effects. Increased resistance to hypoxia and decreased apoptosis are thought to be important mechanisms for tumor progression, including malignant glioma. Because recent studies have demonstrated that EPO and its receptor (EPOR) are expressed in several tumors and can promote tumor growth, in the present study, we investigated EPO and EPOR expression in human glioma and the effect of EPO administration in a rat model of glioma implantation. METHODS: Using Western blotting and immunohistochemical analysis, we examined the expression of EPO, EPOR, platelet endothelial cell adhesion molecule, and Ki-67 in human glioma specimens and experimentally induced glioma in rats. In the experimental setting, a daily dose of recombinant human EPO (rHuEPO) or saline solution were administered for 21 days in Fischer rats subjected to 9L cell line implantation. RESULTS: In both human and animal specimens, we found an increase in EPOR expression as long as the lesion presented with an increasing malignant pattern. A significant direct correlation was found between the expression of EPOR and Ki-67 and EPOR and platelet endothelial cell adhesion molecule in low- and high-grade gliomas. The rats treated with rHuEPO presented with significantly larger tumor spread compared with the saline-treated rats. CONCLUSIONS: The results of our study have shown that the EPO/EPOR complex might play a significant role in the aggressive behavior of high-grade gliomas. The larger tumor spread in rHuEPO-treated rats suggests a feasible role for EPO in the aggressiveness and progression of malignant glioma.


Subject(s)
Brain Neoplasms/metabolism , Erythropoietin/metabolism , Glioma/metabolism , Receptors, Erythropoietin/metabolism , Adult , Aged , Animals , Blotting, Western , Brain Neoplasms/etiology , Brain Neoplasms/pathology , Cell Line, Tumor , Disease Models, Animal , Erythropoietin/pharmacology , Erythropoietin/physiology , Female , Glioma/etiology , Glioma/pathology , Humans , Immunohistochemistry , Ki-67 Antigen/metabolism , Male , Middle Aged , Neoplasm Transplantation , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Rats, Inbred F344 , Receptors, Erythropoietin/physiology , Recombinant Proteins/pharmacology , Tumor Burden/drug effects
7.
Am J Transplant ; 19(9): 2407-2414, 2019 09.
Article in English | MEDLINE | ID: mdl-30903735

ABSTRACT

Erythropoietin (EPO) is a glycoprotein produced mainly by the adult kidney in response to hypoxia and is the crucial regulator of red blood cell production. EPO receptors (EPORs), however, are not confined to erythroid cells, but are expressed by many organs including the heart, brain, retina, pancreas, and kidney, where they mediate EPO-induced, erythropoiesis-independent, tissue-protective effects. Some of these tissues also produce and locally release small amounts of EPO in response to organ injury as a mechanism of self-repair. Growing evidence shows that EPO possesses also important immune-modulating effects. Monocytes can produce EPO, and autocrine EPO/EPOR signaling in these cells is crucial in maintaining immunologic self-tolerance. New data in mice and humans also indicate that EPO has a direct inhibitory effect on effector/memory T cells, while it promotes formation of regulatory T cells. This review examines the nonerythropoietic effects of EPO, with a special emphasis on its modulating activity on innate immune cells and T cells and on how it affects transplant outcomes.


Subject(s)
Erythropoietin/physiology , Receptors, Erythropoietin/physiology , Animals , Apoptosis , Erythrocytes/metabolism , Erythropoietin/chemistry , Humans , Hypoxia , Immunity, Innate , Immunologic Memory , Mice , Monocytes/metabolism , Protein Binding , Receptors, Erythropoietin/chemistry , Regeneration , Signal Transduction , T-Lymphocytes , T-Lymphocytes, Regulatory/cytology , Transplants/immunology
8.
Cell Calcium ; 74: 123-130, 2018 09.
Article in English | MEDLINE | ID: mdl-30048878

