1
|
Kaya M, Cicek N, Guven S, Alpay H, Gokce I. Resistance to Epoetin-Stimulating Agents in Children Receiving Renal Replacement Therapy. Clin Pediatr (Phila) 2024:99228241299893. [PMID: 39690475 DOI: 10.1177/00099228241299893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
The incidence of anemia increases with the stage of chronic kidney disease (CKD). Erythropoietin (EPO) deficiency is a common cause of anemia in CKD. Erythropoietin-stimulating agents (ESAs) are the mainstay of the treatment. Treatment can be challenging due to erythropoietin resistance (ER), which can be assessed using the erythropoietin resistance index (ERI). Our aim was to investigate the factors contributing to high ERI levels in children receiving renal replacement therapy (RRT). Thirty-three children receiving RRT for at least 3 months were included. The mean ERI value was 15.7 IU/kg/w/g/dL. A significant association was observed between serum phosphorus levels and ERI (P = .016, r = 0.41). The mean parathormone (PTH) level was also higher in the high ERI group (599 ± 351 vs 392 ± 320 pg/mL, P = .088). An association, approaching statistical significance, was found between ERI and hypertension (P = .06, r = 0.32). Our study indicated a potential relationship between hyperphosphatemia, possibly secondary hyperparathyroidism, and ERI in children undergoing RRT.
Collapse
Affiliation(s)
- Mehtap Kaya
- Division of Pediatric Nephrology, Department of Pediatrics, School of Medicine, Marmara University, Istanbul, Turkey
| | - Neslihan Cicek
- Division of Pediatric Nephrology, Department of Pediatrics, School of Medicine, Marmara University, Istanbul, Turkey
| | - Sercin Guven
- Division of Pediatric Nephrology, Department of Pediatrics, School of Medicine, Marmara University, Istanbul, Turkey
| | - Harika Alpay
- Division of Pediatric Nephrology, Department of Pediatrics, School of Medicine, Marmara University, Istanbul, Turkey
| | - Ibrahim Gokce
- Division of Pediatric Nephrology, Department of Pediatrics, School of Medicine, Marmara University, Istanbul, Turkey
| |
Collapse
|
2
|
Stockhausen S, Kilani B, Schubert I, Steinsiek AL, Chandraratne S, Wendler F, Eivers L, von Brühl ML, Massberg S, Ott I, Stark K. Differential Effects of Erythropoietin Administration and Overexpression on Venous Thrombosis in Mice. Thromb Haemost 2024; 124:1027-1039. [PMID: 37846465 PMCID: PMC11518618 DOI: 10.1055/s-0043-1775965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 08/06/2023] [Indexed: 10/18/2023]
Abstract
BACKGROUND Deep vein thrombosis (DVT) is a common condition associated with significant mortality due to pulmonary embolism. Despite advanced prevention and anticoagulation therapy, the incidence of venous thromboembolism remains unchanged. Individuals with elevated hematocrit and/or excessively high erythropoietin (EPO) serum levels are particularly susceptible to DVT formation. We investigated the influence of short-term EPO administration compared to chronic EPO overproduction on DVT development. Additionally, we examined the role of the spleen in this context and assessed its impact on thrombus composition. METHODS We induced ligation of the caudal vena cava (VCC) in EPO-overproducing Tg(EPO) mice as well as wildtype mice treated with EPO for two weeks, both with and without splenectomy. The effect on platelet circulation time was evaluated through FACS analysis, and thrombus composition was analyzed using immunohistology. RESULTS We present evidence for an elevated thrombogenic phenotype resulting from chronic EPO overproduction, achieved by combining an EPO-overexpressing mouse model with experimental DVT induction. This increased thrombotic state is largely independent of traditional contributors to DVT, such as neutrophils and platelets. Notably, the pronounced prothrombotic effect of red blood cells (RBCs) only manifests during chronic EPO overproduction and is not influenced by splenic RBC clearance, as demonstrated by splenectomy. In contrast, short-term EPO treatment does not induce thrombogenesis in mice. Consequently, our findings support the existence of a differential thrombogenic effect between chronic enhanced erythropoiesis and exogenous EPO administration. CONCLUSION Chronic EPO overproduction significantly increases the risk of DVT, while short-term EPO treatment does not. These findings underscore the importance of considering EPO-related factors in DVT risk assessment and potential therapeutic strategies.
Collapse
Affiliation(s)
- Sven Stockhausen
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
- Walter-Brendel Center of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
| | - Badr Kilani
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
- Walter-Brendel Center of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
| | - Irene Schubert
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
- Walter-Brendel Center of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
| | | | - Sue Chandraratne
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
- Walter-Brendel Center of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
| | - Franziska Wendler
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
- Walter-Brendel Center of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
| | - Luke Eivers
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
- Walter-Brendel Center of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
| | - Marie-Luise von Brühl
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
- Walter-Brendel Center of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
| | - Steffen Massberg
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
- Walter-Brendel Center of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
| | - Ilka Ott
- Department of cardiology, German Heart Center, Munich, Germany.
| | - Konstantin Stark
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
- Walter-Brendel Center of Experimental Medicine, Ludwig-Maximilians-Universität, Munich, Germany
| |
Collapse
|
3
|
Yin W, Rajvanshi PK, Rogers HM, Yoshida T, Kopp JB, An X, Gassmann M, Noguchi CT. Erythropoietin regulates energy metabolism through EPO-EpoR-RUNX1 axis. Nat Commun 2024; 15:8114. [PMID: 39284834 PMCID: PMC11405798 DOI: 10.1038/s41467-024-52352-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 09/02/2024] [Indexed: 09/20/2024] Open
Abstract
Erythropoietin (EPO) plays a key role in energy metabolism, with EPO receptor (EpoR) expression in white adipose tissue (WAT) mediating its metabolic activity. Here, we show that male mice lacking EpoR in adipose tissue exhibit increased fat mass and susceptibility to diet-induced obesity. Our findings indicate that EpoR is present in WAT, brown adipose tissue, and skeletal muscle. Elevated EPO in male mice improves glucose tolerance and insulin sensitivity while reducing expression of lipogenic-associated genes in WAT, which is linked to an increase in transcription factor RUNX1 that directly inhibits lipogenic genes expression. EPO treatment in wild-type male mice decreases fat mass and lipogenic gene expression and increase in RUNX1 protein in adipose tissue which is not observed in adipose tissue EpoR ablation mice. EPO treatment decreases WAT ubiquitin ligase FBXW7 expression and increases RUNX1 stability, providing evidence that EPO regulates energy metabolism in male mice through the EPO-EpoR-RUNX1 axis.
Collapse
Affiliation(s)
- Weiqin Yin
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Praveen Kumar Rajvanshi
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Heather M Rogers
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Teruhiko Yoshida
- Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Jeffrey B Kopp
- Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Xiuli An
- Laboratory of Membrane Biology, New York Blood Center, New York, NY, USA
| | - Max Gassmann
- Institute of Veterinary Physiology and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Constance T Noguchi
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD, USA.
| |
Collapse
|
4
|
Lee J, Rogers HM, Springer DA, Noguchi CT. Neuronal nitric oxide synthase required for erythropoietin modulation of heart function in mice. Front Physiol 2024; 15:1338476. [PMID: 38628440 PMCID: PMC11019009 DOI: 10.3389/fphys.2024.1338476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 03/04/2024] [Indexed: 04/19/2024] Open
Abstract
Introduction: Erythropoietin (EPO) acts primarily in regulating red blood cell production mediated by high EPO receptor (EPOR) expression in erythroid progenitor cells. EPO activity in non-erythroid tissue is evident in mice with EPOR restricted to erythroid tissues (ΔEPORE) that become obese, glucose-intolerant, and insulin-resistant. In animal models, nitric oxide synthase (NOS) contributes to EPO activities including erythropoiesis, neuroprotection, and cardioprotection against ischemia-reperfusion injury. However, we found that extended EPO treatment to increase hematocrit compromised heart function, while the loss of neuronal NOS (nNOS) was protective against the deleterious activity of EPO to promote heart failure. Methods: Wild-type (WT) mice, ΔEPORE mice, and nNOS-knockout mice (nNOS-/-) were placed on a high-fat diet to match the ΔEPORE obese phenotype and were treated with EPO for 3 weeks. Hematocrit and metabolic response to EPO treatment were monitored. Cardiac function was assessed by echocardiography and ultrasonography. Results: ΔEPORE mice showed a decrease in the left ventricular outflow tract (LVOT) peak velocity, ejection fraction, and fractional shortening, showing that endogenous non-erythroid EPO response is protective for heart function. EPO treatment increased hematocrit in all mice and decreased fat mass in male WT, demonstrating that EPO regulation of fat mass requires non-erythroid EPOR. EPO treatment also compromised heart function in WT mice, and decreased the pulmonary artery peak velocity (PA peak velocity), LVOT peak velocity, ejection fraction, and fractional shortening, but it had minimal effect in further reducing the heart function in ΔEPORE mice, indicating that the adverse effect of EPO on heart function is not related to EPO-stimulated erythropoiesis. ΔEPORE mice had increased expression of heart failure-associated genes, hypertrophic cardiomyopathy-related genes, and sarcomeric genes that were also elevated with EPO treatment in WT mice. Male and female nNOS-/- mice were protected against diet-induced obesity. EPO treatment in nNOS-/- mice increased the hematocrit that tended to be lower than WT mice and decreased the PA peak velocity but did not affect the LVOT peak velocity, ejection fraction, and fractional shortening, suggesting that nNOS is required for the adverse effect of EPO treatment on heart function in WT mice. EPO treatment did not change expression of heart failure-associated gene expression in nNOS-/- mice. Discussion: Endogenous EPO has a protective effect on heart function. With EPO administration, in contrast to the protective effect to the cardiac injury of acute EPO treatment, extended EPO treatment to increase hematocrit in WT mice adversely affected the heart function with a corresponding increase in expression of heart failure-associated genes. This EPO activity was independent of EPO-stimulated erythropoiesis and required EPOR in non-erythroid tissue and nNOS activity, while nNOS-/- mice were protected from the EPO-associated adverse effect on heart function. These data provide evidence that nNOS contributes to the negative impact on the heart function of high-dose EPO treatment for anemia.
Collapse
Affiliation(s)
- Jeeyoung Lee
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Heather M. Rogers
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Danielle A. Springer
- Murine Phenotyping Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Constance T. Noguchi
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
5
|
Muttathukunnel P, Wälti M, Aboouf MA, Köster-Hegmann C, Haenggi T, Gassmann M, Pannzanelli P, Fritschy JM, Schneider Gasser EM. Erythropoietin regulates developmental myelination in the brain stimulating postnatal oligodendrocyte maturation. Sci Rep 2023; 13:19522. [PMID: 37945644 PMCID: PMC10636124 DOI: 10.1038/s41598-023-46783-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/05/2023] [Indexed: 11/12/2023] Open
Abstract
Myelination is a process tightly regulated by a variety of neurotrophic factors. Here, we show-by analyzing two transgenic mouse lines, one overexpressing EPO selectively in the brain Tg21(PDGFB-rhEPO) and another with targeted removal of EPO receptors (EPORs) from oligodendrocyte progenitor cells (OPC)s (Sox10-cre;EpoRfx/fx mice)-a key function for EPO in regulating developmental brain myelination. Overexpression of EPO resulted in faster postnatal brain growth and myelination, an increased number of myelinating oligodendrocytes, faster axonal myelin ensheathment, and improved motor coordination. Conversely, targeted ablation of EPORs from OPCs reduced the number of mature oligodendrocytes and impaired motor coordination during the second postnatal week. Furthermore, we found that EPORs are transiently expressed in the subventricular zone (SVZ) during the second postnatal week and EPO increases the postnatal expression of essential oligodendrocyte pro-differentiation and pro-maturation (Nkx6.2 and Myrf) transcripts, and the Nfatc2/calcineurin pathway. In contrast, ablation of EPORs from OPCs inactivated the Erk1/2 pathway and reduced the postnatal expression of the transcripts. Our results reveal developmental time windows in which EPO therapies could be highly effective for stimulating oligodendrocyte maturation and myelination.
Collapse
Affiliation(s)
- Paola Muttathukunnel
- Institute of Pharmacology and Toxicology, University of Zürich, 8057, Zürich, Switzerland
- Center for Neuroscience Zurich (ZNZ), Zurich, Switzerland
| | - Michael Wälti
- Institute of Pharmacology and Toxicology, University of Zürich, 8057, Zürich, Switzerland
| | - Mostafa A Aboouf
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zürich, 8057, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057, Zurich, Switzerland
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| | - Christina Köster-Hegmann
- Institute of Pharmacology and Toxicology, University of Zürich, 8057, Zürich, Switzerland
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zürich, 8057, Zurich, Switzerland
| | - Tatjana Haenggi
- Institute of Pharmacology and Toxicology, University of Zürich, 8057, Zürich, Switzerland
| | - Max Gassmann
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zürich, 8057, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057, Zurich, Switzerland
| | - Patrizia Pannzanelli
- Rita Levi Montalcini Center for Brain Repair, University of Turin, 10126, Turin, Italy
| | - Jean-Marc Fritschy
- Institute of Pharmacology and Toxicology, University of Zürich, 8057, Zürich, Switzerland
- Center for Neuroscience Zurich (ZNZ), Zurich, Switzerland
| | - Edith M Schneider Gasser
- Institute of Pharmacology and Toxicology, University of Zürich, 8057, Zürich, Switzerland.
- Center for Neuroscience Zurich (ZNZ), Zurich, Switzerland.
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zürich, 8057, Zurich, Switzerland.
| |
Collapse
|
6
|
Emery JM, Chicana B, Taglinao H, Ponce C, Donham C, Padmore H, Sebastian A, Trasti SL, Manilay JO. Vhl deletion in Dmp1 -expressing cells alters MEP metabolism and promotes stress erythropoiesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.25.550559. [PMID: 37546957 PMCID: PMC10402046 DOI: 10.1101/2023.07.25.550559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
In recent years, general hypoxia-inducible factor (HIF)-prolyl hydroxylase (PHD) enzyme inhibitors have been developed for the treatment of anemia due to renal disease and osteoporosis. However, it remains a challenge to target the HIF signaling pathway without dysregulating the skeletal and hematopoietic system. Here, we examined the effects of Vhl deletion in bone by performing longitudinal analyses of Vhl cKO mice at 3, 6, 10, and 24 weeks of age, where at 10 and 24 weeks of age, high bone mass and splenomegaly are present. Using flow cytometry, we observed increased frequency (%) of CD71 lo TER119 hi FSC lo orthochromatophilic erythroblasts and reticulocytes in 10- and 24-week-old Vhl cKO bone marrow (BM), which correlated with elevated erythropoietin levels in the BM and increased number of red blood cells in circulation. The absolute numbers of myeloerythroid progenitors (MEPs) in the BM were significantly reduced at 24 weeks. Bulk RNA-Seq of the MEPs showed upregulation of Epas1 ( Hif1a) and Efnb2 ( Hif2a) in Vhl cKO MEPs, consistent with a response to hypoxia, and genes involved in erythrocyte development, actin filament organization, and response to glucose. Additionally, histological analysis of Vhl cKO spleens revealed red pulp hyperplasia and the presence of megakaryocytes, both of which are features of extramedullary hematopoiesis (EMH). EMH in the spleen was correlated with the presence of mature stress erythroid progenitors, suggesting that stress erythropoiesis is occurring to compensate for the BM microenvironmental irregularities. Our studies implicate that HIF-driven alterations in skeletal homeostasis can accelerate erythropoiesis. Key Points • Dysregulation of HIF signaling in Dmp1+ bone cells induces stress erythropoiesis.• Skeletal homeostasis modulates erythropoiesis.
