1
|
Ziaka M, Exadaktylos A. ARDS associated acute brain injury: from the lung to the brain. Eur J Med Res 2022; 27:150. [PMID: 35964069 PMCID: PMC9375183 DOI: 10.1186/s40001-022-00780-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 07/29/2022] [Indexed: 01/10/2023] Open
Abstract
A complex interrelation between lung and brain in patients with acute lung injury (ALI) has been established by experimental and clinical studies during the last decades. Although, acute brain injury represents one of the most common insufficiencies in patients with ALI and acute respiratory distress syndrome (ARDS), the underlying pathophysiology of the observed crosstalk remains poorly understood due to its complexity. Specifically, it involves numerous pathophysiological parameters such as hypoxemia, neurological adverse events of lung protective ventilation, hypotension, disruption of the BBB, and neuroinflammation in such a manner that the brain of ARDS patients-especially hippocampus-becomes very vulnerable to develop secondary lung-mediated acute brain injury. A protective ventilator strategy could reduce or even minimize further systemic release of inflammatory mediators and thus maintain brain homeostasis. On the other hand, mechanical ventilation with low tidal volumes may lead to self-inflicted lung injury, hypercapnia and subsequent cerebral vasodilatation, increased cerebral blood flow, and intracranial hypertension. Therefore, by describing the pathophysiology of ARDS-associated acute brain injury we aim to highlight and discuss the possible influence of mechanical ventilation on ALI-associated acute brain injury.
Collapse
Affiliation(s)
- Mairi Ziaka
- Department of Internal Medicine, Thun General Hospital, Thun, Switzerland
| | - Aristomenis Exadaktylos
- Department of Emergency Medicine, Inselspital, University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
2
|
Perrone S, Lembo C, Gironi F, Petrolini C, Catalucci T, Corbo G, Buonocore G, Gitto E, Esposito SMR. Erythropoietin as a Neuroprotective Drug for Newborn Infants: Ten Years after the First Use. Antioxidants (Basel) 2022; 11:antiox11040652. [PMID: 35453337 PMCID: PMC9031072 DOI: 10.3390/antiox11040652] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/19/2022] [Accepted: 03/24/2022] [Indexed: 01/27/2023] Open
Abstract
Protective strategies against perinatal brain injury represent a major challenge for modern neonatology. Erythropoietin (Epo) enhances endogenous mechanisms of repair and angiogenesis. In order to analyse the newest evidence on the role of Epo in prematurity, hypoxic ischemic encephalopathy (HIE) and perinatal stroke, a critical review using 2020 PRISMA statement guidelines was conducted. This review uncovered 26 clinical trials examining the use of Epo for prematurity and brain injury-related outcomes. The effects of Epo on prematurity were analysed in 16 clinical trials. Erythropoietin was provided until 32–35 weeks of corrected postnatal age with a dosage between 500–3000 UI/kg/dose. Eight trials reported the Epo effects on HIE term newborn infants: Erythropoietin was administered in the first weeks of life, at different multiple doses between 250–2500 UI/kg/dose, as either an adjuvant therapy with hypothermia or a substitute for hypothermia. Two trials investigated Epo effects in perinatal stroke. Erythropoietin was administered at a dose of 1000 IU/kg for three days. No beneficial effect in improving morbidity was observed after Epo administration in perinatal stroke. A positive effect on neurodevelopmental outcome seems to occur when Epo is used as an adjuvant therapy with hypothermia in the HIE newborns. Administration of Epo in preterm infants still presents inconsistencies with regard to neurodevelopmental outcome. Clinical trials show significant differences mainly in target population and intervention scheme. The identification of specific markers and their temporal expression at different time of recovery after hypoxia-ischemia in neonates might be implemented to optimize the therapeutic scheme after hypoxic-ischemic injury in the developing brain. Additional studies on tailored regimes, accounting for the risk stratification of brain damage in newborns, are required.
Collapse
Affiliation(s)
- Serafina Perrone
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (C.P.); (S.M.R.E.)