ABSTRACT

Tissue hypoxia is a condition that induces calcium influx into living cells. Calcium is a major player in maintaining cell signaling and homeostasis, and mediates the regulation of gene transcription and cell proliferation; however, acute and aggressive calcium influx induced by hypoxia eventually leads to programmed cell death. The blind mole rat, Spalax, is a wild-spread burrowing mammal adapted to hypoxic environments. A tyrosine -to- phenylalanine (F481 in Spalax corresponding to Y485 in human full-length receptor; Y460 in human mature form) substitution is found in the erythropoietin receptor of Spalax and other species, which was previously shown to be strongly involved in the calcium channels activation and subsequent calcium influx. The current work aimed to explore the dynamics of calcium transport across Spalax nonhematopoietic cells' membrane compared to above ground rat and mouse, and the role of the erythropoietin receptor of Spalax in the regulation of calcium influx under hypoxia. We show here that Epo-induced calcium influx in HEK293 cells transfected with Spalax EpoR is significantly lower than that of mouse; in hypoxia this difference was even more pronounced. Western blots confirmed a significant increase of Erk phosphorylation after stimulation with erythropoietin under hypoxia in cells transfected with mouse full length erythropoietin receptor compared to Spalax. Native primary fibroblasts showed lower cytosolic calcium concentrations in Spalax cells when compared to those of rats under normoxic and hypoxic conditions. Spalax EpoR appears to play an important role in preventing deleterious consequences of hypoxia and maintaining cellular homeostasis under stress.


Subject(s)
Calcium/metabolism , Fibroblasts/metabolism , Receptors, Erythropoietin/physiology , Spalax/metabolism , Animals , Cell Hypoxia/physiology , Cells, Cultured , HEK293 Cells , Humans , Mice , Mole Rats , Rats
9.
Physiol Rep ; 6(12): e13751, 2018 06.
Article in English | MEDLINE | ID: mdl-29939494

ABSTRACT

Clinically, erythropoietin (EPO) is known to increase systemic vascular resistance and arterial blood pressure. However, EPO stimulates the production of the potent vasodilator, nitric oxide (NO), in culture endothelial cells. The mechanism by which EPO causes vasoconstriction despite stimulating NO production may be dependent on its ability to activate two receptor complexes, the homodimeric EPO (EPOR2 ) and the heterodimeric EPOR/ß-common receptor (ßCR). The purpose of this study was to investigate the contribution of each receptor to the vasoactive properties of EPO. First-order, mesenteric arteries were isolated from 16-week-old male C57BL/6 mice, and arterial function was studied in pressure arteriographs. To determine the contribution of each receptor complex, EPO-stimulating peptide (ESP), which binds and activates the heterodimeric EPOR/ßCR complex, and EPO, which activates both receptors, were added to the arteriograph chamber 20 min prior to evaluation of endothelium-dependent (acetylcholine, bradykinin, A23187) and endothelium-independent (sodium nitroprusside) vasodilator responses. Only ACh-induced vasodilation was impaired in arteries pretreated with EPO or ESP. EPO and ESP pretreatment abolished ACh-induced vasodilation by 100% and 60%, respectively. EPO and ESP did not affect endothelium-independent vasodilation by SNP. Additionally, a novel ßCR inhibitory peptide (ßIP), which was computationally developed, prevented the impairment of acetylcholine-induced vasodilation by EPO and ESP, further implicating the EPOR/ßCR complex. Last, pretreatment with either EPO or ESP did not affect vasoconstriction by phenylephrine and KCl. Taken together, these findings suggest that acute activation of the heterodimeric EPOR/ßCR in endothelial cells leads to a selective impairment of ACh-mediated vasodilator response in mouse mesenteric resistance arteries.