Collapse
|
7
|
Kittur FS, Hung CY, Li PA, Sane DC, Xie J. Asialo-rhuEPO as a Potential Neuroprotectant for Ischemic Stroke Treatment. Pharmaceuticals (Basel) 2023; 16:610. [PMID: 37111367 PMCID: PMC10143832 DOI: 10.3390/ph16040610] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/11/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Neuroprotective drugs to protect the brain against cerebral ischemia and reperfusion (I/R) injury are urgently needed. Mammalian cell-produced recombinant human erythropoietin (rhuEPOM) has been demonstrated to have excellent neuroprotective functions in preclinical studies, but its neuroprotective properties could not be consistently translated in clinical trials. The clinical failure of rhuEPOM was thought to be mainly due to its erythropoietic activity-associated side effects. To exploit its tissue-protective property, various EPO derivatives with tissue-protective function only have been developed. Among them, asialo-rhuEPO, lacking terminal sialic acid residues, was shown to be neuroprotective but non-erythropoietic. Asialo-rhuEPO can be prepared by enzymatic removal of sialic acid residues from rhuEPOM (asialo-rhuEPOE) or by expressing human EPO gene in glycoengineered transgenic plants (asialo-rhuEPOP). Both types of asialo-rhuEPO, like rhuEPOM, displayed excellent neuroprotective effects by regulating multiple cellular pathways in cerebral I/R animal models. In this review, we describe the structure and properties of EPO and asialo-rhuEPO, summarize the progress on neuroprotective studies of asialo-rhuEPO and rhuEPOM, discuss potential reasons for the clinical failure of rhuEPOM with acute ischemic stroke patients, and advocate future studies needed to develop asialo-rhuEPO as a multimodal neuroprotectant for ischemic stroke treatment.
Collapse
Affiliation(s)
- Farooqahmed S. Kittur
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute & Technology Enterprise, North Carolina Central University, Durham, NC 27707, USA; (C.-Y.H.); (P.A.L.)
| | - Chiu-Yueh Hung
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute & Technology Enterprise, North Carolina Central University, Durham, NC 27707, USA; (C.-Y.H.); (P.A.L.)
| | - P. Andy Li
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute & Technology Enterprise, North Carolina Central University, Durham, NC 27707, USA; (C.-Y.H.); (P.A.L.)
| | - David C. Sane
- Carilion Clinic and Virginia Tech Carilion School of Medicine, Roanoke, VA 24014, USA;
| | - Jiahua Xie
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute & Technology Enterprise, North Carolina Central University, Durham, NC 27707, USA; (C.-Y.H.); (P.A.L.)
| |
Collapse
|
8
|
Al-Rifai R, Vandestienne M, Lavillegrand JR, Mirault T, Cornebise J, Poisson J, Laurans L, Esposito B, James C, Mansier O, Hirsch P, Favale F, Braik R, Knosp C, Vilar J, Rizzo G, Zernecke A, Saliba AE, Tedgui A, Lacroix M, Arrive L, Mallat Z, Taleb S, Diedisheim M, Cochain C, Rautou PE, Ait-Oufella H. JAK2V617F mutation drives vascular resident macrophages toward a pathogenic phenotype and promotes dissecting aortic aneurysm. Nat Commun 2022; 13:6592. [PMID: 36329047 PMCID: PMC9633755 DOI: 10.1038/s41467-022-34469-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022] Open
Abstract
JAK2V617F mutation is associated with an increased risk for athero-thrombotic cardiovascular disease, but its role in aortic disease development and complications remains unknown. In a cohort of patients with myeloproliferative neoplasm, JAK2V617F mutation was identified as an independent risk factor for dilation of both the ascending and descending thoracic aorta. Using single-cell RNA-seq, complementary genetically-modified mouse models, as well as pharmacological approaches, we found that JAK2V617F mutation was associated with a pathogenic pro-inflammatory phenotype of perivascular tissue-resident macrophages, which promoted deleterious aortic wall remodeling at early stages, and dissecting aneurysm through the recruitment of circulating monocytes at later stages. Finally, genetic manipulation of tissue-resident macrophages, or treatment with a Jak2 inhibitor, ruxolitinib, mitigated aortic wall inflammation and reduced aortic dilation and rupture. Overall, JAK2V617F mutation drives vascular resident macrophages toward a pathogenic phenotype and promotes dissecting aortic aneurysm.
Collapse
Affiliation(s)
- Rida Al-Rifai
- grid.462416.30000 0004 0495 1460Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | - Marie Vandestienne
- grid.462416.30000 0004 0495 1460Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | - Jean-Rémi Lavillegrand
- grid.462416.30000 0004 0495 1460Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | - Tristan Mirault
- grid.462416.30000 0004 0495 1460Université Paris Cité, Inserm, PARCC, F-75015, Paris, France ,Service de médecine vasculaire, Hopital Européen G. Pompidou, Paris, France
| | - Julie Cornebise
- grid.462416.30000 0004 0495 1460Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | - Johanne Poisson
- grid.462416.30000 0004 0495 1460Université Paris Cité, Inserm, PARCC, F-75015, Paris, France ,Service de gériatrie, Hopital Européen G. Pompidou, Paris, France ,grid.462374.00000 0004 0620 6317Centre de recherche sur l’inflammation, Inserm, Paris, France
| | - Ludivine Laurans
- grid.462416.30000 0004 0495 1460Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | - Bruno Esposito
- grid.462416.30000 0004 0495 1460Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | - Chloé James
- Université de Bordeaux, UMR1034, Inserm, Biology of Cardiovascular Diseases, CHU de Bordeaux, Laboratoire d’Hématologie, Pessac, France
| | - Olivier Mansier
- Université de Bordeaux, UMR1034, Inserm, Biology of Cardiovascular Diseases, CHU de Bordeaux, Laboratoire d’Hématologie, Pessac, France
| | - Pierre Hirsch
- grid.412370.30000 0004 1937 1100Laboratoire d’Hématologie, Hôpital Saint-Antoine, AP-HP, Paris, France
| | - Fabrizia Favale
- grid.412370.30000 0004 1937 1100Laboratoire d’Hématologie, Hôpital Saint-Antoine, AP-HP, Paris, France
| | - Rayan Braik
- grid.462416.30000 0004 0495 1460Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | - Camille Knosp
- grid.462416.30000 0004 0495 1460Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | - Jose Vilar
- grid.462416.30000 0004 0495 1460Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | - Giuseppe Rizzo
- grid.411760.50000 0001 1378 7891Institute of Experimental Biomedicine, University Hospital Wuerzburg, Würzburg, Germany
| | - Alma Zernecke
- grid.411760.50000 0001 1378 7891Institute of Experimental Biomedicine, University Hospital Wuerzburg, Würzburg, Germany
| | - Antoine-Emmanuel Saliba
- grid.498164.6Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), Würzburg, Germany
| | - Alain Tedgui
- grid.462416.30000 0004 0495 1460Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | - Maxime Lacroix
- grid.412370.30000 0004 1937 1100Service de radiologie, Hôpital Saint-Antoine, AP-HP, Paris, France
| | - Lionel Arrive
- grid.412370.30000 0004 1937 1100Service de radiologie, Hôpital Saint-Antoine, AP-HP, Paris, France
| | - Ziad Mallat
- grid.462416.30000 0004 0495 1460Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | - Soraya Taleb
- grid.462416.30000 0004 0495 1460Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | - Marc Diedisheim
- grid.411784.f0000 0001 0274 3893GlandOmics, 41700 Cheverny, & Department of Diabetology, AP-HP, Hôpital Cochin, Paris, France
| | - Clément Cochain
- grid.411760.50000 0001 1378 7891Institute of Experimental Biomedicine, University Hospital Wuerzburg, Würzburg, Germany
| | - Pierre-Emmanuel Rautou
- grid.462416.30000 0004 0495 1460Université Paris Cité, Inserm, PARCC, F-75015, Paris, France ,grid.462374.00000 0004 0620 6317Centre de recherche sur l’inflammation, Inserm, Paris, France ,grid.411599.10000 0000 8595 4540AP-HP, Hôpital Beaujon, Service d’Hépatologie, DMU DIGEST, Centre de Référence des Maladies Vasculaires du Foie, FILFOIE, ERN RARE-LIVER, Clichy, France
| | - Hafid Ait-Oufella
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France. .,Medical Intensive Care Unit, Hôpital Saint-Antoine, AP-HP, Sorbonne Université, Paris, France.
| |
Collapse
|
9
|
Kralova B, Sochorcova L, Song J, Jahoda O, Hlusickova Kapralova K, Prchal JT, Divoky V, Horvathova M. Developmental changes in iron metabolism and erythropoiesis in mice with human gain-of-function erythropoietin receptor. Am J Hematol 2022; 97:1286-1299. [PMID: 35815815 DOI: 10.1002/ajh.26658] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 07/05/2022] [Indexed: 01/24/2023]
Abstract
Iron availability for erythropoiesis is controlled by the iron-regulatory hormone hepcidin. Increased erythropoiesis negatively regulates hepcidin synthesis by erythroferrone (ERFE), a hormone produced by erythroid precursors in response to erythropoietin (EPO). The mechanisms coordinating erythropoietic activity with iron homeostasis in erythrocytosis with low EPO are not well defined as exemplified by dominantly inherited (heterozygous) gain-of-function mutation of human EPO receptor (mtHEPOR) with low EPO characterized by postnatal erythrocytosis. We previously created a mouse model of this mtHEPOR that develops fetal erythrocytosis with a transient perinatal amelioration of erythrocytosis and its reappearance at 3-6 weeks of age. Prenatally and perinatally, mtHEPOR heterozygous and homozygous mice (differing in erythrocytosis severity) had increased Erfe transcripts, reduced hepcidin, and iron deficiency. Epo was transiently normal in the prenatal life; then decreased at postnatal day 7, and remained reduced in adulthood. Postnatally, hepcidin increased in mtHEPOR heterozygotes and homozygotes, accompanied by low Erfe induction and iron accumulation. With aging, the old, especially mtHEPOR homozygotes had a decline of erythropoiesis, myeloid expansion, and local bone marrow inflammatory stress. In addition, mtHEPOR erythrocytes had a reduced lifespan. This, together with reduced iron demand for erythropoiesis, due to its age-related attenuation, likely contributes to increased iron deposition in the aged mtHEPOR mice. In conclusion, the erythroid drive-mediated inhibition of hepcidin production in mtHEPOR mice in the prenatal/perinatal period is postnatally abrogated by increasing iron stores promoting hepcidin synthesis. The differences observed in studied characteristics between mtHEPOR heterozygotes and homozygotes suggest dose-dependent alterations of downstream EPOR stimulation.
Collapse
Affiliation(s)
- Barbora Kralova
- Department of Biology, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Lucie Sochorcova
- Department of Biology, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Jihyun Song
- Division of Hematology & Hematologic Malignancies, The University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Ondrej Jahoda
- Department of Biology, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | | | - Josef T Prchal
- Division of Hematology & Hematologic Malignancies, The University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Vladimir Divoky
- Department of Biology, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Monika Horvathova
- Department of Biology, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| |
Collapse
|
10
|
Aboouf MA, Guscetti F, von Büren N, Armbruster J, Ademi H, Ruetten M, Meléndez-Rodríguez F, Rülicke T, Seymer A, Jacobs RA, Schneider Gasser EM, Aragones J, Neumann D, Gassmann M, Thiersch M. Erythropoietin receptor regulates tumor mitochondrial biogenesis through iNOS and pAKT. Front Oncol 2022; 12:976961. [PMID: 36052260 PMCID: PMC9425774 DOI: 10.3389/fonc.2022.976961] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Erythropoietin receptor (EPOR) is widely expressed in healthy and malignant tissues. In certain malignancies, EPOR stimulates tumor growth. In healthy tissues, EPOR controls processes other than erythropoiesis, including mitochondrial metabolism. We hypothesized that EPOR also controls the mitochondrial metabolism in cancer cells. To test this hypothesis, we generated EPOR-knockdown cancer cells to grow tumor xenografts in mice and analyzed tumor cellular respiration via high-resolution respirometry. Furthermore, we analyzed cellular respiratory control, mitochondrial content, and regulators of mitochondrial biogenesis in vivo and in vitro in different cancer cell lines. Our results show that EPOR controls tumor growth and mitochondrial biogenesis in tumors by controlling the levels of both, pAKT and inducible NO synthase (iNOS). Furthermore, we observed that the expression of EPOR is associated with the expression of the mitochondrial marker VDAC1 in tissue arrays of lung cancer patients, suggesting that EPOR indeed helps to regulate mitochondrial biogenesis in tumors of cancer patients. Thus, our data imply that EPOR not only stimulates tumor growth but also regulates tumor metabolism and is a target for direct intervention against progression.
Collapse
Affiliation(s)
- Mostafa A. Aboouf
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
- Center for Clinical Studies, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Franco Guscetti
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Nadine von Büren
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
- Center for Clinical Studies, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Julia Armbruster
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
- Center for Clinical Studies, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Hyrije Ademi
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
- Center for Clinical Studies, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Maja Ruetten
- PathoVet AG, Pathology Diagnostic Laboratory, Tagelswangen, Switzerland
| | | | - Thomas Rülicke
- Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Alexander Seymer
- Department for Sociology and Social Geography, Paris Lodron University of Salzburg (PLUS), Salzburg, Austria
| | - Robert A. Jacobs
- Department of Human Physiology & Nutrition, University of Colorado Colorado Springs (UCCS), Colorado Springs, CO, United States
| | - Edith M. Schneider Gasser
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
- Center of Neuroscience Zurich (ZNZ), University of Zurich, Zurich, Switzerland
| | - Julian Aragones
- Hospital Universitario Santa Cristina, Autonomous University of Madrid, Madrid, Spain
| | - Drorit Neumann
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Max Gassmann
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Markus Thiersch
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
- Center for Clinical Studies, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
- *Correspondence: Markus Thiersch,
| |
Collapse
|
11
|
Dahl SL, Bapst AM, Khodo SN, Scholz CC, Wenger RH. Fount, fate, features, and function of renal erythropoietin-producing cells. Pflugers Arch 2022; 474:783-797. [PMID: 35750861 PMCID: PMC9338912 DOI: 10.1007/s00424-022-02714-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 05/18/2022] [Accepted: 05/27/2022] [Indexed: 12/19/2022]
Abstract
Renal erythropoietin (Epo)-producing (REP) cells represent a rare and incompletely understood cell type. REP cells are fibroblast-like cells located in close proximity to blood vessels and tubules of the corticomedullary border region. Epo mRNA in REP cells is produced in a pronounced "on-off" mode, showing transient transcriptional bursts upon exposure to hypoxia. In contrast to "ordinary" fibroblasts, REP cells do not proliferate ex vivo, cease to produce Epo, and lose their identity following immortalization and prolonged in vitro culture, consistent with the loss of Epo production following REP cell proliferation during tissue remodelling in chronic kidney disease. Because Epo protein is usually not detectable in kidney tissue, and Epo mRNA is only transiently induced under hypoxic conditions, transgenic mouse models have been developed to permanently label REP cell precursors, active Epo producers, and inactive descendants. Future single-cell analyses of the renal stromal compartment will identify novel characteristic markers of tagged REP cells, which will provide novel insights into the regulation of Epo expression in this unique cell type.