- Correspondence:
| | - Chiara Lembo
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (C.L.); (F.G.); (T.C.); (G.C.); (G.B.)
| | - Federica Gironi
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (C.L.); (F.G.); (T.C.); (G.C.); (G.B.)
| | - Chiara Petrolini
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (C.P.); (S.M.R.E.)
| | - Tiziana Catalucci
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (C.L.); (F.G.); (T.C.); (G.C.); (G.B.)
| | - Giulia Corbo
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (C.L.); (F.G.); (T.C.); (G.C.); (G.B.)
| | - Giuseppe Buonocore
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy; (C.L.); (F.G.); (T.C.); (G.C.); (G.B.)
| | - Eloisa Gitto
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
| | | |
Collapse
|
3
|
Sergio CM, Rolando CA. Erythropoietin regulates signaling pathways associated with neuroprotective events. Exp Brain Res 2022; 240:1303-1315. [PMID: 35234993 DOI: 10.1007/s00221-022-06331-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 02/09/2022] [Indexed: 11/25/2022]
Abstract
Erythropoietin is a cytokine that binds to the Erythropoietin receptor and regulates the formation of erythroid cells during erythropoiesis in the bone marrow. However, many other organs and tissues express Erythropoietin and its receptor, such as the Nervous System, which principally regulates tissue protection. In the Central Nervous System, Erythropoietin is principally expressed by astrocytes, while neurons mainly express Erythropoietin receptors. Moreover, Erythropoietin acts as a pleiotropic molecule with neuroprotective effects, and its mechanisms of signal transduction pathways are defined, and there is a growing interest in its therapeutic potential. This review focuses on the role of Erythropoietin and its relationship with HIF1, PI3/Akt, GSK3B, JAK/STAT, and MAPKs signaling pathways that leads to cell survival after injury in the Central Nervous System. Knowledge of these signaling systems comprehensively could better guide EPO treatment to restoring different SNC alterations mediated by different insults.
Collapse
Affiliation(s)
- Cornelio-Martínez Sergio
- Universidad del Valle de México, Escuela de Ciencias de la Salud, Campus Zapopan, Zapopan, Mexico
| | - Castañeda-Arellano Rolando
- Laboratorio de Farmacología, Centro de Investigación Multidisciplinario en Salud, Departamento de Ciencias Biomédicas, Centro Universitario de Tonalá, Universidad de Guadalajara, Av. Nuevo Periférico No. 555, 45425, Tonalá, Mexico.
| |
Collapse
|
4
|
Mitroshina EV, Savyuk MO, Ponimaskin E, Vedunova MV. Hypoxia-Inducible Factor (HIF) in Ischemic Stroke and Neurodegenerative Disease. Front Cell Dev Biol 2021; 9:703084. [PMID: 34395432 PMCID: PMC8355741 DOI: 10.3389/fcell.2021.703084] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/05/2021] [Indexed: 01/09/2023] Open
Abstract
Hypoxia is one of the most common pathological conditions, which can be induced by multiple events, including ischemic injury, trauma, inflammation, tumors, etc. The body's adaptation to hypoxia is a highly important phenomenon in both health and disease. Most cellular responses to hypoxia are associated with a family of transcription factors called hypoxia-inducible factors (HIFs), which induce the expression of a wide range of genes that help cells adapt to a hypoxic environment. Basic mechanisms of adaptation to hypoxia, and particularly HIF functions, have being extensively studied over recent decades, leading to the 2019 Nobel Prize in Physiology or Medicine. Based on their pivotal physiological importance, HIFs are attracting increasing attention as a new potential target for treating a large number of hypoxia-associated diseases. Most of the experimental work related to HIFs has focused on roles in the liver and kidney. However, increasing evidence clearly demonstrates that HIF-based responses represent an universal adaptation mechanism in all tissue types, including the central nervous system (CNS). In the CNS, HIFs are critically involved in the regulation of neurogenesis, nerve cell differentiation, and neuronal apoptosis. In this mini-review, we provide an overview of the complex role of HIF-1 in the adaptation of neurons and glia cells to hypoxia, with a focus on its potential involvement into various neuronal pathologies and on its possible role as a novel therapeutic target.