Subject(s)
Acetylcholine/antagonists & inhibitors , Cytokine Receptor Common beta Subunit/drug effects , Erythropoietin/pharmacology , Mesenteric Arteries/physiology , Receptors, Erythropoietin/drug effects , Vasodilation/physiology , Acetylcholine/pharmacology , Animals , Arterioles/physiology , Cytokine Receptor Common beta Subunit/physiology , Endothelium, Vascular/physiology , Male , Mesenteric Arteries/drug effects , Mice, Inbred C57BL , Nitroprusside/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Receptors, Erythropoietin/physiology , Recombinant Proteins/pharmacology , Vasodilation/drug effects , Vasodilator Agents/antagonists & inhibitors , Vasodilator Agents/pharmacology
10.
PLoS One ; 13(5): e0196684, 2018.
Article in English | MEDLINE | ID: mdl-29738538

ABSTRACT

OBJECTIVES: Anemia is a known driver for hypoxia inducible factor (HIF) which leads to increased renal erythropoietin (EPO) synthesis. Bone marrow (BM) EPO receptor (EPOR) signals are transduced through a JAK2-STAT5 pathway. The origins of anemia of chronic kidney disease (CKD) are multifactorial, including impairment of both renal EPO synthesis as well as intestinal iron absorption. We investigated the HIF- EPO- EPOR axis in kidney, BM and proximal tibia in anemic juvenile CKD rats. METHODS: CKD was induced by 5/6 nephrectomy in young (20 days old) male Sprague-Dawley rats while C group was sham operated. Rats were sacrificed 4 weeks after CKD induction and 5 minutes after a single bolus of IV recombinant human EPO. An additional control anemic (C-A) group was daily bled for 7 days. RESULTS: Hemoglobin levels were similarly reduced in CKD and C-A (11.4 ± 0.3 and 10.8±0.2 Vs 13.5±0.3 g/dL in C, p<0.0001). Liver hepcidin mRNA was decreased in CA but increased in CKD. Serum iron was unchanged while transferrin levels were mildly decreased in CKD. Kidney HIF2α protein was elevated in C-A but unchanged in CKD. Kidney EPO protein and mRNA levels were unchanged between groups. However, BM EPO protein (which reflects circulating EPO) was increased in C-A but remained unchanged in CKD. BM and proximal tibia EPOR were unchanged in C-A but decreased in CKD. Proximal tibial phospho-STAT5 increased after the EPO bolus in C but not in CKD. CONCLUSIONS: Compared to blood loss, anemia in young CKD rats is associated with inappropriate responses in the HIF-EPO-EPO-R axis: kidney HIF2α and renal EPO are not increased, BM and bone EPOR levels, as well as bone pSTAT5 response to EPO are reduced. Thus, anemia of CKD may be treated with additional therapeutic avenues beyond iron and EPO supplementation.


Subject(s)
Anemia/etiology , Basic Helix-Loop-Helix Transcription Factors/physiology , Erythropoietin/physiology , Receptors, Erythropoietin/physiology , Renal Insufficiency, Chronic/complications , Signal Transduction/physiology , Anemia/physiopathology , Animals , Bone Marrow/metabolism , Disease Models, Animal , Erythropoietin/biosynthesis , Erythropoietin/genetics , Erythropoietin/pharmacology , Hepcidins/physiology , Kidney/metabolism , Liver/metabolism , Male , Nephrectomy/adverse effects , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/physiopathology , STAT5 Transcription Factor/physiology
11.
Sci Rep ; 8(1): 2161, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29391474

ABSTRACT

Erythropoietin (EPO) is recognized for neuroprotective and angiogenic effects and has been associated with aging and neovascular age-related macular degeneration (AMD). We hypothesized that systemic EPO facilitates the development of choroidal neovascularization (CNV). Wild type mice expressed murine EPOR (mWtEPOR) in RPE/choroids at baseline and had significantly increased serum EPO after laser treatment. To test the role of EPO signaling, we used human EPOR knock-in mice with the mWtEPOR gene replaced by either the human EPOR gene (hWtEPOR) or a mutated human EPOR gene (hMtEPOR) in a laser-induced choroidal neovascularization (LCNV) model. Loss-of-function hWtEPOR mice have reduced downstream activation, whereas gain-of-function hMtEPOR mice have increased EPOR signaling. Compared to littermate controls (mWtEPOR), hMtEPOR with increased EPOR signaling developed larger CNV lesions. At baseline, hMtEPOR mice had increased numbers of macrophages, greater expression of macrophage markers F4/80 and CD206, and following laser injury, had greater expression of cytokines CCL2, CXCL10, CCL22, IL-6, and IL-10 than mWtEPOR controls. These data support a hypothesis that injury from age- and AMD-related changes in the RPE/choroid leads to choroidal neovascularization through EPOR-mediated cytokine production.