Collapse
Affiliation(s)
- Sophie L Dahl
- Institute of Physiology and National Centre of Competence in Research "Kidney.CH", University of Zürich, CH-8057, Zurich, Switzerland
| | - Andreas M Bapst
- Institute of Physiology and National Centre of Competence in Research "Kidney.CH", University of Zürich, CH-8057, Zurich, Switzerland
| | - Stellor Nlandu Khodo
- Institute of Physiology and National Centre of Competence in Research "Kidney.CH", University of Zürich, CH-8057, Zurich, Switzerland
| | - Carsten C Scholz
- Institute of Physiology and National Centre of Competence in Research "Kidney.CH", University of Zürich, CH-8057, Zurich, Switzerland
- Institute of Physiology, University Medicine Greifswald, D-17475, Greifswald, Germany
| | - Roland H Wenger
- Institute of Physiology and National Centre of Competence in Research "Kidney.CH", University of Zürich, CH-8057, Zurich, Switzerland.
| |
Collapse
|
12
|
Ranasinghe R, Mathai M, Zulli A. Revisiting the therapeutic potential of tocotrienol. Biofactors 2022; 48:813-856. [PMID: 35719120 PMCID: PMC9544065 DOI: 10.1002/biof.1873] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 05/13/2022] [Indexed: 12/14/2022]
Abstract
The therapeutic potential of the tocotrienol group stems from its nutraceutical properties as a dietary supplement. It is largely considered to be safe when consumed at low doses for attenuating pathophysiology as shown by animal models, in vitro assays, and ongoing human trials. Medical researchers and the allied sciences have experimented with tocotrienols for many decades, but its therapeutic potential was limited to adjuvant or concurrent treatment regimens. Recent studies have focused on targeted drug delivery by enhancing the bioavailability through carriers, self-sustained emulsions, nanoparticles, and ethosomes. Epigenetic modulation and computer remodeling are other means that will help increase chemosensitivity. This review will focus on the systemic intracellular anti-cancer, antioxidant, and anti-inflammatory mechanisms that are stimulated and/or regulated by tocotrienols while highlighting its potent therapeutic properties in a diverse group of clinical diseases.
Collapse
Affiliation(s)
- Ranmali Ranasinghe
- Institute of Health and Sport, College of Health and MedicineVictoria UniversityMelbourneVictoriaAustralia
| | - Michael Mathai
- Institute of Health and Sport, College of Health and MedicineVictoria UniversityMelbourneVictoriaAustralia
| | - Anthony Zulli
- Institute of Health and Sport, College of Health and MedicineVictoria UniversityMelbourneVictoriaAustralia
| |
Collapse
|
13
|
Kaur D, Behl T, Sehgal A, Singh S, Sharma N, Badavath VN, Ul Hassan SS, Hasan MM, Bhatia S, Al-Harassi A, Khan H, Bungau S. Unravelling the potential neuroprotective facets of erythropoietin for the treatment of Alzheimer's disease. Metab Brain Dis 2022; 37:1-16. [PMID: 34436747 DOI: 10.1007/s11011-021-00820-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 08/09/2021] [Indexed: 02/06/2023]
Abstract
During the last three decades, recombinant DNA technology has produced a wide range of hematopoietic and neurotrophic growth factors, including erythropoietin (EPO), which has emerged as a promising protein drug in the treatment of several diseases. Cumulative studies have recently indicated the neuroprotective role of EPO in preclinical models of acute and chronic neurodegenerative disorders, including Alzheimer's disease (AD). AD is one of the most prevalent neurodegenerative illnesses in the elderly, characterized by the accumulation of extracellular amyloid-ß (Aß) plaques and intracellular neurofibrillary tangles (NFTs), which serve as the disease's two hallmarks. Unfortunately, AD lacks a successful treatment strategy due to its multifaceted and complex pathology. Various clinical studies, both in vitro and in vivo, have been conducted to identify the various mechanisms by which erythropoietin exerts its neuroprotective effects. The results of clinical trials in patients with AD are also promising. Herein, it is summarized and reviews all such studies demonstrating erythropoietin's potential therapeutic benefits as a pleiotropic neuroprotective agent in the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Dapinder Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | | | - Syed Shams Ul Hassan
- School of Medicine and Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Mohammad Mehedi Hasan
- Department of Biochemistry and Molecular Biology, Faculty of Life Science, Mawlana Bhashani Science and Technology University, Tangail, Bangladesh
| | - Saurabh Bhatia
- Natural & Medical Sciences Research Centre, University of Nizwa, Nizwa, Oman
- Amity Institute of Pharmacy, Amity University, Noida, Haryana, India
| | - Ahmed Al-Harassi
- Natural & Medical Sciences Research Centre, University of Nizwa, Nizwa, Oman
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
14
|
Elliot-Portal E, Arias-Reyes C, Laouafa S, Tam R, Kinkead R, Soliz J. Cerebral Erythropoietin Prevents Sex-Dependent Disruption of Respiratory Control Induced by Early Life Stress. Front Physiol 2021; 12:701344. [PMID: 34987412 PMCID: PMC8720854 DOI: 10.3389/fphys.2021.701344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 11/22/2021] [Indexed: 11/29/2022] Open
Abstract
Injuries that occur early in life are often at the root of adult illness. Neonatal maternal separation (NMS) is a form of early life stress that has persistent and sex-specific effects on the development of neural networks, including those that regulate breathing. The release of stress hormones during a critical period of development contributes to the deleterious consequences of NMS, but the role of increased corticosterone (CORT) in NMS-induced respiratory disturbance is unknown. Because erythropoietin (EPO) is a potent neuroprotectant that prevents conditions associated with hyperactivation of the stress neuroaxis in a sex-specific manner, we hypothesized that EPO reduces the sex-specific alteration of respiratory regulation induced by NMS in adult mice. Animals were either raised under standard conditions (controls) or exposed to NMS 3 h/day from postnatal days 3–12. We tested the efficacy of EPO in preventing the effects of NMS by comparing wild-type mice with transgenic mice that overexpress EPO only in the brain (Tg21). In 7-days-old pups, NMS augmented CORT levels ~2.5-fold by comparison with controls but only in males; this response was reduced in Tg21 mice. Respiratory function was assessed using whole-body plethysmography. Apneas were detected during sleep; the responsiveness to stimuli was measured by exposing mice to hypoxia (10% O2; 15 min) and hypercapnia (5% CO2; 10 min). In wild-type, NMS increased the number of apneas and the hypercapnic ventilatory response (HcVR) only in males; with no effect on Tg21. In wild-type males, the incidence of apneas was positively correlated with HcVR and inversely related to the tachypneic response to hypoxia. We conclude that neural EPO reduces early life stress-induced respiratory disturbances observed in males.
Collapse
Affiliation(s)
- Elizabeth Elliot-Portal
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, QC, Canada
| | - Christian Arias-Reyes
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, QC, Canada
| | - Sofien Laouafa
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, QC, Canada
| | - Rose Tam
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, QC, Canada
| | - Richard Kinkead
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, QC, Canada
| | - Jorge Soliz
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, QC, Canada
- High Altitude Pulmonary and Pathology Institute (HAPPI–IPPA), La Paz, Bolivia
- *Correspondence: Jorge Soliz,
| |
Collapse
|
15
|
Watts D, Bechmann N, Meneses A, Poutakidou IK, Kaden D, Conrad C, Krüger A, Stein J, El-Armouche A, Chavakis T, Eisenhofer G, Peitzsch M, Wielockx B. HIF2α regulates the synthesis and release of epinephrine in the adrenal medulla. J Mol Med (Berl) 2021; 99:1655-1666. [PMID: 34480587 PMCID: PMC8542008 DOI: 10.1007/s00109-021-02121-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 02/06/2023]
Abstract
The adrenal gland and its hormones regulate numerous fundamental biological processes; however, the impact of hypoxia signaling on adrenal function remains poorly understood. Here, we reveal that deficiency of HIF (hypoxia inducible factors) prolyl hydroxylase domain protein-2 (PHD2) in the adrenal medulla of mice results in HIF2α-mediated reduction in phenylethanolamine N-methyltransferase (PNMT) expression, and consequent reduction in epinephrine synthesis. Simultaneous loss of PHD2 in renal erythropoietin (EPO)-producing cells (REPCs) stimulated HIF2α-driven EPO overproduction, excessive RBC formation (erythrocytosis), and systemic hypoglycemia, which is necessary and sufficient to enhance exocytosis of epinephrine from the adrenal medulla. Based on these results, we propose that the PHD2-HIF2α axis in the adrenal medulla regulates the synthesis of epinephrine, whereas in REPCs, it indirectly induces the release of this hormone. Our findings are also highly relevant to the testing of small molecule PHD inhibitors in phase III clinical trials for patients with renal anemia. KEY MESSAGES: HIF2α and not HIF1α modulates PNMT during epinephrine synthesis in chromaffin cells. The PHD2-HIF2α-EPO axis induces erythrocytosis and hypoglycemia. Reduced systemic glucose facilitates exocytosis of epinephrine from adrenal gland.
Collapse
Affiliation(s)
- Deepika Watts
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
| | - Nicole Bechmann
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany.,Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke, 14558, Nuthetal, Germany.,German Center for Diabetes Research (DZD), 85764, München-Neuherberg, Germany
| | - Ana Meneses
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
| | - Ioanna K Poutakidou
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
| | - Denise Kaden
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
| | - Catleen Conrad
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
| | - Anja Krüger
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
| | - Johanna Stein
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
| | - Ali El-Armouche
- Department of Pharmacology and Toxicology, Medical Faculty, Technische Universität Dresden, 01307, Dresden, Germany
| | - Triantafyllos Chavakis
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
| | - Graeme Eisenhofer
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany.,Department of Medicine III, Medical Faculty, Technische Universität Dresden, 01307, Dresden, Germany
| | - Mirko Peitzsch
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
| | - Ben Wielockx
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany.
| |
Collapse
|
16
|
Jacobs RA, Aboouf MA, Koester-Hegmann C, Muttathukunnel P, Laouafa S, Arias-Reyes C, Thiersch M, Soliz J, Gassmann M, Schneider Gasser EM. Erythropoietin promotes hippocampal mitochondrial function and enhances cognition in mice. Commun Biol 2021; 4:938. [PMID: 34354241 PMCID: PMC8342552 DOI: 10.1038/s42003-021-02465-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 07/19/2021] [Indexed: 11/22/2022] Open
Abstract
Erythropoietin (EPO) improves neuronal mitochondrial function and cognition in adults after brain injury and in those afflicted by psychiatric disorders. However, the influence of EPO on mitochondria and cognition during development remains unexplored. We previously observed that EPO stimulates hippocampal-specific neuronal maturation and synaptogenesis early in postnatal development in mice. Here we show that EPO promotes mitochondrial respiration in developing postnatal hippocampus by increasing mitochondrial content and enhancing cellular respiratory potential. Ultrastructurally, mitochondria profiles and total vesicle content were greater in presynaptic axon terminals, suggesting that EPO enhances oxidative metabolism and synaptic transmission capabilities. Behavioural tests of hippocampus-dependent memory at early adulthood, showed that EPO improves spatial and short-term memory. Collectively, we identify a role for EPO in the murine postnatal hippocampus by promoting mitochondrial function throughout early postnatal development, which corresponds to enhanced cognition by early adulthood. Robert Jacobs, Mostafa Aboouf, et al. examined the effect of erythropoietin (EPO) in hippocampal mitochondrial function and memory in two mouse models: one overexpressing EPO in the brain, and juvenile mice treated during three days with a high dose of intraperitoneal EPO. Their results suggest that erythropoietin in the neonatal brain may impact spatial memory by increasing mitochondrial content.
Collapse
Affiliation(s)
- Robert A Jacobs
- Institute of Veterinary Physiology, Vetsuisse-Faculty, University of Zurich, Zurich, Switzerland.,Department of Human Physiology & Nutrition, University of Colorado, Colorado Springs, CO, USA
| | - Mostafa A Aboouf
- Institute of Veterinary Physiology, Vetsuisse-Faculty, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIPH), University of Zurich, Zurich, Switzerland.,Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Christina Koester-Hegmann
- Institute of Veterinary Physiology, Vetsuisse-Faculty, University of Zurich, Zurich, Switzerland.,Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Paola Muttathukunnel
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.,Center for Neuroscience Zurich (ZNZ), Zurich, Switzerland
| | - Sofien Laouafa
- Faculty of Medicine, Centre Hospitalier Universitaire de Québec (CHUQ), Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, QC, Canada
| | - Christian Arias-Reyes
- Faculty of Medicine, Centre Hospitalier Universitaire de Québec (CHUQ), Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, QC, Canada
| | - Markus Thiersch
- Institute of Veterinary Physiology, Vetsuisse-Faculty, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIPH), University of Zurich, Zurich, Switzerland
| | - Jorge Soliz
- Faculty of Medicine, Centre Hospitalier Universitaire de Québec (CHUQ), Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, QC, Canada
| | - Max Gassmann
- Institute of Veterinary Physiology, Vetsuisse-Faculty, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIPH), University of Zurich, Zurich, Switzerland
| | - Edith M Schneider Gasser
- Institute of Veterinary Physiology, Vetsuisse-Faculty, University of Zurich, Zurich, Switzerland. .,Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland. .,Center for Neuroscience Zurich (ZNZ), Zurich, Switzerland.
| |
Collapse
|
17
|
Erythropoietin Stimulates GABAergic Maturation in the Mouse Hippocampus. eNeuro 2021; 8:ENEURO.0006-21.2021. [PMID: 33495244 PMCID: PMC7890522 DOI: 10.1523/eneuro.0006-21.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 01/09/2021] [Indexed: 12/15/2022] Open
Abstract
Several neurodevelopmental disabilities are strongly associated with alterations in GABAergic transmission, and therapies to stimulate its normal development are lacking. Erythropoietin (EPO) is clinically used in neonatology to mitigate acute brain injury, and to stimulate neuronal maturation. Yet it remains unclear whether EPO can stimulate maturation of the GABAergic system. Here, with the use of a transgenic mouse line that constitutively overexpresses neuronal EPO (Tg21), we show that EPO stimulates postnatal GABAergic maturation in the hippocampus. We show an increase in hippocampal GABA-immunoreactive neurons, and postnatal elevation of interneurons expressing parvalbumin (PV), somatostatin (SST), and neuropeptide Y (NPY). Analysis of perineuronal net (PNN) formation and innervation of glutamatergic terminals onto PV+ cells, shows to be enhanced early in postnatal development. Additionally, an increase in GABAAergic synapse density and IPSCs in CA1 pyramidal cells from Tg21 mice is observed. Detection of EPO receptor (EPOR) mRNA was observed to be restricted to glutamatergic pyramidal cells and increased in Tg21 mice at postnatal day (P)7, along with reduced apoptosis. Our findings show that EPO can stimulate postnatal GABAergic maturation in the hippocampus, by increasing neuronal survival, modulating critical plasticity periods, and increasing synaptic transmission. Our data supports EPO’s clinical use to balance GABAergic dysfunction.
Collapse
|
18
|
Ratsma DMA, Zillikens MC, van der Eerden BCJ. Upstream Regulators of Fibroblast Growth Factor 23. Front Endocrinol (Lausanne) 2021; 12:588096. [PMID: 33716961 PMCID: PMC7952762 DOI: 10.3389/fendo.2021.588096] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 01/11/2021] [Indexed: 12/11/2022] Open
Abstract
Fibroblast growth factor 23 (FGF23) has been described as an important regulator of mineral homeostasis, but has lately also been linked to iron deficiency, inflammation, and erythropoiesis. FGF23 is essential for the maintenance of phosphate homeostasis in the body and activating mutations in the gene itself or inactivating mutations in its upstream regulators can result in severe chronic hypophosphatemia, where an unbalanced mineral homeostasis often leads to rickets in children and osteomalacia in adults. FGF23 can be regulated by changes in transcriptional activity or by changes at the post-translational level. The balance between O-glycosylation and phosphorylation is an important determinant of how much active intact or inactive cleaved FGF23 will be released in the circulation. In the past years, it has become evident that iron deficiency and inflammation regulate FGF23 in a way that is not associated with its classical role in mineral metabolism. These conditions will not only result in an upregulation of FGF23 transcription, but also in increased cleavage, leaving the levels of active intact FGF23 unchanged. The exact mechanisms behind and function of this process are still unclear. However, a deeper understanding of FGF23 regulation in both the classical and non-classical way is important to develop better treatment options for diseases associated with disturbed FGF23 biology. In this review, we describe how the currently known upstream regulators of FGF23 change FGF23 transcription and affect its post-translational modifications at the molecular level.