Collapse
Affiliation(s)
- Elena V. Mitroshina
- Department of Neurotechnologe, Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhni Novgorod, Nizhny Novgorod, Russia
| | - Maria O. Savyuk
- Department of Neurotechnologe, Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhni Novgorod, Nizhny Novgorod, Russia
| | - Evgeni Ponimaskin
- Department of Neurotechnologe, Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhni Novgorod, Nizhny Novgorod, Russia
- Department of Cellular Neurophysiology, Hannover Medical School, Hanover, Germany
| | - Maria V. Vedunova
- Department of Neurotechnologe, Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhni Novgorod, Nizhny Novgorod, Russia
| |
Collapse
|
5
|
Ma X, Shi Y. Whether erythropoietin can be a neuroprotective agent against premature brain injury: cellular mechanisms and clinical efficacy. Curr Neuropharmacol 2021; 20:611-629. [PMID: 34030616 DOI: 10.2174/1570159x19666210524154519] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 04/27/2021] [Accepted: 05/08/2021] [Indexed: 11/22/2022] Open
Abstract
Preterm infants are at high risk of brain injury. With more understanding of the preterm brain injury's pathogenesis, neuroscientists are looking for more effective methods to prevent and treat it, among which erythropoietin (Epo) is considered as a prime candidate. This review tries to clarify the possible mechanisms of Epo in preterm neuroprotection and summarize updated evidence considering Epo as a pharmacological neuroprotective strategy in animal models and clinical trials. To date, various animal models have validated that Epo is an anti-apoptotic, anti-inflammatory, anti-oxidant, anti-excitotoxic, neurogenetic, erythropoietic, angiogenetic, and neurotrophic agent, thus preventing preterm brain injury. However, although the scientific rationale and preclinical data for Epo's neuroprotective effect are promising, when translated to bedside, the results vary in different studies, especially in its long-term efficacy. Based on existing evidence, it is still too early to recommend Epo as the standard treatment for preterm brain injury.
Collapse
Affiliation(s)
- Xueling Ma
- Department of Neonatology, Children's Hospital of Chongqing Medical University; National Clinical Research Center for Child Health and Disorders; Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing 400014, China
| | - Yuan Shi
- Department of Neonatology, Children's Hospital of Chongqing Medical University; National Clinical Research Center for Child Health and Disorders; Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing 400014, China
| |
Collapse
|
6
|
Lee J, Walter MF, Korach KS, Noguchi CT. Erythropoietin reduces fat mass in female mice lacking estrogen receptor alpha. Mol Metab 2020; 45:101142. [PMID: 33309599 PMCID: PMC7809438 DOI: 10.1016/j.molmet.2020.101142] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/23/2020] [Accepted: 12/07/2020] [Indexed: 12/17/2022] Open
Abstract
Objective Erythropoietin (EPO), the cytokine required for erythropoiesis, contributes to metabolic regulation of fat mass and glycemic control. EPO treatment in mice on high-fat diets (HFD) improved glucose tolerance and decreased body weight gain via reduced fat mass in males and ovariectomized females. The decreased fat accumulation with EPO treatment during HFD in ovariectomized females was abrogated with estradiol supplementation, providing evidence for estrogen-related gender-specific EPO action in metabolic regulation. In this study, we examined the cross-talk between estrogen mediated through estrogen receptor α (ERα) and EPO for the regulation of glucose metabolism and fat mass accumulation. Methods Wild-type (WT) mice and mouse models with ERα knockout (ERα−/−) and targeted deletion of ERα in adipose tissue (ERαadipoKO) were used to examine EPO treatment during high-fat diet feeding and after diet-induced obesity. Results ERα−/− mice on HFD exhibited increased fat mass and glucose intolerance. EPO treatment on HFD reduced fat accumulation in male WT and ERα−/− mice and female ERα−/− mice but not female WT mice. EPO reduced HFD increase in adipocyte size in WT mice but not in mice with deletion of ERα independent of EPO-stimulated reduction in fat mass. EPO treatment also improved glucose and insulin tolerance significantly greater in female ERα−/− mice and female ERαadipoKO compared with WT controls. Increased metabolic activity by EPO was associated with browning of white adipocytes as shown by reductions in white fat-associated genes and induction of brown fat-specific uncoupling protein 1 (UCP1). Conclusions This study clearly identified the role of estrogen signaling in modifying EPO regulation of glucose metabolism and the sex-differential EPO effect on fat mass regulation. Cross-talk between EPO and estrogen was implicated for metabolic homeostasis and regulation of body mass in female mice. Erythropoietin regulates fat mass in male but not female mice on high-fat diets. Female estrogen receptor alpha deletion restores erythropoietin fat mass regulation. Estrogen receptor alpha deletion increases erythropoietin regulation of glucose tolerance. Erythropoietin reduced white fat-associated genes and increased uncoupling protein 1. Erythropoietin and estrogen cross-talk is implicated for metabolic homeostasis.