Subject(s)
Choroid/blood supply , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Cytokines/metabolism , Erythropoietin/metabolism , Macrophages/physiology , Receptors, Erythropoietin/physiology , Animals , Cells, Cultured , Choroid/metabolism , Disease Models, Animal , Female , Macrophages/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction
12.
Biosci Rep ; 37(6)2017 Dec 22.
Article in English | MEDLINE | ID: mdl-29127105

ABSTRACT

Ischemia-reperfusion injury (IRI) remains a key component of graft damage during transplantation. Erythropoietin (EPO) induces anti-inflammatory and anti-apoptotic effects via the EPOR2/ßcR2 complex, with a potential risk of thrombosis. Previous work indicates that EPO has EPOR2/ßcR2-independent protective effects via direct effects on the endothelium. As the EPOR2/ßcR2 receptor has a very low affinity for EPO, we aimed to test the hypothesis that EPO doses below the level that stimulate this receptor elicit cytoprotective effects via endothelial stimulation in a porcine liver transplantation model. Landrace pigs underwent allogenic liver transplantation (follow-up: 6 h) with a portojugular shunt. Animals were divided into two groups: donor and recipient treatment with low-dose EPO (65 IU/kg) or vehicle, administered 6 h before cold perfusion and 30 min after warm reperfusion. Fourteen of 17 animals (82.4%) fulfilled the inclusion criteria. No differences were noted in operative values between the groups including hemoglobin, cold or warm ischemic time. EPO-treated animals showed a significantly lower histopathology score, reduced apoptosis, oxidative stress, and most important a significant up-regulation of endothelial nitric oxide (NO) synthase (eNOS). Donor and recipient treatment with low-dose EPO reduces the hepatic IRI via EPOR2/ßcR2-independent cytoprotective mechanisms and represents a clinically applicable way to reduce IRI.


Subject(s)
Epoetin Alfa/pharmacology , Receptors, Erythropoietin/physiology , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Drug Evaluation, Preclinical , Epoetin Alfa/physiology , Female , Humans , Liver/enzymology , Liver/pathology , Liver/surgery , Liver Transplantation , Protective Agents/pharmacology , Reperfusion Injury/prevention & control , Sus scrofa
13.
Vitam Horm ; 105: 233-248, 2017.
Article in English | MEDLINE | ID: mdl-28629520

ABSTRACT

Erythropoietin (EPO) is a growth hormone, widely known for its role in erythropoiesis. The broad expression of erythropoietin receptor (EPOR) in adult organs suggested that EPO may also affect other cells besides late erythroid progenitors. In the embryonic heart, EPOR is expressed in all cells including the immature proliferating cardiomyocytes. In contrast to the embryonic heart in adulthood, EPOR expression is decreased and mainly detected in immature proliferating cells (i.e., resident cardiac progenitor cells) rather than in terminally differentiated cells (i.e., cardiomyocytes). Since cardiac progenitor cells are considered a regenerative cell source upon cardiac injury, the protective action of the EPO system was tested by creating an erythroid-rescued EPOR knockout mouse model. Although these mice appear to have less immature proliferating myocytes during embryogenesis, they reach adulthood without apparent morphological defects. However, upon ischemia reperfusion, these animals show a greater infarct size, suggesting that the EPO/EPOR protects the heart upon injury. Indeed preclinical studies showed that EPO administration postinfarction improves cardiac function via neoangiogenesis, antiapoptotic mechanisms, and/or CPC activation. Despite the promising preclinical data, large cohort clinical studies in humans failed to show a significant amelioration in cardiac function upon systemic injection of EPO in patients with myocardial infarctions. The discrepancy between preclinical and clinical trials may be due to differences between the doses, the way of delivery, the homogeneity of the cohorts, and last but not least the species differences. These data pinpoint the importance of carrying out preclinical studies in human models of disease as engineered human cardiac tissue that will provide a better understanding of the expression pattern of EPOR and the role of its ligand in human cardiac cells. Such studies may be able to bridge the gap between preclinical rodent data and human clinical trials and thus lead to the design of more successful clinical studies.