Collapse
|
19
|
Riou R, Ladli M, Gerbal-Chaloin S, Bossard P, Gougelet A, Godard C, Loesch R, Lagoutte I, Lager F, Calderaro J, Dos Santos A, Wang Z, Verdier F, Colnot S. ARID1A loss in adult hepatocytes activates β-catenin-mediated erythropoietin transcription. eLife 2020; 9:e53550. [PMID: 33084574 PMCID: PMC7641585 DOI: 10.7554/elife.53550] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 10/20/2020] [Indexed: 12/13/2022] Open
Abstract
Erythropoietin (EPO) is a key regulator of erythropoiesis. The embryonic liver is the main site of erythropoietin synthesis, after which the kidney takes over. The adult liver retains the ability to express EPO, and we discovered here new players of this transcription, distinct from the classical hypoxia-inducible factor pathway. In mice, genetically invalidated in hepatocytes for the chromatin remodeler Arid1a, and for Apc, the major silencer of Wnt pathway, chromatin was more accessible and histone marks turned into active ones at the Epo downstream enhancer. Activating β-catenin signaling increased binding of Tcf4/β-catenin complex and upregulated its enhancer function. The loss of Arid1a together with β-catenin signaling, resulted in cell-autonomous EPO transcription in mouse and human hepatocytes. In mice with Apc-Arid1a gene invalidations in single hepatocytes, Epo de novo synthesis led to its secretion, to splenic erythropoiesis and to dramatic erythrocytosis. Thus, we identified new hepatic EPO regulation mechanism stimulating erythropoiesis.
Collapse
Affiliation(s)
- Rozenn Riou
- INSERM, Sorbonne Université, Université de Paris, Centre de Recherche des Cordeliers (CRC)ParisFrance
- Equipe labellisée Ligue Nationale Contre le CancerParisFrance
- INSERM, CNRS, Institut COCHINParisFrance
| | | | - Sabine Gerbal-Chaloin
- INSERM U1183, Université Montpellier, Institute for Regenerative Medicine & Biotherapy (IRMB)MontpellierFrance
| | - Pascale Bossard
- Equipe labellisée Ligue Nationale Contre le CancerParisFrance
- INSERM, CNRS, Institut COCHINParisFrance
| | - Angélique Gougelet
- INSERM, Sorbonne Université, Université de Paris, Centre de Recherche des Cordeliers (CRC)ParisFrance
- Equipe labellisée Ligue Nationale Contre le CancerParisFrance
- INSERM, CNRS, Institut COCHINParisFrance
| | - Cécile Godard
- INSERM, Sorbonne Université, Université de Paris, Centre de Recherche des Cordeliers (CRC)ParisFrance
- Equipe labellisée Ligue Nationale Contre le CancerParisFrance
- INSERM, CNRS, Institut COCHINParisFrance
| | - Robin Loesch
- INSERM, Sorbonne Université, Université de Paris, Centre de Recherche des Cordeliers (CRC)ParisFrance
- Equipe labellisée Ligue Nationale Contre le CancerParisFrance
- INSERM, CNRS, Institut COCHINParisFrance
| | - Isabelle Lagoutte
- INSERM, CNRS, Institut COCHINParisFrance
- Plateforme d’Imageries du Vivant de l’Université de ParisParisFrance
| | - Franck Lager
- INSERM, CNRS, Institut COCHINParisFrance
- Plateforme d’Imageries du Vivant de l’Université de ParisParisFrance
| | - Julien Calderaro
- INSERM, Université Paris-Est UPECCréteilFrance
- Department of Pathology, Henri Mondor HospitalCréteilFrance
| | | | - Zhong Wang
- Department of Cardiac Surgery Cardiovascular Research Center, University of MichiganAnn ArborUnited States
| | | | - Sabine Colnot
- INSERM, Sorbonne Université, Université de Paris, Centre de Recherche des Cordeliers (CRC)ParisFrance
- Equipe labellisée Ligue Nationale Contre le CancerParisFrance
- INSERM, CNRS, Institut COCHINParisFrance
| |
Collapse
|
20
|
Soliz J, Schneider-Gasser EM, Arias-Reyes C, Aliaga-Raduan F, Poma-Machicao L, Zubieta-Calleja G, Furuya WI, Trevizan-Baú P, Dhingra RR, Dutschmann M. Coping with hypoxemia: Could erythropoietin (EPO) be an adjuvant treatment of COVID-19? Respir Physiol Neurobiol 2020; 279:103476. [PMID: 32522574 PMCID: PMC7275159 DOI: 10.1016/j.resp.2020.103476] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 05/29/2020] [Accepted: 05/30/2020] [Indexed: 12/13/2022]
Abstract
A very recent epidemiological study provides preliminary evidence that living in habitats located at 2500 m above sea level (masl) might protect from the development of severe respiratory symptoms following infection with the novel SARS-CoV-2 virus. This epidemiological finding raises the question of whether physiological mechanisms underlying the acclimatization to high altitude identifies therapeutic targets for the effective treatment of severe acute respiratory syndrome pivotal to the reduction of global mortality during the COVID-19 pandemic. This article compares the symptoms of acute mountain sickness (AMS) with those of SARS-CoV-2 infection and explores overlapping patho-physiological mechanisms of the respiratory system including impaired oxygen transport, pulmonary gas exchange and brainstem circuits controlling respiration. In this context, we also discuss the potential impact of SARS-CoV-2 infection on oxygen sensing in the carotid body. Finally, since erythropoietin (EPO) is an effective prophylactic treatment for AMS, this article reviews the potential benefits of implementing FDA-approved erythropoietin-based (EPO) drug therapies to counteract a variety of acute respiratory and non-respiratory (e.g. excessive inflammation of vascular beds) symptoms of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Jorge Soliz
- Institute Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ), Faculty of Medicine, Université Laval, Québec, QC, Canada; High Altitude Pulmonary and Pathology Institute IPPA, La Paz, Bolivia.
| | - Edith M Schneider-Gasser
- Institute of Veterinary Physiology, Vetsuisse-Faculty University of Zurich, Winterthurerstrasse 260, Switzerland
| | - Christian Arias-Reyes
- Institute Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ), Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Fernanda Aliaga-Raduan
- Institute Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ), Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Liliana Poma-Machicao
- Institute Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ), Faculty of Medicine, Université Laval, Québec, QC, Canada
| | | | - Werner I Furuya
- Florey Institute of Neuroscience and Mental Health, Melbourne Victoria, Australia; The Florey Department of Neuroscience, University of Melbourne, Melbourne Victoria, Australia
| | - Pedro Trevizan-Baú
- Florey Institute of Neuroscience and Mental Health, Melbourne Victoria, Australia; The Florey Department of Neuroscience, University of Melbourne, Melbourne Victoria, Australia
| | - Rishi R Dhingra
- Florey Institute of Neuroscience and Mental Health, Melbourne Victoria, Australia
| | - Mathias Dutschmann
- Florey Institute of Neuroscience and Mental Health, Melbourne Victoria, Australia; The Florey Department of Neuroscience, University of Melbourne, Melbourne Victoria, Australia
| |
Collapse
|
21
|
Hanudel MR, Eisenga MF, Rappaport M, Chua K, Qiao B, Jung G, Gabayan V, Gales B, Ramos G, de Jong MA, van Zanden JJ, de Borst MH, Bakker SJL, Nemeth E, Salusky IB, Gaillard CAJM, Ganz T. Effects of erythropoietin on fibroblast growth factor 23 in mice and humans. Nephrol Dial Transplant 2020; 34:2057-2065. [PMID: 30007314 DOI: 10.1093/ndt/gfy189] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 05/22/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Erythropoietin (EPO) has been reported as a novel determinant of fibroblast growth factor 23 (FGF23) production; however, it is unknown whether FGF23 is stimulated by chronic exposure to EPO or by EPO administration in nonpolycystic chronic kidney disease (CKD) models. METHODS We analyzed the effects of chronic EPO on FGF23 in murine models with chronically high EPO levels and normal kidney function. We studied the effects of exogenous EPO on FGF23 in wild-type mice, with and without CKD, injected with EPO. Also, in four independent human CKD cohorts, we evaluated associations between FGF23 and serum EPO levels or exogenous EPO dose. RESULTS Mice with high endogenous EPO have elevated circulating total FGF23, increased disproportionately to intact FGF23, suggesting coupling of increased FGF23 production with increased proteolytic cleavage. Similarly, in wild-type mice with and without CKD, a single exogenous EPO dose acutely increases circulating total FGF23 out of proportion to intact FGF23. In these murine models, the bone marrow is shown to be a novel source of EPO-stimulated FGF23 production. In humans, serum EPO levels and recombinant human EPO dose are positively and independently associated with total FGF23 levels across the spectrum of CKD and after kidney transplantation. In our largest cohort of 680 renal transplant recipients, serum EPO levels are associated with total FGF23, but not intact FGF23, consistent with the effects of EPO on FGF23 production and metabolism observed in our murine models. CONCLUSION EPO affects FGF23 production and metabolism, which may have important implications for CKD patients.
Collapse
Affiliation(s)
- Mark R Hanudel
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Michele F Eisenga
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Maxime Rappaport
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Kristine Chua
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Bo Qiao
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Grace Jung
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Victoria Gabayan
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Barbara Gales
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Georgina Ramos
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Maarten A de Jong
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jelmer J van Zanden
- Certe, Department of Clinical Chemistry, Martini Hospital, Groningen, The Netherlands
| | - Martin H de Borst
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Stephan J L Bakker
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Elizabeta Nemeth
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Isidro B Salusky
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Carlo A J M Gaillard
- Department of Internal Medicine and Dermatology, University of Utrecht, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Tomas Ganz
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
22
|
Dey S, Cui Z, Gavrilova O, Zhang X, Gassmann M, Noguchi CT. Sex-specific brain erythropoietin regulation of mouse metabolism and hypothalamic inflammation. JCI Insight 2020; 5:134061. [PMID: 32078583 DOI: 10.1172/jci.insight.134061] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 02/12/2020] [Indexed: 12/17/2022] Open
Abstract
The blood hormone erythropoietin (EPO), upon binding to its receptor (EpoR), modulates high-fat diet-induced (HFD-induced) obesity in mice, improves glucose tolerance, and prevents white adipose tissue inflammation. Transgenic mice with constitutive overexpression of human EPO solely in the brain (Tg21) were used to assess the neuroendocrine EPO effect without increasing the hematocrit. Male Tg21 mice resisted HFD-induced weight gain; showed lower serum adrenocorticotropic hormone, corticosterone, and C-reactive protein levels; and prevented myeloid cell recruitment to the hypothalamus compared with WT male mice. HFD-induced hypothalamic inflammation (HI) and microglial activation were higher in male mice, and Tg21 male mice exhibited a lower increase in HI than WT male mice. Physiological EPO function in the brain also showed sexual dimorphism in regulating HFD response. Female estrogen production blocked reduced weight gain and HI. Targeted deletion of EpoR gene expression in neuronal cells worsened HFD-induced glucose intolerance in both male and female mice but increased weight gain and HI in the hypothalamus in male mice only. Both male and female Tg21 mice kept on normal chow and HFD showed significantly improved glycemic control. Our data indicate that cerebral EPO regulates weight gain and HI in a sex-dependent response, distinct from EPO regulation of glycemic control, and independent of erythropoietic EPO response.
Collapse
Affiliation(s)
| | | | | | - Xiaojie Zhang
- Laboratory of Animal Sciences Section, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, Maryland, USA
| | - Max Gassmann
- Institute of Veterinary Physiology and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.,Universidad Peruana Cayetano Heredia, Lima, Peru
| | | |
Collapse
|
23
|
Suresh S, Rajvanshi PK, Noguchi CT. The Many Facets of Erythropoietin Physiologic and Metabolic Response. Front Physiol 2020; 10:1534. [PMID: 32038269 PMCID: PMC6984352 DOI: 10.3389/fphys.2019.01534] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 12/05/2019] [Indexed: 12/30/2022] Open
Abstract
In mammals, erythropoietin (EPO), produced in the kidney, is essential for bone marrow erythropoiesis, and hypoxia induction of EPO production provides for the important erythropoietic response to ischemic stress, such as during blood loss and at high altitude. Erythropoietin acts by binding to its cell surface receptor which is expressed at the highest level on erythroid progenitor cells to promote cell survival, proliferation, and differentiation in production of mature red blood cells. In addition to bone marrow erythropoiesis, EPO causes multi-tissue responses associated with erythropoietin receptor (EPOR) expression in non-erythroid cells such neural cells, endothelial cells, and skeletal muscle myoblasts. Animal and cell models of ischemic stress have been useful in elucidating the potential benefit of EPO affecting maintenance and repair of several non-hematopoietic organs including brain, heart and skeletal muscle. Metabolic and glucose homeostasis are affected by endogenous EPO and erythropoietin administration affect, in part via EPOR expression in white adipose tissue. In diet-induced obese mice, EPO is protective for white adipose tissue inflammation and gives rise to a gender specific response in weight control associated with white fat mass accumulation. Erythropoietin regulation of fat mass is masked in female mice due to estrogen production. EPOR is also expressed in bone marrow stromal cells (BMSC) and EPO administration in mice results in reduced bone independent of the increase in hematocrit. Concomitant reduction in bone marrow adipocytes and bone morphogenic protein suggests that high EPO inhibits adipogenesis and osteogenesis. These multi-tissue responses underscore the pleiotropic potential of the EPO response and may contribute to various physiological manifestations accompanying anemia or ischemic response and pharmacological uses of EPO.
Collapse
Affiliation(s)
- Sukanya Suresh
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Praveen Kumar Rajvanshi
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Constance T Noguchi
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
24
|
Rao TN, Hansen N, Hilfiker J, Rai S, Majewska JM, Leković D, Gezer D, Andina N, Galli S, Cassel T, Geier F, Delezie J, Nienhold R, Hao-Shen H, Beisel C, Di Palma S, Dimeloe S, Trebicka J, Wolf D, Gassmann M, Fan TWM, Lane AN, Handschin C, Dirnhofer S, Kröger N, Hess C, Radimerski T, Koschmieder S, Čokić VP, Skoda RC. JAK2-mutant hematopoietic cells display metabolic alterations that can be targeted to treat myeloproliferative neoplasms. Blood 2019; 134:1832-1846. [PMID: 31511238 PMCID: PMC6872961 DOI: 10.1182/blood.2019000162] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 08/17/2019] [Indexed: 12/20/2022] Open
Abstract
Increased energy requirement and metabolic reprogramming are hallmarks of cancer cells. We show that metabolic alterations in hematopoietic cells are fundamental to the pathogenesis of mutant JAK2-driven myeloproliferative neoplasms (MPNs). We found that expression of mutant JAK2 augmented and subverted metabolic activity of MPN cells, resulting in systemic metabolic changes in vivo, including hypoglycemia, adipose tissue atrophy, and early mortality. Hypoglycemia in MPN mouse models correlated with hyperactive erythropoiesis and was due to a combination of elevated glycolysis and increased oxidative phosphorylation. Modulating nutrient supply through high-fat diet improved survival, whereas high-glucose diet augmented the MPN phenotype. Transcriptomic and metabolomic analyses identified numerous metabolic nodes in JAK2-mutant hematopoietic stem and progenitor cells that were altered in comparison with wild-type controls. We studied the consequences of elevated levels of Pfkfb3, a key regulatory enzyme of glycolysis, and found that pharmacological inhibition of Pfkfb3 with the small molecule 3PO reversed hypoglycemia and reduced hematopoietic manifestations of MPNs. These effects were additive with the JAK1/2 inhibitor ruxolitinib in vivo and in vitro. Inhibition of glycolysis by 3PO altered the redox homeostasis, leading to accumulation of reactive oxygen species and augmented apoptosis rate. Our findings reveal the contribution of metabolic alterations to the pathogenesis of MPNs and suggest that metabolic dependencies of mutant cells represent vulnerabilities that can be targeted for treating MPNs.