Collapse
Affiliation(s)
- Jeeyoung Lee
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mary F Walter
- Clinical Laboratory Core, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kenneth S Korach
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Constance Tom Noguchi
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
7
|
Insights into Potential Targets for Therapeutic Intervention in Epilepsy. Int J Mol Sci 2020; 21:ijms21228573. [PMID: 33202963 PMCID: PMC7697405 DOI: 10.3390/ijms21228573] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/04/2020] [Accepted: 11/11/2020] [Indexed: 02/06/2023] Open
Abstract
Epilepsy is a chronic brain disease that affects approximately 65 million people worldwide. However, despite the continuous development of antiepileptic drugs, over 30% patients with epilepsy progress to drug-resistant epilepsy. For this reason, it is a high priority objective in preclinical research to find novel therapeutic targets and to develop effective drugs that prevent or reverse the molecular mechanisms underlying epilepsy progression. Among these potential therapeutic targets, we highlight currently available information involving signaling pathways (Wnt/β-catenin, Mammalian Target of Rapamycin (mTOR) signaling and zinc signaling), enzymes (carbonic anhydrase), proteins (erythropoietin, copine 6 and complement system), channels (Transient Receptor Potential Vanilloid Type 1 (TRPV1) channel) and receptors (galanin and melatonin receptors). All of them have demonstrated a certain degree of efficacy not only in controlling seizures but also in displaying neuroprotective activity and in modifying the progression of epilepsy. Although some research with these specific targets has been done in relation with epilepsy, they have not been fully explored as potential therapeutic targets that could help address the unsolved issue of drug-resistant epilepsy and develop new antiseizure therapies for the treatment of epilepsy.
Collapse
|
8
|
Zhang J, Luo X, Huang C, Pei Z, Xiao H, Luo X, Huang S, Chang Y. Erythropoietin prevents LPS-induced preterm birth and increases offspring survival. Am J Reprod Immunol 2020; 84:e13283. [PMID: 32506750 PMCID: PMC7507205 DOI: 10.1111/aji.13283] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 05/13/2020] [Accepted: 06/01/2020] [Indexed: 12/17/2022] Open
Abstract
PROBLEM Preterm delivery is the leading cause of neonatal mortality and contributes to delayed physical and cognitive development in children. At present, there is no efficient therapy to prevent preterm labor. A large body of evidence suggests that infections might play a significant and potentially preventable cause of premature birth. This work assessed the effects of erythropoietin (EPO) in a murine model of inflammation-associated preterm delivery, which mimics central features of preterm infections in humans. METHOD OF STUDY BALB/c mice were injected i.p. with 20 000 IU/kg EPO or normal saline twice on gestational day (GD) 15, with a 3 hours time interval between injections. An hour after the first EPO or normal saline injection, all mice received two injections of 50 μg/kg LPS, also given 3 hours apart. RESULTS EPO significantly prevented preterm labor and increased offspring survival in an LPS induced preterm delivery model. EPO prevented LPS-induced leukocyte infiltration into the placenta. Moreover, EPO inhibited the expression of pro-inflammatory cytokines, interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumour necrosis factor-α (TNF-α) in maternal serum and amniotic fluid. EPO also prevented LPS-induced increase in placental prostaglandin (PG)E2 and uterine inducible nitric oxide synthase (iNOS) production, while decreasing nuclear factor kappa-B (NF-κβ) activity in the myometrium. EPO also increased the gene expression of placental programmed cell death ligand 1 (PD-L1) in LPS-treated mice. CONCLUSIONS Our results suggest that EPO could be a potential novel therapeutic strategy to tackle infection-related preterm labor.