Subject(s)
Erythropoietin/physiology , Heart Injuries/metabolism , Receptors, Erythropoietin/physiology , Stem Cells/physiology , Animals , Humans , Myocardium/cytology , Myocardium/metabolism
14.
J Am Soc Nephrol ; 28(8): 2377-2392, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28302753

ABSTRACT

Although spontaneous kidney transplant acceptance/tolerance occurs in mice and occasionally in humans, mechanisms remain unclear. Herein we test the hypothesis that EPO, a hormone predominantly produced by the adult kidney, has immunomodulating properties that are required for spontaneous kidney graft acceptance. In vitro, in a manner dependent on the EPO receptor and CD131 on antigen-presenting cells, EPO induced the secretion of active TGFß by antigen-presenting cells, which in turn converted naïve CD4+ T cells into functional Foxp3+ regulatory T cells (Treg). In murine transplant models, pharmacologic downregulation of kidney-derived EPO prevented spontaneous Treg generation. In a controlled, prospective cohort clinical study, EPO administration at doses used to correct anemia augmented the frequency of peripheral CD4+CD25+CD127lo T cells in humans with CKD. Furthermore, EPO directly inhibited conventional T cell proliferation in vitro via tyrosine phosphatase SHP-1-dependent uncoupling of IL-2Rß signaling. Conversely, EPO-initiated signals facilitated Treg proliferation by augmenting IL-2Rγ signaling and maintaining constitutively quenched IL-2Rß signaling. In additional murine transplant models, recombinant EPO administration prolonged heart allograft survival, whereas pharmacologic downregulation of kidney-derived EPO reduced the expression of TGFß mRNA and abrogated kidney allograft acceptance. Together, our findings delineate the protolerogenic properties of EPO in inhibiting conventional T cells while simultaneously promoting Treg induction, and suggest that manipulating the EPO/EPO receptor signaling axis could be exploited to prevent and/or treat T cell-mediated pathologies, including transplant rejection.


Subject(s)
Graft Survival/immunology , Kidney Transplantation , Receptor Cross-Talk , Receptors, Erythropoietin/physiology , T-Lymphocytes, Regulatory/immunology , Animals , Humans , Mice , Prospective Studies
15.
J Hematol Oncol ; 9(1): 75, 2016 08 31.
Article in English | MEDLINE | ID: mdl-27581518