Collapse
Affiliation(s)
- Tata Nageswara Rao
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Nils Hansen
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Julian Hilfiker
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Shivam Rai
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Julia-Magdalena Majewska
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Danijela Leković
- Clinic of Hematology, Clinical Center of Serbia, Belgrade, Serbia
| | - Deniz Gezer
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Nicola Andina
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Serena Galli
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Teresa Cassel
- Center for Environmental and Systems Biochemistry, Department of Toxicology and Cancer Biology and Markey Cancer Center, University of Kentucky, Lexington, KY
| | - Florian Geier
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | | | - Ronny Nienhold
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Hui Hao-Shen
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Christian Beisel
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Serena Di Palma
- Functional Genomics Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Sarah Dimeloe
- Immunobiology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Jonel Trebicka
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
- European Foundation for the Study of Chronic Liver Failure, Barcelona, Spain
- Department of Gastroenterology, Odense Hospital, University of Southern Denmark, Odense, Denmark
- Institute for Bioengineering of Catalonia, Barcelona, Spain
| | - Dominik Wolf
- Internal Medicine V, Department of Hematology and Oncology, Medical University Innsbruck, Innsbruck, Austria
- Medical Clinic III for Oncology, Hematology, Immunoncology and Rheumatology, University Hospital Bonn, Bonn, Germany
| | - Max Gassmann
- Institute of Veterinary Physiology, Vetsuisse Faculty and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Teresa W-M Fan
- Center for Environmental and Systems Biochemistry, Department of Toxicology and Cancer Biology and Markey Cancer Center, University of Kentucky, Lexington, KY
| | - Andrew N Lane
- Center for Environmental and Systems Biochemistry, Department of Toxicology and Cancer Biology and Markey Cancer Center, University of Kentucky, Lexington, KY
| | | | - Stefan Dirnhofer
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Hospital Eppendorf, Hamburg, Germany
| | - Christoph Hess
- Immunobiology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Thomas Radimerski
- Disease Area Oncology, Novartis Institutes for Biomedical Research, Basel, Switzerland; and
| | - Steffen Koschmieder
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Vladan P Čokić
- Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Radek C Skoda
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| |
Collapse
|
25
|
Daryadel A, Natale L, Seebeck P, Bettoni C, Schnitzbauer U, Gassmann M, Wagner CA. Elevated FGF23 and disordered renal mineral handling with reduced bone mineralization in chronically erythropoietin over-expressing transgenic mice. Sci Rep 2019; 9:14989. [PMID: 31628396 PMCID: PMC6802194 DOI: 10.1038/s41598-019-51577-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 09/09/2019] [Indexed: 12/22/2022] Open
Abstract
Fibroblast Growth Factor 23 (FGF23) is a phosphaturic factor causing increased renal phosphate excretion as well as suppression of 1,25 (OH)2-vitamin D3. Highly elevated FGF23 can promote development of rickets and osteomalacia. We and others previously reported that acute application of erythropoietin (EPO) stimulates FGF23 production. Considering that EPO is clinically used as chronic treatment against anemia, we used here the Tg6 mouse model that constitutively overexpresses human EPO in an oxygen-independent manner, to examine the consequences of long-term EPO therapy on mineral and bone metabolism. Six to eight weeks old female Tg6 mice showed elevated intact and C-terminal fragment of FGF23 but normal plasma levels of PTH, calcitriol, calcium and phosphate. Renal function showed moderate alterations with higher urea and creatinine clearance and mild albuminuria. Renal phosphate excretion was normal whereas mild hypercalciuria was found. Renal expression of the key proteins TRPV5 and calbindin D28k involved in active calcium reabsorption was reduced in Tg6 mice. Plasma levels of the bone turnover marker osteocalcin were comparable between groups. However, urinary excretion of deoxypyridinoline (DPD) was lower in Tg6 mice. MicroCT analysis showed reduced total, cortical, and trabecular bone mineral density in femora from Tg6 mice. Our data reveal that chronic elevation of EPO is associated with high FGF23 levels and disturbed mineral homeostasis resulting in reduced bone mineral density. These observations imply the need to study the impact of therapeutically applied EPO on bone mineralization in patients, especially those suffering from chronic kidney disease.
Collapse
Affiliation(s)
- Arezoo Daryadel
- Institute of Physiology, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland.,National Centre for Competence in Research NCCR "Kidney.CH", Zurich, Switzerland
| | - Luciano Natale
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Petra Seebeck
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland.,Zurich Integrative Rodent Physiology (ZIRP), University of Zurich, Zurich, Switzerland
| | - Carla Bettoni
- Institute of Physiology, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland.,National Centre for Competence in Research NCCR "Kidney.CH", Zurich, Switzerland
| | - Udo Schnitzbauer
- Institute of Physiology, University of Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland.,National Centre for Competence in Research NCCR "Kidney.CH", Zurich, Switzerland
| | - Max Gassmann
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland.,Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland.,Universidad Peruana Cayetano Heredia (UPCH), Lima, Peru
| | - Carsten A Wagner
- Institute of Physiology, University of Zurich, Zurich, Switzerland. .,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland. .,National Centre for Competence in Research NCCR "Kidney.CH", Zurich, Switzerland.
| |
Collapse
|
26
|
Suresh S, de Castro LF, Dey S, Robey PG, Noguchi CT. Erythropoietin modulates bone marrow stromal cell differentiation. Bone Res 2019; 7:21. [PMID: 31666996 PMCID: PMC6804931 DOI: 10.1038/s41413-019-0060-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 04/01/2019] [Accepted: 05/17/2019] [Indexed: 01/08/2023] Open
Abstract
Erythropoietin is essential for bone marrow erythropoiesis and erythropoietin receptor on non-erythroid cells including bone marrow stromal cells suggests systemic effects of erythropoietin. Tg6 mice with chronic erythropoietin overexpression have a high hematocrit, reduced trabecular and cortical bone and bone marrow adipocytes, and decreased bone morphogenic protein 2 driven ectopic bone and adipocyte formation. Erythropoietin treatment (1 200 IU·kg–1) for 10 days similarly exhibit increased hematocrit, reduced bone and bone marrow adipocytes without increased osteoclasts, and reduced bone morphogenic protein signaling in the bone marrow. Interestingly, endogenous erythropoietin is required for normal differentiation of bone marrow stromal cells to osteoblasts and bone marrow adipocytes. ΔEpoRE mice with erythroid restricted erythropoietin receptor exhibit reduced trabecular bone, increased bone marrow adipocytes, and decreased bone morphogenic protein 2 ectopic bone formation. Erythropoietin treated ΔEpoRE mice achieved hematocrit similar to wild-type mice without reduced bone, suggesting that bone reduction with erythropoietin treatment is associated with non-erythropoietic erythropoietin response. Bone marrow stromal cells from wild-type, Tg6, and ΔEpoRE-mice were transplanted into immunodeficient mice to assess development into a bone/marrow organ. Like endogenous bone formation, Tg6 bone marrow cells exhibited reduced differentiation to bone and adipocytes indicating that high erythropoietin inhibits osteogenesis and adipogenesis, while ΔEpoRE bone marrow cells formed ectopic bones with reduced trabecular regions and increased adipocytes, indicating that loss of erythropoietin signaling favors adipogenesis at the expense of osteogenesis. In summary, endogenous erythropoietin signaling regulates bone marrow stromal cell fate and aberrant erythropoietin levels result in their impaired differentiation.
Collapse
Affiliation(s)
- Sukanya Suresh
- 1Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| | - Luis Fernandez de Castro
- 2Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892 USA
| | - Soumyadeep Dey
- 1Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| | - Pamela G Robey
- 2Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892 USA
| | - Constance Tom Noguchi
- 1Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| |
Collapse
|
27
|
Moringa oleifera Seeds Improve Aging-Related Endothelial Dysfunction in Wistar Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:2567198. [PMID: 31214278 PMCID: PMC6535829 DOI: 10.1155/2019/2567198] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 02/14/2019] [Indexed: 12/16/2022]
Abstract
Vascular aging is characterized by functional and structural changes of the vessel wall, including endothelial dysfunction, with decreased endothelial NO· bioavailability and elevated vasoconstrictor and inflammatory mediator production, vascular rigidity, and tone impairment. Moringa oleifera (MOI) is a little tree, and different parts of which are used in traditional medicine in tropical Africa, America, and Asia for therapeutic applications in several disorders including cardiovascular disease. The present study is aimed at assessing the effect of MOI on aging-associated alteration of the endothelial function in Wistar rats. Middle-aged Wistar rats (46-week-old males) have been fed with food containing or not 750 mg/kg/day of MOI seed powder for 4 weeks. A group of young Wistar rats (16-week-old) was used as control. Measurement of isometric contraction, western blot analysis, and immunostaining has then been performed in the aortas and mesenteric arteries to assess the endothelium function. MOI treatment improved carbachol-induced relaxation in both aortas and mesenteric arteries of middle-aged rats. In the aortas, this was associated with an increased Akt signalling and endothelial NO synthase activation and a downregulation of arginase-1. In the mesenteric arteries, the improvement of the endothelial-dependent relaxation was related to an EDHF-dependent mechanism. These results suggest a vascular protective effect of MOI seeds against the vascular dysfunction that develops during aging through different mechanisms in conductance and resistance arteries.
Collapse
|
28
|
Kittur FS, Lin Y, Arthur E, Hung CY, Li PA, Sane DC, Xie J. Recombinant asialoerythropoetin protects HL-1 cardiomyocytes from injury via suppression of Mst1 activation. Biochem Biophys Rep 2019; 17:157-168. [PMID: 30671548 PMCID: PMC6327940 DOI: 10.1016/j.bbrep.2019.01.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 01/02/2019] [Accepted: 01/04/2019] [Indexed: 12/04/2022] Open
Abstract
Background Recombinant human erythropoietin (rhuEPO) and asialoerythropoietin (asialo-rhuEPO) are cardioprotective. However, the protective effects of rhuEPO could not be translated into clinical practice because of its hematopoiesis-associated side effects while non-erythropoietic asialo-rhuEPO is unavailable in large quantities for clinical studies. This study was designed to investigate the cardiomyocyte protective potential of plant-produced asialo-rhuEPO (asialo-rhuEPOP) against staurosporine (STS)-induced injury in HL-1 murine cardiomyocytes and identify cellular pathway(s) responsible for its cardioprotection. Methods HL-1 cardiomyocytes were simultaneously treated with STS and asialo-rhuEPOP. Cellular injury, apoptosis, and cell viabilities were measured by LDH assay, Hoechst staining and trypan blue exclusion method, respectively while western blotting was used to study its effects on apoptosis and autophagy hallmarks. Results Our results showed that 20 IU/ml asialo-rhuEPOP provided 39% protection to cardiomyocytes compared to STS-treated cells, which is 2-fold better than that of mammalian cell-produce rhuEPO (rhuEPOM). Asialo-rhuEPOP was found to suppress activation of proapoptotic kinase Mst1 (mammalian Sterile-20-like kinase 1) and FOXO3, leading to inhibition of apoptotic pathway and restoration of autophagy as indicated by the reduction of fragmented/condensed nuclei, altered ratios of Bax/Bcl2, p-Bad/Bad, cytosol/mitochondrial cyt c and caspase-3 activation, and the restored levels of autophagy markers Beclin1, p62 and LC3B-II. Additionally, Akt was found to be activated and FOXO3 was phosphorylated on Ser253, suggesting inhibition of FOXO3 transcriptional function. Conclusions Asialo-rhuEPOP-mediated cardioprotection occurs through activation of PI3K/Akt pathway leading to suppression of Mst1 activation and promoting cardiomyocyte survival. General significance Asialo-rhuEPOP could be used to modulate Mst1 activity elevated under numerous pathological states. Recombinant asialo-rhuEPO protect HL-1 cardiomyocytes against STS-induced injury. Protective effect of recombinant asialo-rhuEPO is superior to sialylated EPO. Asialo-rhuEPO suppresses activation of proapoptotic kinase MSt1 by activating Akt. Asialo-rhuEPO restores autophagy and inhibits apoptosis to promote cell survival.
Collapse
Affiliation(s)
- Farooqahmed S Kittur
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute & Technology Enterprise, North Carolina Central University, Durham, NC 27707, USA
| | - Yuan Lin
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute & Technology Enterprise, North Carolina Central University, Durham, NC 27707, USA.,School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Elena Arthur
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute & Technology Enterprise, North Carolina Central University, Durham, NC 27707, USA
| | - Chiu-Yueh Hung
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute & Technology Enterprise, North Carolina Central University, Durham, NC 27707, USA
| | - P Andy Li
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute & Technology Enterprise, North Carolina Central University, Durham, NC 27707, USA
| | - David C Sane
- Carilion Clinic and Virginia Tech Carilion School of Medicine, Roanoke, VA 24014, USA
| | - Jiahua Xie
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute & Technology Enterprise, North Carolina Central University, Durham, NC 27707, USA
| |
Collapse
|
29
|
Erythropoietin stimulates fibroblast growth factor 23 (FGF23) in mice and men. Pflugers Arch 2018; 470:1569-1582. [DOI: 10.1007/s00424-018-2171-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 06/08/2018] [Accepted: 06/20/2018] [Indexed: 02/06/2023]
|
30
|
Elliot-Portal E, Laouafa S, Arias-Reyes C, Janes TA, Joseph V, Soliz J. Brain-derived erythropoietin protects from intermittent hypoxia-induced cardiorespiratory dysfunction and oxidative stress in mice. Sleep 2018; 41:4985474. [PMID: 29697839 PMCID: PMC6047438 DOI: 10.1093/sleep/zsy072] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 02/21/2018] [Indexed: 12/21/2022] Open
Abstract
Study Objectives Based on the fact that erythropoietin (Epo) administration in rodents protects against spatial learning and cognitive deficits induced by chronic intermittent hypoxia (CIH)-mediated oxidative damage, here we tested the hypothesis that Epo in the brain protects against cardiorespiratory disorders and oxidative stress induced by CIH in adult mice. Methods Adult control and transgenic mice overexpressing Epo in the brain only (Tg21) were exposed to CIH (21%-10% O2-10 cycles/hour-8 hours/day-7 days) or room air. After CIH exposure, we used the tail cuff method to measure arterial pressure, and whole-body plethysmography to assess the frequency of apneic episodes at rest, minute ventilation, and ventilatory responses to hypoxia and hypercapnia. Finally, the activity of pro-oxidant (XO-xanthine oxidase, and NADPH) and antioxidant (super oxide dismutase) enzymes was evaluated in the cerebral cortex and brainstem. Results Exposure of control mice to CIH significantly increased the heart rate and arterial pressure, the number of apneic events, and the ventilatory response to hypoxia and hypercapnia. Furthermore, CIH increased the ratio of pro-oxidant to antioxidant enzymes in cortex and brainstem tissues. Both physiological and molecular changes induced by CIH were prevented in transgenic Tg21 mice. Conclusions We conclude that the neuroprotective effect of Epo prevents oxidative damage in the brain and cardiorespiratory disorders induced by CIH. Considering that Epo is used in clinics to treat chronic kidney disease and stroke, our data show convincing evidence suggesting that Epo may be a promising alternative drug to treat sleep-disorder breathing.