Collapse
Affiliation(s)
- Jie Zhang
- Department of RehabilitationGuangdong Women and Children HospitalGuangzhouChina
| | - Xianqiong Luo
- Department of PediatricsGuangdong Women and Children HospitalGuangzhouChina
| | - Caicai Huang
- Department of ObstetricsGuangdong Women and Children HospitalGuangzhouChina
| | - Zheng Pei
- Department of RehabilitationGuangdong Women and Children HospitalGuangzhouChina
| | - Huimei Xiao
- Department of RehabilitationGuangdong Women and Children HospitalGuangzhouChina
| | - Xingang Luo
- Department of RehabilitationGuangdong Women and Children HospitalGuangzhouChina
| | - Shuangmiao Huang
- Department of RehabilitationGuangdong Women and Children HospitalGuangzhouChina
| | - Yanqun Chang
- Department of RehabilitationGuangdong Women and Children HospitalGuangzhouChina
| |
Collapse
|
9
|
Yasuoka Y, Fukuyama T, Izumi Y, Nakayama Y, Inoue H, Yanagita K, Oshima T, Yamazaki T, Uematsu T, Kobayashi N, Shimada Y, Nagaba Y, Mukoyama M, Yamashita T, Sato Y, Sands JM, Kawahara K, Nonoguchi H. Erythropoietin production by the kidney and the liver in response to severe hypoxia evaluated by Western blotting with deglycosylation. Physiol Rep 2020; 8:e14485. [PMID: 32592328 PMCID: PMC7319944 DOI: 10.14814/phy2.14485] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 05/11/2020] [Accepted: 05/19/2020] [Indexed: 01/17/2023] Open
Abstract
The detection of erythropoietin (Epo) protein by Western blotting has required pre‐purification of the sample. We developed a new Western blot method to detect plasma and urinary Epo using deglycosylation. Epo in urine and tissue, and erythropoiesis‐stimulating agents (ESAs) in urine were directly detected by our Western blotting. Plasma Epo and ESAs were not detected by direct application but were detected by our Western blotting after deglycosylation. The broad bands of Epo and ESAs were shifted to 22 kDa by deglycosylation except for PEG‐bound epoetin β pegol. The 22 kDa band from an anemic patient's urine was confirmed by Liquid Chromatography/Mass Spectrometry (LC/MS) to contain human Epo. Severe hypoxia (7% O2, 4 hr) caused a 400‐fold increase in deglycosylated Epo expression in rat kidneys, which is consistent with the increases in both Epo gene expression and plasma Epo concentration. Immunohistochemistry showed Epo expression in nephrons but not in interstitial cells under control conditions, and hypoxia increased Epo expression in interstitial cells but not in tubules. These data show that intrinsic Epo and all ESAs can be detected by Western blot either directly in urine or after deglycosylation in blood, and that the kidney but not the liver is the main site of Epo production in control and severe hypoxia. Our method will make the tests for Epo doping and detection easy.
Collapse
Affiliation(s)
- Yukiko Yasuoka
- Department of Physiology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Takashi Fukuyama
- Division of Biomedical Research, Kitasato University Medical Center, Kitamoto, Japan
| | - Yuichiro Izumi
- Department of Nephrology, Kumamoto University Graduate School of Medicine, Kumamoto, Japan
| | - Yushi Nakayama
- Department of Nephrology, Kumamoto University Graduate School of Medicine, Kumamoto, Japan
| | - Hideki Inoue
- Department of Nephrology, Kumamoto University Graduate School of Medicine, Kumamoto, Japan
| | - Kengo Yanagita
- Department of Molecular Diagnostics, Kitasato University School of Allied Health Sciences, Sagamihara, Japan
| | - Tomomi Oshima
- Department of Physiology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Taiga Yamazaki
- Division of Biomedical Research, Kitasato University Medical Center, Kitamoto, Japan
| | - Takayuki Uematsu
- Division of Biomedical Research, Kitasato University Medical Center, Kitamoto, Japan
| | - Noritada Kobayashi
- Division of Biomedical Research, Kitasato University Medical Center, Kitamoto, Japan
| | - Yoshitaka Shimada
- Division of Internal Medicine, Kitasato University Medical Center, Kitamoto, Japan
| | - Yasushi Nagaba
- Division of Internal Medicine, Kitasato University Medical Center, Kitamoto, Japan
| | - Masashi Mukoyama
- Department of Nephrology, Kumamoto University Graduate School of Medicine, Kumamoto, Japan
| | - Tetsuro Yamashita
- Department of Biological Chemistry and Food Sciences, Faculty of Agriculture, Iwate University, Morioka, Japan
| | - Yuichi Sato
- Department of Molecular Diagnostics, Kitasato University School of Allied Health Sciences, Sagamihara, Japan
| | - Jeff M Sands
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Katsumasa Kawahara
- Department of Physiology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Hiroshi Nonoguchi
- Division of Internal Medicine, Kitasato University Medical Center, Kitamoto, Japan
| |
Collapse
|
10
|
Li J, Tao T, Xu J, Liu Z, Zou Z, Jin M. HIF‑1α attenuates neuronal apoptosis by upregulating EPO expression following cerebral ischemia‑reperfusion injury in a rat MCAO model. Int J Mol Med 2020; 45:1027-1036. [PMID: 32124933 PMCID: PMC7053873 DOI: 10.3892/ijmm.2020.4480] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 12/30/2019] [Indexed: 01/16/2023] Open