ABSTRACT

BACKGROUND: Multiple myeloma is an incurable complex disease characterized by clonal proliferation of malignant plasma cells in a hypoxic bone marrow environment. Hypoxia-dependent erythropoietin (EPO)-receptor (EPOR) signaling is central in various cancers, but the relevance of EPOR signaling in multiple myeloma cells has not yet been thoroughly investigated. METHODS: Myeloma cell lines and malignant plasma cells isolated from bone marrow of myeloma patients were used in this study. Transcript levels were analysed by quantitative PCR and cell surface levels of EPOR in primary cells by flow cytometry. Knockdown of EPOR by short interfering RNA was used to show specific EPOR signaling in the myeloma cell line INA-6. Flow cytometry was used to assess viability in primary cells treated with EPO in the presence and absence of neutralizing anti-EPOR antibodies. Gene expression data for total therapy 2 (TT2), total therapy 3A (TT3A) trials and APEX 039 and 040 were retrieved from NIH GEO omnibus and EBI ArrayExpress. RESULTS: We show that the EPOR is expressed in myeloma cell lines and in primary myeloma cells both at the mRNA and protein level. Exposure to recombinant human EPO (rhEPO) reduced viability of INA-6 myeloma cell line and of primary myeloma cells. This effect could be partially reversed by neutralizing antibodies against EPOR. In INA-6 cells and primary myeloma cells, janus kinase 2 (JAK-2) and extracellular signal regulated kinase 1 and 2 (ERK-1/2) were phosphorylated by rhEPO treatment. Knockdown of EPOR expression in INA-6 cells reduced rhEPO-induced phospo-JAK-2 and phospho-ERK-1/2. Co-cultures of primary myeloma cells with bone marrow-derived stroma cells did not protect the myeloma cells from rhEPO-induced cell death. In four different clinical trials, survival data linked to gene expression analysis indicated that high levels of EPOR mRNA were associated with better survival. CONCLUSIONS: Our results demonstrate for the first time active EPOR signaling in malignant plasma cells. EPO-mediated EPOR signaling reduced the viability of myeloma cell lines and of malignant primary plasma cells in vitro. Our results encourage further studies to investigate the importance of EPO/EPOR in multiple myeloma progression and treatment. TRIAL REGISTRATION: [Trial registration number for Total Therapy (TT) 2: NCT00083551 and TT3: NCT00081939 ].


Subject(s)
Cell Death , Multiple Myeloma/pathology , Receptors, Erythropoietin/physiology , Bone Marrow/pathology , Cell Survival/drug effects , Erythropoietin/pharmacology , Humans , Phosphorylation/drug effects , Protein Kinases/metabolism , RNA, Messenger/blood , Receptors, Erythropoietin/analysis , Receptors, Erythropoietin/genetics , Signal Transduction/physiology , Tumor Cells, Cultured
16.
BMC Evol Biol ; 16: 11, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26768152

ABSTRACT

BACKGROUND: Erythropoietin (EPO) is a glycoprotein hormone that plays a principal regulatory role in erythropoiesis and initiates cell homeostatic responses to environmental challenges. The Qinghai-Tibet Plateau is a natural laboratory for hypoxia adaptation. Gymnocypris dobula is a highly specialized plateau schizothoracine fish that is restricted to > 4500 m high-altitude freshwater rivers and ponds in the Qinghai-Tibet Plateau. The role of EPO in the adaptation of schizothoracine fish to hypoxia is unknown. RESULTS: The EPO and EPO receptor genes from G. dobula and four other schizothoracine fish from various altitudinal habitats were characterized. Schizothoracine EPOs are predicted to possess 2-3 N-glycosylation (NGS) sites, 4-5 casein kinase II phosphorylation (CK2) sites, 1-2 protein kinase C (PKC) phosphorylation sites, and four conserved cysteine residues within four helical domains, with variations in the numbers of NGS and CK2 sites in G. dobula. PAML analysis indicated a d N/d S value (ω) = 1.112 in the G. dobula lineage, and a few amino acids potentially under lineage-specific positive selection were detected within the G. dobula EPO. Similarly, EPO receptors of the two high-altitude schizothoracines (G. dobula and Ptychobarbus kaznakovi), were found to be statistically on the border of positive selection using the branch-site model (P-value = 0.096), and some amino acids located in the ligand-binding domain and the fibronectin type III domain were identified as potentially positive selection sites. Tissue EPO expression profiling based on transcriptome sequencing of three schizothoracines (G. dobula, Schizothorax nukiangensis Tsao, and Schizothorax prenanti) showed significant upregulation of EPO expression in the brain and less significantly in the gill of G. dobula. The elevated expression together with the rapid evolution of the EPO gene in G. dobula suggested a possible role for EPO in adaptation to hypoxia. To test this hypothesis, Gd-EPO and Sp-EPO were cloned into an expression vector and transfected into the cultured cell line 293 T. Significantly higher cell viability was observed in cells transfected with Gd-EPO than cells harboring Sp-EPO when challenged by hypoxia. CONCLUSION: The deduced EPO proteins of the schizothoracine fish contain characteristic structures and important domains similar to EPOs from other taxa. The presence of potentially positive selection sites in both EPO and EPOR in G. dobula suggest possible adaptive evolution in the ligand-receptor binding activity of the EPO signaling cascade in G. dobula. Functional study indicated that the EPO from high-altitude schizothoracine species demonstrated features of hypoxic adaptation by reducing toxic effects or improving cell survival when expressed in cultured cells, providing evidence of molecular adaptation to hypoxic conditions in the Qinghai-Tibet Plateau.