Collapse
Affiliation(s)
- Elizabeth Elliot-Portal
- Centre de Recherche de l’Institut Universitaire, de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Sofien Laouafa
- Centre de Recherche de l’Institut Universitaire, de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Christian Arias-Reyes
- Centre de Recherche de l’Institut Universitaire, de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Tara Adele Janes
- Centre de Recherche de l’Institut Universitaire, de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Vincent Joseph
- Centre de Recherche de l’Institut Universitaire, de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Jorge Soliz
- Centre de Recherche de l’Institut Universitaire, de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| |
Collapse
|
31
|
Hematopoietic Stem Cells but Not Multipotent Progenitors Drive Erythropoiesis during Chronic Erythroid Stress in EPO Transgenic Mice. Stem Cell Reports 2018; 10:1908-1919. [PMID: 29754961 PMCID: PMC5989815 DOI: 10.1016/j.stemcr.2018.04.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 04/13/2018] [Accepted: 04/16/2018] [Indexed: 12/30/2022] Open
Abstract
The hematopoietic stem cell (HSC) compartment consists of a small pool of cells capable of replenishing all blood cells. Although it is established that the hematopoietic system is assembled as a hierarchical organization under steady-state conditions, emerging evidence suggests that distinct differentiation pathways may exist in response to acute stress. However, it remains unclear how different hematopoietic stem and progenitor cell subpopulations behave under sustained chronic stress. Here, by using adult transgenic mice overexpressing erythropoietin (EPO; Tg6) and a combination of in vivo, in vitro, and deep-sequencing approaches, we found that HSCs respond differentially to chronic erythroid stress compared with their closely related multipotent progenitors (MPPs). Specifically, HSCs exhibit a vastly committed erythroid progenitor profile with enhanced cell division, while MPPs display erythroid and myeloid cell signatures and an accumulation of uncommitted cells. Thus, our results identify HSCs as master regulators of chronic stress erythropoiesis, potentially circumventing the hierarchical differentiation-detour.
Collapse
|
32
|
Mechanisms and mediators of hypertension induced by erythropoietin and related molecules. Nephrol Dial Transplant 2017; 33:1690-1698. [DOI: 10.1093/ndt/gfx324] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 10/23/2017] [Indexed: 11/14/2022] Open
|
33
|
Moransard M, Bednar M, Frei K, Gassmann M, Ogunshola OO. Erythropoietin reduces experimental autoimmune encephalomyelitis severity via neuroprotective mechanisms. J Neuroinflammation 2017; 14:202. [PMID: 29029628 PMCID: PMC5640948 DOI: 10.1186/s12974-017-0976-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 10/03/2017] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Treatment with erythropoietin (Epo) in experimental autoimmune encephalomyelitis (EAE), the rodent model of multiple sclerosis (MS), has consistently been shown to ameliorate disease progression and improve overall outcome. The effect has been attributed to modulation of the immune response and/or preservation of the central nervous system (CNS) tissue integrity. It remains unclear, however, if (a) Epo acts primarily in the CNS or the periphery and if (b) Epo's beneficial effect in EAE is mainly due to maintaining CNS tissue integrity or to modulation of the immune response. If Epo acts primarily by modulating the immune system, where is this modulation required? In the periphery, the CNS or both? METHODS To address these questions, we used two well-characterized transgenic mouse strains that constitutively overexpress recombinant human Epo (rhEpo) either systemically (tg6) or in CNS only (tg21) in a MOG-induced EAE model. We assessed clinical severity, disease progression, immunomodulation, and CNS tissue integrity, including neuronal survival. RESULTS Although disease onset remained unaffected, EAE progression was alleviated in transgenic animals compared to controls with both lines performing equally well showing that expression of Epo in the periphery is not required; Epo expression in the CNS is sufficient. Immunomodulation was observed in both strains but surprisingly the profile of modulation differed substantially between strains. Modulation in the tg21 strain was limited to a reduction in macrophages in the CNS, with no peripheral immunomodulatory effects observed. In contrast, in the tg6 strain, macrophages were upregulated in the CNS, and, in the periphery of this strain, T cells and macrophages were downregulated. The lack of a consistent immunomodulatory profile across both transgenic species suggests that immunomodulation by Epo is unlikely to be the primary mechanism driving amelioration of EAE. Finally, CNS tissue integrity was affected in all strains. Although myelin appeared equally damaged in all strains, neuronal survival was significantly improved in the spinal cord of tg21 mice, indicating that Epo may ameliorate EAE predominantly by protecting neurons. CONCLUSIONS Our data suggests that moderate elevated brain Epo levels provide clinically significant neuroprotection in EAE without modulation of the immune response making a significant contribution.
Collapse
Affiliation(s)
- M Moransard
- Department of Internal Medicine, Section of Clinical Immunology, University Hospital Zürich, Zurich, Switzerland
| | - M Bednar
- Institute of Veterinary Physiology and Zurich Center of Integrative Human Physiology (ZIHP), Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 260, CH-8057, Zurich, Switzerland
| | - K Frei
- Department of Neurosurgery, University Hospital Zurich, CH-8006, Zurich, Switzerland
| | - M Gassmann
- Institute of Veterinary Physiology and Zurich Center of Integrative Human Physiology (ZIHP), Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 260, CH-8057, Zurich, Switzerland
- Universidad Peruana Cayetano Heredia (UPCH), Lima, Peru
| | - O O Ogunshola
- Institute of Veterinary Physiology and Zurich Center of Integrative Human Physiology (ZIHP), Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 260, CH-8057, Zurich, Switzerland.
| |
Collapse
|
34
|
Gilboa D, Haim-Ohana Y, Deshet-Unger N, Ben-Califa N, Hiram-Bab S, Reuveni D, Zigmond E, Gassmann M, Gabet Y, Varol C, Neumann D. Erythropoietin enhances Kupffer cell number and activity in the challenged liver. Sci Rep 2017; 7:10379. [PMID: 28871174 PMCID: PMC5583293 DOI: 10.1038/s41598-017-11082-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 08/15/2017] [Indexed: 02/07/2023] Open
Abstract
Erythropoietin (EPO) is the main hormone driving mammalian erythropoiesis, with activity mediated via the surface receptor, EPO-R, on erythroid progenitor cells. Recombinant human EPO is currently used clinically for the treatment of anemia in patients with end-stage renal disease, and in certain cancer patients suffering from anemia induced either by the tumor itself or by chemotherapy. EPO-R expression is also detected in non-erythroid cells, including macrophages present in the peritoneum, spleen, and bone marrow (BM). Here we demonstrate that Kupffer cells (KCs) - the liver-resident macrophages - are EPO targets. We show that, in vitro, EPO initiated intracellular signalling and enhanced phagocytosis in a rat KC line (RKC-2) and in sorted KCs. Moreover, continuous EPO administration in mice, resulted in an increased number of KCs, up-regulation of liver EPO-R expression and elevated production of the monocyte chemoattractant CCL2, with corresponding egress of Ly6Chi monocytes from the BM. In a model of acute acetaminophen-induced liver injury, EPO administration increased the recruitment of Ly6Chi monocytes and neutrophils to the liver. Taken together, our results reveal a new role for EPO in stimulating KC proliferation and phagocytosis, and in recruiting Ly6Chi monocytes in response to liver injury.
Collapse
Affiliation(s)
- Dafna Gilboa
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yasmin Haim-Ohana
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Naamit Deshet-Unger
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nathalie Ben-Califa
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sahar Hiram-Bab
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Debby Reuveni
- The Research Center for Digestive Tract and Liver Diseases, Sourasky Medical Center and Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ehud Zigmond
- The Research Center for Digestive Tract and Liver Diseases, Sourasky Medical Center and Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Max Gassmann
- Institute for Veterinary Physiology, Vetsuisse Faculty and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Yankel Gabet
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Chen Varol
- The Research Center for Digestive Tract and Liver Diseases, Sourasky Medical Center and Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Drorit Neumann
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
35
|
Baum O, Jentsch L, Odriozola A, Tschanz SA, Olfert IM. Ultrastructure of Skeletal Muscles in Mice Lacking Muscle‐Specific VEGF Expression. Anat Rec (Hoboken) 2017; 300:2239-2249. [DOI: 10.1002/ar.23644] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 04/11/2017] [Accepted: 04/30/2017] [Indexed: 01/22/2023]
Affiliation(s)
- Oliver Baum
- Institute of Physiology, Charité‐Universitätsmedizin BerlinBerlin Germany
| | - Lena Jentsch
- Institute of Anatomy, University of BernBern Switzerland
| | | | | | - I. Mark Olfert
- West Virginia Clinical and Translational Institute, Department of Exercise PhysiologyWest Virginia University School of MedicineMorgantown West Virginia
| |
Collapse
|
36
|
Nunomiya A, Shin J, Kitajima Y, Dan T, Miyata T, Nagatomi R. Activation of the hypoxia-inducible factor pathway induced by prolyl hydroxylase domain 2 deficiency enhances the effect of running training in mice. Acta Physiol (Oxf) 2017; 220:99-112. [PMID: 27393382 PMCID: PMC5412909 DOI: 10.1111/apha.12751] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 12/19/2015] [Accepted: 07/04/2016] [Indexed: 12/15/2022]
Abstract
AIMS Hypoxic response mediated by hypoxia-inducible factor (HIF) seems to contribute to the benefit of endurance training. To verify the direct contribution of HIF activation to running training without exposure to atmospheric hypoxia, we used prolyl hydroxylase domain 2 (PHD2) conditional knockout mice (cKO), which exhibit HIF activation independent of oxygen concentration, and we examined their maximal exercise capacity before and after 4 weeks of treadmill exercise training. METHODS Phd2f/f mice (n = 26) and Phd2 cKO mice (n = 24) were randomly divided into two groups, trained and untrained, and were subjected to maximal running test before and after a 4-week treadmill-training regimen. RESULTS Prolyl hydroxylase domain 2 deficiency resulted in HIF-α protein accumulation. Phd2 cKO mice exhibited marked increases in haematocrit values and haemoglobin concentrations, as well as an increase in the capillary number in the skeletal muscle. The 4-week training elicited an increase in the capillary-to-fibre (C/F) ratio and succinyl dehydrogenase activity of the skeletal muscle. Importantly, trained Phd2 cKO mice showed a significantly greater improvement in running time than trained control mice (P < 0.05). Collectively, these data suggest that the combination of training and the activation of the HIF pathway are important for maximizing the effect of running training. CONCLUSION We conclude that the activation of the HIF pathway induced by PHD2 deficiency enhances the effect of running training.
Collapse
Affiliation(s)
- A. Nunomiya
- Department of Medicine and Science in Sports and Exercise; Tohoku University Graduate School of Medicine; Sendai Japan
| | - J. Shin
- Division of Biomedical Engineering for Health & Welfare; Tohoku University Graduate School of Biomedical Engineering; Sendai Japan
| | - Y. Kitajima
- Division of Biomedical Engineering for Health & Welfare; Tohoku University Graduate School of Biomedical Engineering; Sendai Japan
| | - T. Dan
- Division of Molecular Medicine and Therapy; United Centers for Advanced Research and Translational Medicine; Tohoku University Graduate School of Medicine; Sendai Japan
| | - T. Miyata
- Division of Molecular Medicine and Therapy; United Centers for Advanced Research and Translational Medicine; Tohoku University Graduate School of Medicine; Sendai Japan
| | - R. Nagatomi
- Department of Medicine and Science in Sports and Exercise; Tohoku University Graduate School of Medicine; Sendai Japan
- Division of Biomedical Engineering for Health & Welfare; Tohoku University Graduate School of Biomedical Engineering; Sendai Japan
- Center for Sports Medicine and Science; United Centers for Advanced Research and Translational Medicine; Tohoku University Graduate School of Medicine; Sendai Japan
| |
Collapse
|
37
|
Laouafa S, Perrin-Terrin AS, Jeton F, Elliot-Portal E, Tam R, Bodineau L, Voituron N, Soliz J. Pharmacological, but not genetic, alteration of neural Epo modifies the CO 2/H + central chemosensitivity in postnatal mice. Respir Physiol Neurobiol 2017; 242:73-79. [PMID: 28396201 DOI: 10.1016/j.resp.2017.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 03/24/2017] [Accepted: 04/05/2017] [Indexed: 11/24/2022]
Abstract
Cerebral erythropoietin (Epo) plays a crucial role for respiratory control in newborn rodents. We showed previously that soluble Epo receptor (sEpoR: an Epo antagonist) reduces basal ventilation and hypoxic hyperventilation at postnatal day 10 (P10) and in adult mice. However, at these ages (P10 and adulthood), Epo had no effect on central chemosensitivity. Nevertheless, it is known that the sensitivity to CO2/H+ during the mammalian respiratory network maturation process is age-dependent. Accordingly, in this study we wanted to test the hypothesis that cerebral Epo is involved in the breathing stimulation induced by the activation of central CO2/H+ chemoreceptors at earlier postnatal ages. To this end, en bloc brainstem-spinal cord preparations were obtained from P4 mice and the fictive breathing response to CO2-induced acidosis or metabolic acidosis was analyzed. This age (P4) was chosen because previous research from our laboratory showed that Epo altered (in a dose- and time-dependent manner) the fictive ventilation elicited in brainstem-spinal cord preparations. Moreover, as it was observed that peripheral chemoreceptors determined the respiratory sensitivity of central chemoreceptors to CO2, the use of this technique restricts our observations to central modulation. Our results did not show differences between preparations from control and transgenic animals (Tg21: overexpressing cerebral Epo; Epo-TAgh: cerebral Epo deficient mice). However, when Tg21 brainstem preparations were incubated for 1h with sEpoR, or with inhibitors of ERK/Akt (thus blocking the activation of the Epo molecular pathway), the fictive breathing response to CO2-induced acidosis was blunted. Our data suggest that variation of the Epo/sEpoR ratio is central to breathing modulation during CO2 challenges, and calls attention to clinical perspectives based on the use of Epo drugs at birth in hypoventilation cases.