Abstract
Hypoxia-inducible factor-1α (HIF-1α) is a key transcriptional factor in response to hypoxia and is involved in ischemic stroke. In the present study, the potential for HIF-1α to inhibit neuronal apoptosis through upregulating erythropoietin (EPO) was investigated in a transient middle cerebral artery occlusion (tMCAO) rat stroke model. For this purpose, a recombinant adenovirus expressing HIF-1α was engineered (Ad-HIF-1α). Control adenovirus (Ad group), Ad-HIF-1α (Ad-HIF-1α group) or Ad-HIF-1α in addition to erythropoietin mimetic peptide-9 (EMP9), an EPO-receptor (-R) antagonist (Ad-HIF-1α+EMP9 group), were used for an intracranial injection into rat ischemic penumbra 1 h following MCAO. All rats demonstrated functional improvement following tMCAO, while the improvement rate was faster in rats treated by Ad-HIF-1α compared with all other groups. The EPO-R inhibitor partially reversed the benefits of Ad-HIF-1α. Apoptosis induced by tMCAO was significantly inhibited by Ad-HIF-1α (P<0.05). The expression of HIF-1α, evaluated by immunohistochemistry either in neurons or astrocytes, was upregulated by Ad-HIF-1α. Both EPO mRNA and protein expression were increased by Ad-HIF-1α, however, there was no significant change of EPO-R either on an mRNA level or protein level. Furthermore, EMP9 did not change the EPO expression which was upregulated by Ad-HIF-1α. Activated caspase 3 in neurons was suppressed by Ad-HIF-1α. Activated caspase 3 downregulated by HIF-1α was partially blocked by EMP9. Altogether, the present data demonstrated that HIF-1α attenuates neuronal apoptosis partially through upregulating EPO following cerebral ischemia in rat. Thus, upregulating HIF-1α subsequent to a stroke may be a potential treatment for ischemic stroke.
Collapse
Affiliation(s)
- Jun Li
- Department of Rehabilitation Medicine, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Tao Tao
- Department of Rehabilitation Medicine, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Jian Xu
- Department of Neurology, The Affiliated Hospital Guizhou Medical University, Guiyang, Guizhou 550001, P.R. China
| | - Zhi Liu
- Department of Pharmacy, The Affiliated Hospital Guizhou Medical University, Guiyang, Guizhou 550001, P.R. China
| | - Zhehua Zou
- Department of General Practice, The First Hospital of Qinhuangdao, Qinhuangdao, Hebei 066000, P.R. China
| | - Minglu Jin
- Department of Neurology, Qijiang Hospital of The First Affiliated Hospital of Chongqing Medical University, Chongqing 404100, P.R. China
| |
Collapse
|
11
|
Davidović S, Babić N, Jovanović S, Barišić S, Grković D, Miljković A. Serum erythropoietin concentration and its correlation with stage of diabetic retinopathy. BMC Ophthalmol 2019; 19:227. [PMID: 31727007 PMCID: PMC6857223 DOI: 10.1186/s12886-019-1240-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 11/06/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Erythropoietin (EPO) is one of the systemic angiogenic factors, and its role in ocular angiogenesis and in diabetic retinopathy (DR) is not yet fully understood. The latest research data reveal a possible correlation of higher erythropoietin concentrations in the blood and in the eye with the development of more advanced stages of DR. The main aim of this work was to examine the possible influence of serum concentrations of erythropoietin on the development of diabetic retinopathy in patients with diabetes mellitus type 2. METHODS The research involved 90 patients examined at the University Eye Clinic of the Clinical Center of Vojvodina, Novi Sad, Serbia. The first group comprised 60 patients with diabetes mellitus lasting for 10 years or more, with diabetic retinopathy. The second, control group consisted of 30 healthy individuals. In the first group of 60 patients with diabetes, 30 of them had non-proliferative diabetic retinopathy (NPDR), and 30 had proliferative diabetic retinopathy (PDR). Laboratory EPO serum levels were determined, and they were correlated to the stage of DR. Concentration of EPO was assessed by ELISA method. RESULTS The highest average concentration of EPO in serum (9.95 mIU/ml) was determined in the group of people with diabetes with PDR. The lowest average concentration of EPO in the serum (6.90 mIU/ml) was found in the control group. The average concentration of EPO in serum in the group of patients with diabetes with NPDR was 7.00 mIU/ml. The EPO concentration in serum was elevated in the group of PDR, and it was directly proportional to the level of the clinical stadium of PDR, being significantly higher in the moderate and severe subgroup of PDR comparing to the control healthy subjects, NPDR and mild PDR (p = 0.007). CONCLUSIONS Significantly elevated serum concentration of EPO in the advanced stages of DR, and positive correlation between EPO serum concentration and clinical stages of PDR, suggest that erythropoietin represents an important growth factor from blood, which plays a significant role in retinal ischemia and angiogenesis in diabetic retinopathy, especially in the proliferative stage of this disease.