Subject(s)
Cyprinidae/physiology , Cytoprotection , Ecosystem , Erythropoietin/physiology , Oxygen , Acclimatization , Adaptation, Physiological , Animals , Biological Evolution , Cyprinidae/genetics , Erythropoietin/genetics , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/physiology , Tibet
17.
J Hypertens ; 33(6): 1267-75, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25715089

ABSTRACT

OBJECTIVE: Anemia is a common comorbidity of patients with heart failure, and iron deficiency is known as one of the causes of anemia in heart failure. Recent studies have shown that iron deficiency alone, without overt anemia, is associated with poor outcomes in patients with heart failure. Thus, to minimize the mortality in patients with heart failure, it is important to understand the link between iron deficiency and cardiac function. Chronic untreated iron deficiency results in cardiac remodeling, and we have previously reported that erythropoietin (Epo) and cardiac Epo receptor (EpoR) signaling may be associated with its remodeling. However, the link between EpoR signaling and its remodeling remains to be elucidated. Herein, we investigated the role of EpoR signaling on cardiac remodeling in response to chronic iron deficiency. METHODS: Wild-type mice and transgene-rescued EpoR-null mutant mice, which express EpoR only in the hematopoietic lineage (EpoR-restricted mice), were fed with either a normal or an iron-restricted diet, and the molecular mechanisms were investigated. RESULTS: Dietary iron restriction gradually induced anemia, Epo secretion, and cardiac hypertrophy in wild-type mice. In contrast, EpoR-restricted mice fed with an iron-restricted diet exhibited anemia, left ventricular dilatation, and cardiac dysfunction compared with wild-type mice. Interestingly, altered cardiac mitochondrial biogenesis was observed in EpoR-restricted mice following iron deficiency. Moreover, cardiac p53 expression was increased in EpoR-restricted mice compared with wild-type mice following iron deficiency. CONCLUSION: These data indicate that EpoR signaling is associated with cardiac remodeling following chronic iron deficiency.


Subject(s)
Anemia, Iron-Deficiency/complications , Heart Failure/etiology , Heart Failure/pathology , Iron Deficiencies , Myocardium/pathology , Receptors, Erythropoietin/physiology , Animals , Chronic Disease , Disease Models, Animal , Erythropoietin/metabolism , Male , Mice , Mice, Knockout , Receptors, Erythropoietin/genetics , Signal Transduction/drug effects
18.
J Exp Med ; 211(9): 1715-22, 2014 Aug 25.
Article in English | MEDLINE | ID: mdl-25092874