Collapse
Affiliation(s)
- Sofien Laouafa
- Université Laval, Faculté de Médecine, Centre de Recherche Institut universitaire de cardiologie et de pneumologie de Québec, Département de Pédiatrie, Québec, QC, Canada; LEHNA, UMR CNRS 5023, Ecologie des Hydrosystèmes Naturels et Anthropisés, Université de Lyon, Université Lyon 1, ENTPE, 6 rue Raphael Dubois, 69622 Villeurbanne, France
| | - Anne-Sophie Perrin-Terrin
- Université Paris 13, Sorbonne Paris Cité, UFR SMBH, Laboratoire "Hypoxie et poumons", EA 2363, 93017 Bobigny, France; Sorbonne Universités, UPMC Univ Paris 06, INSERM, UMR_S1158 Neurophysiologie Respiratoire Expérimentale et Clinique, F-75013, Paris, France
| | - Florine Jeton
- Université Paris 13, Sorbonne Paris Cité, UFR SMBH, Laboratoire "Hypoxie et poumons", EA 2363, 93017 Bobigny, France
| | - Elizabeth Elliot-Portal
- Université Laval, Faculté de Médecine, Centre de Recherche Institut universitaire de cardiologie et de pneumologie de Québec, Département de Pédiatrie, Québec, QC, Canada; Molecular biology and Biotechnology Institute, Universidad Mayor de San Andres, La Paz, Bolivia
| | - Rose Tam
- Université Laval, Faculté de Médecine, Centre de Recherche Institut universitaire de cardiologie et de pneumologie de Québec, Département de Pédiatrie, Québec, QC, Canada
| | - Laurence Bodineau
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, UMR_S1158 Neurophysiologie Respiratoire Expérimentale et Clinique, F-75013, Paris, France
| | - Nicolas Voituron
- Université Paris 13, Sorbonne Paris Cité, UFR SMBH, Laboratoire "Hypoxie et poumons", EA 2363, 93017 Bobigny, France
| | - Jorge Soliz
- Université Laval, Faculté de Médecine, Centre de Recherche Institut universitaire de cardiologie et de pneumologie de Québec, Département de Pédiatrie, Québec, QC, Canada; Molecular biology and Biotechnology Institute, Universidad Mayor de San Andres, La Paz, Bolivia.
| |
Collapse
|
38
|
|
39
|
Ma HQ, Low BS, Chan KL, Khan NAK. Lignans of Phyllanthus niruri Solid Dispersion: A Potential Alternative Gout Therapy. INT J PHARMACOL 2016. [DOI: 10.3923/ijp.2017.11.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
40
|
Laouafa S, Elliot-Portal E, Revollo S, Schneider Gasser EM, Joseph V, Voituron N, Gassmann M, Soliz J. Hypercapnic ventilatory response is decreased in a mouse model of excessive erythrocytosis. Am J Physiol Regul Integr Comp Physiol 2016; 311:R940-R947. [PMID: 27605561 DOI: 10.1152/ajpregu.00226.2016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 08/10/2016] [Indexed: 11/22/2022]
Abstract
The impact of cerebral erythropoietin (Epo) in the regulation of the hypercapnic ventilatory response (HcVR) is controversial. While we reported that cerebral Epo does not affect the central chemosensitivity in C57Bl6 mice receiving an intracisternal injection of sEpoR (the endogenous antagonist of Epo), a recent study in transgenic mice with constitutive high levels of human Epo in brain and circulation (Tg6) and in brain only (Tg21), showed that Epo blunts the HcVR, maybe by interacting with central and peripheral chemoreceptors. High Epo serum levels in Tg6 mice lead to excessive erythrocytosis (hematocrit ~80-90%), the main symptom of chronic mountain sickness (CMS). These latter results support the hypothesis that reduced central chemosensitivity accounts for the hypoventilation observed in CMS patients. To solve this intriguing divergence, we reevaluate HcVR in Tg6 and Tg21 mouse lines, by assessing the metabolic rate [O consumption (V̇) and CO production (V̇)], a key factor modulating ventilation, the effect of which was not considered in the previous study. Our results showed that the decreased HcVR observed in Tg6 mice (~70% reduction; < 0.01) was due to a significant decrease in the metabolism (~40%; < 0.0001) rather than Epo's effect on CO chemosensitivity. Additional analysis in Tg21 mice did not reveal differences of HcVR or metabolism. We concluded that cerebral Epo does not modulate the central chemosensitivity system, and that a metabolic effect upon CO inhalation is responsible for decreased HcVR observed in Tg6 animals. As CMS patients also show decreased HcVR, our findings might help to better understand respiratory disorders at high altitude.
Collapse
Affiliation(s)
- Sofien Laouafa
- Centre de Recherche du CHU de Québec, Pavillon St François d'Assise, Département de Pédiatrie, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Elizabeth Elliot-Portal
- Centre de Recherche du CHU de Québec, Pavillon St François d'Assise, Département de Pédiatrie, Faculté de Médecine, Université Laval, Québec, QC, Canada.,Molecular biology and Biotechnology Institute, Universidad Mayor de San Andres, La Paz, Bolivia
| | - Susana Revollo
- Centre de Recherche du CHU de Québec, Pavillon St François d'Assise, Département de Pédiatrie, Faculté de Médecine, Université Laval, Québec, QC, Canada.,Molecular biology and Biotechnology Institute, Universidad Mayor de San Andres, La Paz, Bolivia
| | - Edith M Schneider Gasser
- Institute of Veterinary Physiology, Vetsuisse Faculty, and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland; and
| | - Vincent Joseph
- Centre de Recherche du CHU de Québec, Pavillon St François d'Assise, Département de Pédiatrie, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Nicolas Voituron
- Université Paris 13, Sorbonne Paris Cité, UFR SMBH, Laboratoire "Hypoxie et poumons," Bobigny, France
| | - Max Gassmann
- Institute of Veterinary Physiology, Vetsuisse Faculty, and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland; and.,Universidad Peruana Cayetano Heredia (UPCH), Lima, Peru
| | - Jorge Soliz
- Centre de Recherche du CHU de Québec, Pavillon St François d'Assise, Département de Pédiatrie, Faculté de Médecine, Université Laval, Québec, QC, Canada;
| |
Collapse
|
41
|
Rauner M, Franke K, Murray M, Singh RP, Hiram-Bab S, Platzbecker U, Gassmann M, Socolovsky M, Neumann D, Gabet Y, Chavakis T, Hofbauer LC, Wielockx B. Increased EPO Levels Are Associated With Bone Loss in Mice Lacking PHD2 in EPO-Producing Cells. J Bone Miner Res 2016; 31:1877-1887. [PMID: 27082941 DOI: 10.1002/jbmr.2857] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 03/28/2016] [Accepted: 04/12/2016] [Indexed: 12/25/2022]
Abstract
The main oxygen sensor hypoxia inducible factor (HIF) prolyl hydroxylase 2 (PHD2) is a critical regulator of tissue homeostasis during erythropoiesis, hematopoietic stem cell maintenance, and wound healing. Recent studies point toward a role for the PHD2-erythropoietin (EPO) axis in the modulation of bone remodeling, even though the studies produced conflicting results. Here, we used a number of mouse strains deficient of PHD2 in different cell types to address the role of PHD2 and its downstream targets HIF-1α and HIF-2α in bone remodeling. Mice deficient for PHD2 in several cell lineages, including EPO-producing cells, osteoblasts, and hematopoietic cells (CD68:cre-PHD2f/f ) displayed a severe reduction of bone density at the distal femur as well as the vertebral body due to impaired bone formation but not bone resorption. Importantly, using osteoblast-specific (Osx:cre-PHD2f/f ) and osteoclast-specific PHD2 knock-out mice (Vav:cre- PHD2f/f ), we show that this effect is independent of the loss of PHD2 in osteoblast and osteoclasts. Using different in vivo and in vitro approaches, we show here that this bone phenotype, including the suppression of bone formation, is directly linked to the stabilization of the α-subunit of HIF-2, and possibly to the subsequent moderate induction of serum EPO, which directly influenced the differentiation and mineralization of osteoblast progenitors resulting in lower bone density. Taken together, our data identify the PHD2:HIF-2α:EPO axis as a so far unknown regulator of osteohematology by controlling bone homeostasis. Further, these data suggest that patients treated with PHD inhibitors or EPO should be monitored with respect to their bone status. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Martina Rauner
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Kristin Franke
- Department of Clinical Pathobiochemistry, Technische Universität Dresden, Dresden, Germany.,Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany
| | - Marta Murray
- Department of Clinical Pathobiochemistry, Technische Universität Dresden, Dresden, Germany.,Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany
| | - Rashim Pal Singh
- Department of Clinical Pathobiochemistry, Technische Universität Dresden, Dresden, Germany.,Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany
| | - Sahar Hiram-Bab
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Uwe Platzbecker
- Department of Medicine I, Technische Universität Dresden, Dresden, Germany
| | - Max Gassmann
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zürich, Zürich, Switzerland.,Universidad Peruana Cayetano Heredia (UPCH), Lima, Peru
| | - Merav Socolovsky
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA.,Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA
| | - Drorit Neumann
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Yankel Gabet
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Triantafyllos Chavakis
- Department of Clinical Pathobiochemistry, Technische Universität Dresden, Dresden, Germany.,Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany.,Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Lorenz C Hofbauer
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany.,Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Ben Wielockx
- Department of Clinical Pathobiochemistry, Technische Universität Dresden, Dresden, Germany. .,Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany. .,Center for Regenerative Therapies Dresden, Dresden, Germany.
| |
Collapse
|
42
|
Bianchi S, Fusi J, Franzoni F, Giovannini L, Galetta F, Mannari C, Guidotti E, Tocchini L, Santoro G. "Effects of recombinant human erythropoietin high mimicking abuse doses on oxidative stress processes in rats". Biomed Pharmacother 2016; 82:355-63. [PMID: 27470373 DOI: 10.1016/j.biopha.2016.05.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 05/12/2016] [Accepted: 05/13/2016] [Indexed: 12/21/2022] Open
Abstract
Although many studies highlight how long-term moderate dose of Recombinant Human Erythropoietin (rHuEPO) treatments result in beneficial and antioxidants effects, few studies take into account the effects that short-term high doses of rHuEPO (mimicking abuse conditions) might have on the oxidative stress processes. Thus, the aim of this study was to investigate the in vivo antioxidant activity of rHuEPO, administered for a short time and at high doses to mimic its sports abuse as doping. Male Wistar healthy rats (n=36) were recruited for the study and were treated with three different concentrations of rHuEPO: 7.5, 15, 30μg/kg. Plasma concentrations of erythropoietin, 8-epi Prostaglandin F2α, plasma and urinary concentrations of NOx were evaluated with specific assay kit, while hematocrit levels were analyzed with an automated cell counter. Antioxidant activity of rHuEPO was assessed analyzing the possible variation of the plasma scavenger capacity against hydroxylic and peroxylic radicals by TOSC (Total Oxyradical Scavenging Capacity) assay. Statistical analyses showed higher hematocrit values, confirmed by a statistically significant increase of plasmatic EPO concentration. An increase in plasma scavenging capacity against peroxyl and hydroxyl radicals, in 8-isoprostane plasmatic concentrations and in plasmatic and urinary levels of NOX were also found in all the treated animals, though not always statistically significant. Our results confirm the literature data regarding the antioxidant action of erythropoietin administered at low doses and for short times, whereas they showed an opposite incremental oxidative stress action when erythropoietin is administered at high doses.
Collapse
Affiliation(s)
- Sara Bianchi
- Department of Translational Research and New Technologies in Medicine and Surgery, Pharmacology, Medical School, University of Pisa, Via Roma 55, 56126, Pisa, Italy.
| | - Jonathan Fusi
- Department of Clinical and Experimental Medicine, University of Pisa, Via Savi 10, 56126, Pisa, Italy
| | - Ferdinando Franzoni
- Department of Clinical and Experimental Medicine, University of Pisa, Via Savi 10, 56126, Pisa, Italy
| | - Luca Giovannini
- Department of Translational Research and New Technologies in Medicine and Surgery, Pharmacology, Medical School, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Fabio Galetta
- Department of Clinical and Experimental Medicine, University of Pisa, Via Savi 10, 56126, Pisa, Italy
| | - Claudio Mannari
- Department of Translational Research and New Technologies in Medicine and Surgery, Pharmacology, Medical School, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Emanuele Guidotti
- Department of Clinical and Experimental Medicine, University of Pisa, Via Savi 10, 56126, Pisa, Italy
| | - Leonardo Tocchini
- Department of Clinical and Experimental Medicine, University of Pisa, Via Savi 10, 56126, Pisa, Italy
| | - Gino Santoro
- Department of Clinical and Experimental Medicine, University of Pisa, Via Savi 10, 56126, Pisa, Italy
| |
Collapse
|
43
|
Menuet C, Khemiri H, de la Poëze d'Harambure T, Gestreau C. Polycythemia and high levels of erythropoietin in blood and brain blunt the hypercapnic ventilatory response in adult mice. Am J Physiol Regul Integr Comp Physiol 2016; 310:R979-91. [PMID: 26936784 DOI: 10.1152/ajpregu.00474.2015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 02/26/2016] [Indexed: 11/22/2022]
Abstract
Changes in arterial Po2, Pco2, and pH are the strongest stimuli sensed by peripheral and central chemoreceptors to adjust ventilation to the metabolic demand. Erythropoietin (Epo), the main regulator of red blood cell production, increases the hypoxic ventilatory response, an effect attributed to the presence of Epo receptors in both carotid bodies and key brainstem structures involved in integration of peripheral inputs and control of breathing. However, it is not known whether Epo also has an effect on the hypercapnic chemoreflex. In a first attempt to answer this question, we tested the hypothesis that Epo alters the ventilatory response to increased CO2 levels. Basal ventilation and hypercapnic ventilatory response (HCVR) were recorded from control mice and from two transgenic mouse lines constitutively expressing high levels of human Epo in brain only (Tg21) or in brain and plasma (Tg6), the latter leading to polycythemia. To tease apart the potential effects of polycythemia and levels of plasma Epo in the HCVR, control animals were injected with an Epo analog (Aranesp), and Tg6 mice were treated with the hemolytic agent phenylhydrazine after splenectomy. Ventilatory parameters measured by plethysmography in conscious mice were consistent with data from electrophysiological recordings in anesthetized animals and revealed a blunted HCVR in Tg6 mice. Polycythemia alone and increased levels of plasma Epo blunt the HCVR. In addition, Tg21 mice with an augmented level of cerebral Epo also had a decreased HCVR. We discuss the potential implications of these findings in several physiopathological conditions.
Collapse
|
44
|
Oikonomidou PR, Casu C, Yang Z, Crielaard B, Shim JH, Rivella S, Vogiatzi MG. Polycythemia is associated with bone loss and reduced osteoblast activity in mice. Osteoporos Int 2016; 27:1559-1568. [PMID: 26650379 PMCID: PMC5319412 DOI: 10.1007/s00198-015-3412-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 11/05/2015] [Indexed: 01/19/2023]
Abstract
UNLABELLED Increased fragility has been described in humans with polycythemia vera (PV). Herein, we describe an osteoporotic phenotype associated with decreased osteoblast activity in a mouse model of PV and another mouse of polycythemia and elevated circulating erythropoietin (EPO). Our results are important for patients with PV or those treated with recombinant EPO (rEPO). INTRODUCTION PV and other myeloproliferative syndromes have been recently associated with an increased risk for fractures. However, the presence of osteoporosis in these patients has not been well documented. EPO, a hormone primarily known to stimulate erythropoiesis, has been shown recently to regulate bone homeostasis in mice. The aim of this study was to examine the bone phenotype of a mouse model of PV and compare it to that of animals with polycythemia caused by elevated circulating EPO. METHODS Bone mass and remodeling were evaluated by micro-computed tomography and histomorphometry. The JAK2(V617F) knock-in mouse, a model of human PV, manifests polycythemia and low circulating EPO levels. Results from this mouse were compared to wild type (wt) controls and the tg6 transgenic mouse that shows polycythemia caused by increased constitutive expression of EPO. RESULTS Compared to wt, both JAK2(V617F) and tg6 mice had a decrease in trabecular bone mass. Tg6 mice showed an additional modest decrease in cortical thickness and cortical bone volume per tissue volume (P < 0.01) suggesting a more severe bone phenotype than JAK2(V617F). Decreased osteoblast numbers and bone formation along with normal osteoclast numbers and activity were found in both mice. CONCLUSIONS This study indicates that PV is associated with low bone mass and decreased osteoblast activity in mice. Our results support future studies of osteoporosis in affected humans. Polycythemia caused by chronically elevated circulating EPO also results in bone loss, and implications on patients treated with rEPO should be evaluated.
Collapse
Affiliation(s)
- P R Oikonomidou
- Division of Pediatric Endocrinology, Weill Cornell Medical College, New York, NY, USA.