ABSTRACT

Ligation of erythropoietin (EPO) receptor (EPOR) JAK2 kinase complexes propagates signals within erythroid progenitor cells (EPCs) that are essential for red blood cell production. To reveal hypothesized novel EPOR/JAK2 targets, a phosphotyrosine (PY) phosphoproteomics approach was applied. Beyond known signal transduction factors, 32 new targets of EPO-modulated tyrosine phosphorylation were defined. Molecular adaptors comprised one major set including growth factor receptor-bound protein 2 (GRB2)-associated binding proteins 1-3 (GAB1-3), insulin receptor substrate 2 (IRS2), docking protein 1 (DOK1), Src homology 2 domain containing transforming protein 1 (SHC1), and sprouty homologue 1 (SPRY1) as validating targets, and SPRY2, SH2 domain containing 2A (SH2D2A), and signal transducing adaptor molecule 2 (STAM2) as novel candidate adaptors together with an ORF factor designated as regulator of human erythroid cell expansion (RHEX). RHEX is well conserved in Homo sapiens and primates but absent from mouse, rat, and lower vertebrate genomes. Among tissues and lineages, RHEX was elevated in EPCs, occurred as a plasma membrane protein, was rapidly PY-phosphorylated >20-fold upon EPO exposure, and coimmunoprecipitated with the EPOR. In UT7epo cells, knockdown of RHEX inhibited EPO-dependent growth. This was associated with extracellular signal-regulated kinase 1,2 (ERK1,2) modulation, and RHEX coupling to GRB2. In primary human EPCs, shRNA knockdown studies confirmed RHEX regulation of erythroid progenitor expansion and further revealed roles in promoting the formation of hemoglobinizing erythroblasts. RHEX therefore comprises a new EPO/EPOR target and regulator of human erythroid cell expansion that additionally acts to support late-stage erythroblast development.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Erythroblasts/cytology , Erythroblasts/physiology , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/physiology , Erythropoietin/physiology , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Animals , Cell Line , Cells, Cultured , Erythropoiesis/physiology , Gene Knockdown Techniques , Humans , Janus Kinase 2/metabolism , Molecular Sequence Data , Proteomics , Receptors, Erythropoietin/physiology , Signal Transduction
19.
Int J Cardiol ; 171(2): 116-25, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24377712

ABSTRACT

Erythropoietin (Epo) has been thought to act exclusively on erythroid progenitor cells. The identification of Epo receptor (EpoR) in non-haematopoietic cells and tissues including neurons, astrocytes, microglia, immune cells, cancer cell lines, endothelial cells, bone marrow stromal cells, as well as cells of myocardium, reproductive system, gastrointestinal tract, kidney, pancreas and skeletal muscle indicates that Epo has pleiotropic actions. Epo shows signals through protein kinases, anti-apoptotic proteins and transcription factors. In light of interest of administering recombinant human erythropoietin (rhEpo) and its analogues for limiting infarct size and left ventricular (LV) remodelling after acute myocardial infarction (AMI) in humans, the foremost studies utilising rhEpo are reviewed. The putative mechanisms involved in Epo-induced cardioprotection are related to the antiapoptotic, anti-inflammatory and angiogenic effects of Epo. Thus, cardioprotective potentials of rhEpo are reviewed in this article by focusing on clinical applicability. An overview of non-haematopoietic Epo analogues, which are a reliable alternative to the classic EpoR agonists and may prevent undesired side effects, is also provided.


Subject(s)
Airway Remodeling/physiology , Erythropoietin/physiology , Heart/physiology , Neovascularization, Physiologic/physiology , Receptors, Erythropoietin/physiology , Cardiotonic Agents , Erythropoietin/therapeutic use , Humans
20.
Kidney Int ; 86(1): 34-9, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24402094

ABSTRACT

Treatment of anemia remains an important component in the care of patients with nondialysis chronic kidney disease (CKD) and end-stage renal disease (ESRD). Erythropoietin-stimulating agents (ESAs) remains a key anemia treatment strategy in this patient population. However, anemia management in this group can become more complicated by prior or current history of malignancy. There has been a great deal of work both scientifically and in clinical trials in oncology that have revealed certain concerns and risks of ESA use in patients with cancer. In this review, we will bring together knowledge from nephrology and oncology literature to help nephrologists understand the implications for ESA treatment when CKD/ESRD is complicated by cancer. We also suggest an approach to the management of anemia in this patient group with active or previous malignancy.


Subject(s)
Hematinics/adverse effects , Hematinics/therapeutic use , Neoplasms/complications , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/drug therapy , Anemia/blood , Anemia/drug therapy , Anemia/etiology , Contraindications , Erythropoietin/genetics , Erythropoietin/physiology , Female , Humans , Male , Neoplasms/blood , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/physiology , Renal Insufficiency, Chronic/blood , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...