- Division of Hematology, Children's Hospital of Philadelphia, Abramson Research Center, 3615 Civic Center Blvd., Room 309 F, Philadelphia, PA, 19104, USA.
| | - C Casu
- Division of Pediatric Hematology/Oncology, Weill Cornell Medical College, New York, NY, USA
- Division of Hematology, Children's Hospital of Philadelphia, Abramson Research Center, 3615 Civic Center Blvd., Room 309 F, Philadelphia, PA, 19104, USA
| | - Z Yang
- Department of Medicine, Cancer Center, Weill Cornell Medical College, Belfer Research Building, 413 East 69th Street, 13th Floor, New York, NY,, 10021, USA
| | - B Crielaard
- Division of Pediatric Hematology/Oncology, Weill Cornell Medical College, New York, NY, USA
- Department of Polymer Chemistry and Bioengineering, Zernike Institute for Advanced Materials, University of Groningen, Nijenborgh 4, 9747 AG, Groningen, Netherlands
| | - J H Shim
- Department of Pathology and Laboratory medicine, Weill Cornell Medical College, 1300 York Avenue, E-904, New York, NY,, 10065, USA
| | - S Rivella
- Division of Pediatric Hematology/Oncology, Weill Cornell Medical College, New York, NY, USA
- Division of Hematology, Children's Hospital of Philadelphia, Abramson Research Center, 3615 Civic Center Blvd., Room 316 B, Philadelphia, PA, 19104, USA
| | - M G Vogiatzi
- Division of Pediatric Endocrinology, Weill Cornell Medical College, New York, NY, USA
- Division of Endocrinology, Children's Hospital of Philadelphia, 3401 Civic Center Bvld., Philadelphia, PA, 19104, USA
| |
Collapse
|
45
|
Kara A, Cetin H, Oktem F, Metin Ciris I, Altuntas I, Kaya S. Amikacin induced renal damage and the role of the antioxidants on neonatal rats. Ren Fail 2016; 38:671-7. [PMID: 26982694 DOI: 10.3109/0886022x.2016.1155393] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Amikacin (AK) is frequently used on the treatment of Gram-negative infections on neonates, but its usage is restricted because of nephrotoxicity. In this study, on neonatal rats, we aimed to investigate the effects of erythropoietin and vitamin E on AK induced nephrotoxicity. A total of 35 newborn Wistar Albino rats were divided into four groups: (1) injected with saline (serum physiological was administered to placebo controls), (2) injected with AK (1200 mg/kg), (3) injected with AK + vitamin E (150 mg/kg), (4) injected with AK + erythropoietin (EPO) (300 IU/kg/day). In renal tissue, AK levels were significantly high in all groups except the control. Tissue malondialdehyde (MDA) and nitric oxide (NO) levels were statistically higher in AK -treated group than the control. MDA and NO levels were significantly decreased with the administration of vitamin E and EPO. Glutathione peroxidase (GPX) levels were statistically low in AK group compared with the controls. The levels of GPX, in vitamin E group, were increased significantly. However, superoxide dismutase and catalase levels were not significantly different in none of the groups. Insulin-like growth factor-1 values in AK, EPO and vitamin E groups were significantly higher than the control group. Histomorphological changes such as tubular epithelial necrosis were seen in AK treated group. Histopathological improvements observed with EPO and vitamin E administration. AK nephrotoxicity is related to oxidative stress and is supported with biochemical and histopathological findings. Vitamin E and EPO, as antioxidants, can be useful renoprotective agents for ameliorating AK induced nephrotoxicity in neonates.
Collapse
Affiliation(s)
- Aslihan Kara
- a Department of Pediatric Nephrology, Medical Faculty , Firat University , Elazig , Turkey
| | - Hasan Cetin
- b Department of Pediatric Neonatology, Medical Faculty , Suleyman Demirel University , Isparta , Turkey
| | - Faruk Oktem
- c Department of Pediatric Nephrology, Medical Faculty , Bezmialem Vakif University , Istanbul , Turkey
| | - Ibrahim Metin Ciris
- d Department of Pathology, Medical Faculty , Suleyman Demirel University , Isparta , Turkey
| | - Irfan Altuntas
- e Department of Biochemistry, Medical Faculty , Dumlupinar University , Kutahya , Turkey
| | - Selcuk Kaya
- f Department of Microbiology, Medical Faculty , Katip Celebi University , Izmir , Turkey
| |
Collapse
|
46
|
Recombinant Human Erythropoietin Protects Myocardial Cells from Apoptosis via the Janus-Activated Kinase 2/Signal Transducer and Activator of Transcription 5 Pathway in Rats with Epilepsy. Curr Ther Res Clin Exp 2015; 77:90-8. [PMID: 26649078 PMCID: PMC4644243 DOI: 10.1016/j.curtheres.2015.07.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2015] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE To investigate the potential mechanisms underlying the protective effects of recombinant human erythropoietin (rhEPO) and carbamylated EPO (CEPO) against myocardial cell apoptosis in epilepsy. METHODS Rats were given an intra-amygdala injection of kainic acid to induce epilepsy. Groups of rats were treated with rhEPO or CEPO before induction of epilepsy, whereas additional rats were given a caudal vein injection of AG490, a selective inhibitor of Janus kinase 2 (JAK2). At different time points after seizure onset, electroencephalogram changes were recorded, and myocardium samples were taken for the detection of myocardial cell apoptosis and expression of JAK2, signal transducer and activator of transcription 5 (STAT5), caspase-3, and bcl-xl mRNAs and proteins. RESULTS Induction of epilepsy significantly enhanced myocardial cell apoptosis and upregulated the expression of caspase-3 and bcl-xl proteins and JAK2 and STAT5a at both the mRNA and protein levels. Pretreatment with either rhEPO or CEPO reduced the number of apoptotic cells, upregulated bcl-xl expression, and downregulated caspase-3 expression in the myocardium of epileptic rats. Both myocardial JAK2 and STAT5a mRNAs, as well as phosphorylated species of JAK2 and STAT5a, were upregulated in epileptic rats in response to rhEPO-but not to CEPO-pretreatment. AG490 treatment increased apoptosis, upregulated caspase-3 protein expression, and downregulated bcl-xl protein expression in the myocardium of epileptic rats. CONCLUSIONS These results indicate that myocardial cell apoptosis may contribute to myocardial injury in epilepsy. EPO protects myocardial cells from apoptosis via the JAK2/STAT5 pathway in rats with experimental epilepsy, whereas CEPO exerts antiapoptotic activity perhaps via a pathway independent of JAK2/STAT5 signaling.
Collapse
|
47
|
Caravagna C, Schneider Gasser EM, Ballot O, Joseph V, Soliz J. Chronic overexpression of cerebral Epo improves the ventilatory response to acute hypoxia during the postnatal development. Int J Dev Neurosci 2015; 44:84-91. [DOI: 10.1016/j.ijdevneu.2015.06.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 06/01/2015] [Accepted: 06/04/2015] [Indexed: 10/23/2022] Open
Affiliation(s)
- Céline Caravagna
- Department of PediatricsLaval University Center de Recherche du Center Hospitalier Universitaire (CHU) de Québec, Hôpital St‐François d'Assise10 Rue de l'EspinayQuébecQCG1L 3L5Canada
| | - Edith M. Schneider Gasser
- Institute of Veterinary PhysiologyVetsuisse‐Faculty University of ZurichWinterthurerstrasse 260CH‐8057ZurichSwitzerland
| | - Orlane Ballot
- Department of PediatricsLaval University Center de Recherche du Center Hospitalier Universitaire (CHU) de Québec, Hôpital St‐François d'Assise10 Rue de l'EspinayQuébecQCG1L 3L5Canada
| | - Vincent Joseph
- Department of PediatricsLaval University Center de Recherche du Center Hospitalier Universitaire (CHU) de Québec, Hôpital St‐François d'Assise10 Rue de l'EspinayQuébecQCG1L 3L5Canada
| | - Jorge Soliz
- Department of PediatricsLaval University Center de Recherche du Center Hospitalier Universitaire (CHU) de Québec, Hôpital St‐François d'Assise10 Rue de l'EspinayQuébecQCG1L 3L5Canada
| |
Collapse
|
48
|
β Common Receptor Mediates Erythropoietin-Conferred Protection on OxLDL-Induced Lipid Accumulation and Inflammation in Macrophages. Mediators Inflamm 2015; 2015:439759. [PMID: 26101463 PMCID: PMC4458544 DOI: 10.1155/2015/439759] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 12/31/2014] [Indexed: 11/17/2022] Open
Abstract
Erythropoietin (EPO), the key factor for erythropoiesis, also protects macrophage foam cells from lipid accumulation, yet the definitive mechanisms are not fully understood. β common receptor (βCR) plays a crucial role in the nonhematopoietic effects of EPO. In the current study, we investigated the role of βCR in EPO-mediated protection in macrophages against oxidized low-density lipoprotein- (oxLDL-) induced deregulation of lipid metabolism and inflammation. Here, we show that βCR expression was mainly in foamy macrophages of atherosclerotic aortas from apolipoprotein E-deficient mice. Results of confocal microscopy and immunoprecipitation analyses revealed that βCR was colocalized and interacted with EPO receptor (EPOR) in macrophages. Inhibition of βCR activation by neutralizing antibody or small interfering RNA (siRNA) abolished the EPO-conferred protection in oxLDL-induced lipid accumulation. Furthermore, EPO-promoted cholesterol efflux and upregulation of ATP-binding cassette (ABC) transporters ABCA1 and ABCG1 were prevented by pretreatment with βCR neutralizing antibody or βCR siRNA. Additionally, blockage of βCR abrogated the EPO-conferred anti-inflammatory action on oxLDL-induced production of macrophage inflammatory protein-2. Collectively, our findings suggest that βCR may play an important role in the beneficial effects of EPO against oxLDL-elicited dysfunction of macrophage foam cells.
Collapse
|
49
|
Hiram-Bab S, Liron T, Deshet-Unger N, Mittelman M, Gassmann M, Rauner M, Franke K, Wielockx B, Neumann D, Gabet Y. Erythropoietin directly stimulates osteoclast precursors and induces bone loss. FASEB J 2015; 29:1890-900. [PMID: 25630969 DOI: 10.1096/fj.14-259085] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 12/23/2014] [Indexed: 12/24/2022]
Abstract
Erythropoietin (EPO) primarily regulates red blood cell formation, and EPO serum levels are increased on hypoxic stress (e.g., anemia and altitude). In addition to anemia, recent discoveries suggest new therapeutic indications for EPO, unrelated to erythropoiesis. We investigated the skeletal role of EPO using several models of overexpression (Tg6 mice) and EPO administration (intermittent/continuous, high/low doses) in adult C57Bl6 female mice. Using microcomputed tomography, histology, and serum markers, we found that EPO induced a 32%-61% trabecular bone loss caused by increased bone resorption (+60%-88% osteoclast number) and reduced bone formation rate (-19 to -74%; P < 0.05 throughout). EPO targeted the monocytic lineage by increasing the number of bone monocytes/macrophages, preosteoclasts, and mature osteoclasts. In contrast to the attenuated bone formation in vivo, EPO treatment in vitro did not inhibit osteoblast differentiation and activity, suggesting an indirect effect of EPO on osteoblasts. However, EPO had a direct effect on preosteoclasts by stimulating osteoclastogenesis in isolated cultures (+60%) via the Jak2 and PI3K pathways. In summary, our findings demonstrate that EPO negatively regulates bone mass and thus bears significant clinical implications for the potential management of patients with endogenously or therapeutically elevated EPO levels.
Collapse
Affiliation(s)
- Sahar Hiram-Bab
- *Department of Cell and Developmental Biology, Department of Anatomy and Anthropology, and Department of Medicine, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Institute of Veterinary Physiology, Vetsuisse Faculty, and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland; Department of Medicine III, Dresden University Medical Center, Dresden, Germany; and Institute of Pathology, University of Technology, Dresden, Germany
| | - Tamar Liron
- *Department of Cell and Developmental Biology, Department of Anatomy and Anthropology, and Department of Medicine, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Institute of Veterinary Physiology, Vetsuisse Faculty, and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland; Department of Medicine III, Dresden University Medical Center, Dresden, Germany; and Institute of Pathology, University of Technology, Dresden, Germany
| | - Naamit Deshet-Unger
- *Department of Cell and Developmental Biology, Department of Anatomy and Anthropology, and Department of Medicine, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Institute of Veterinary Physiology, Vetsuisse Faculty, and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland; Department of Medicine III, Dresden University Medical Center, Dresden, Germany; and Institute of Pathology, University of Technology, Dresden, Germany
| | - Moshe Mittelman
- *Department of Cell and Developmental Biology, Department of Anatomy and Anthropology, and Department of Medicine, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Institute of Veterinary Physiology, Vetsuisse Faculty, and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland; Department of Medicine III, Dresden University Medical Center, Dresden, Germany; and Institute of Pathology, University of Technology, Dresden, Germany
| | - Max Gassmann
- *Department of Cell and Developmental Biology, Department of Anatomy and Anthropology, and Department of Medicine, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Institute of Veterinary Physiology, Vetsuisse Faculty, and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland; Department of Medicine III, Dresden University Medical Center, Dresden, Germany; and Institute of Pathology, University of Technology, Dresden, Germany
| | - Martina Rauner
- *Department of Cell and Developmental Biology, Department of Anatomy and Anthropology, and Department of Medicine, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Institute of Veterinary Physiology, Vetsuisse Faculty, and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland; Department of Medicine III, Dresden University Medical Center, Dresden, Germany; and Institute of Pathology, University of Technology, Dresden, Germany
| | - Kristin Franke
- *Department of Cell and Developmental Biology, Department of Anatomy and Anthropology, and Department of Medicine, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Institute of Veterinary Physiology, Vetsuisse Faculty, and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland; Department of Medicine III, Dresden University Medical Center, Dresden, Germany; and Institute of Pathology, University of Technology, Dresden, Germany
| | - Ben Wielockx
- *Department of Cell and Developmental Biology, Department of Anatomy and Anthropology, and Department of Medicine, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Institute of Veterinary Physiology, Vetsuisse Faculty, and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland; Department of Medicine III, Dresden University Medical Center, Dresden, Germany; and Institute of Pathology, University of Technology, Dresden, Germany
| | - Drorit Neumann
- *Department of Cell and Developmental Biology, Department of Anatomy and Anthropology, and Department of Medicine, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Institute of Veterinary Physiology, Vetsuisse Faculty, and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland; Department of Medicine III, Dresden University Medical Center, Dresden, Germany; and Institute of Pathology, University of Technology, Dresden, Germany
| | - Yankel Gabet
- *Department of Cell and Developmental Biology, Department of Anatomy and Anthropology, and Department of Medicine, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Institute of Veterinary Physiology, Vetsuisse Faculty, and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland; Department of Medicine III, Dresden University Medical Center, Dresden, Germany; and Institute of Pathology, University of Technology, Dresden, Germany
| |
Collapse
|
50
|
dʼUscio LV, Santhanam AVR, Katusic ZS. Erythropoietin prevents endothelial dysfunction in GTP-cyclohydrolase I-deficient hph1 mice. J Cardiovasc Pharmacol 2014; 64:514-21. [PMID: 25490417 PMCID: PMC4261745 DOI: 10.1097/fjc.0000000000000145] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
: In this study, we used the mutant hph1 mouse model, which has deficiency in GTP-cyclohydrolase I (GTPCH I) activity, to test the hypothesis that erythropoietin (EPO) protects aortic wall from oxidative stress induced by uncoupling of endothelial nitric oxide synthase (eNOS). Both GTPCH I activity and tetrahydrobiopterin (BH4) levels were reduced in hph1 mice, whereas 7,8-dihydrobiopterin (7,8-BH2) levels were significantly increased. Furthermore, BH4 deficiency caused increased production of superoxide anion and hydrogen peroxide in the aorta thus resulting in impairment of endothelium-dependent relaxations to acetylcholine. Treatment of hph1 mice with recombinant human EPO (1000 U/kg, subcutaneously for 3 days) significantly decreased superoxide anion production by eNOS and improved BH4 to 7,8-BH2 ratio in aortas. EPO also significantly decreased production of hydrogen peroxide and improved endothelium-dependent relaxations in aortas of hph1 mice. In addition, EPO treatment increased protein expressions of copper-/zinc-superoxide dismutase, manganese-superoxide dismutase, and catalase in the aorta of hph1 mice. Our findings demonstrate that treatment with EPO prevented oxidative stress and endothelial dysfunction caused by eNOS uncoupling. Increased vascular expressions of antioxidants seem to be an important molecular mechanism underlying vascular protection by EPO during chronic BH4 deficiency.
Collapse
Affiliation(s)
- Livius V dʼUscio
- Departments of *Anesthesiology; and †Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN
| | | | | |
Collapse
|