Collapse
Affiliation(s)
- Sofija Davidović
- Faculty of Medicine, Department of Ophthalmology, University of Novi Sad, Novi Sad, 21000, Serbia.
- Eye Clinic, Clinical Center of Vojvodina, Hajduk Veljkova 1-9, Novi Sad, 21000, Serbia.
| | - Nikola Babić
- Faculty of Medicine, Department of Ophthalmology, University of Novi Sad, Novi Sad, 21000, Serbia
- Eye Clinic, Clinical Center of Vojvodina, Hajduk Veljkova 1-9, Novi Sad, 21000, Serbia
| | - Sandra Jovanović
- Faculty of Medicine, Department of Ophthalmology, University of Novi Sad, Novi Sad, 21000, Serbia
- Eye Clinic, Clinical Center of Vojvodina, Hajduk Veljkova 1-9, Novi Sad, 21000, Serbia
| | - Sava Barišić
- Eye Clinic, Clinical Center of Vojvodina, Hajduk Veljkova 1-9, Novi Sad, 21000, Serbia
| | - Desanka Grković
- Faculty of Medicine, Department of Ophthalmology, University of Novi Sad, Novi Sad, 21000, Serbia
- Eye Clinic, Clinical Center of Vojvodina, Hajduk Veljkova 1-9, Novi Sad, 21000, Serbia
| | - Aleksandar Miljković
- Faculty of Medicine, Department of Ophthalmology, University of Novi Sad, Novi Sad, 21000, Serbia
- Eye Clinic, Clinical Center of Vojvodina, Hajduk Veljkova 1-9, Novi Sad, 21000, Serbia
| |
Collapse
|
12
|
Asano R, Asai-Sato M, Matsukuma S, Mizushima T, Taguri M, Yoshihara M, Inada M, Fukui A, Suzuki Y, Miyagi Y, Miyagi E. Expression of erythropoietin messenger ribonucleic acid in wild-type MED12 uterine leiomyomas under estrogenic influence: new insights into related growth disparities. Fertil Steril 2019; 111:178-185. [DOI: 10.1016/j.fertnstert.2018.09.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 09/25/2018] [Accepted: 09/25/2018] [Indexed: 12/28/2022]
|
13
|
Rostami S, Emami-Aleagha MS, Ghasemi-Kasman M, Allameh A. Cross-talks between the kidneys and the central nervous system in multiple sclerosis. CASPIAN JOURNAL OF INTERNAL MEDICINE 2018; 9:206-210. [PMID: 30197763 PMCID: PMC6121345 DOI: 10.22088/cjim.9.3.206] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease, which is considered as a common autoimmune disorder in young adults. A growing number of evidences indicated that the impairment in non-neural tissues plays a significant role in pathology of MS disease. There are bidirectional relationship, metabolic activities and functional similarity between central nervous system (CNS) and kidneys which suggest that kidney tissue may exert remarkable effects on some aspects of MS disorder and CNS impairment in these patients compels the kidney to respond to central inflammation. Recently, it has been well documented that hormonal secretion possesses the important role on CNS abnormalities. In this regard, due to the functional similarity and significant hormonal and non-hormonal relationship between CNS and kidneys, we hypothesized that kidneys exert significant effect on initiation, progression or amelioration of MS disease which might be regarded as potential therapeutic approach in the treatment of MS patients in the future.
Collapse
Affiliation(s)
- Sahar Rostami
- Department of Clinical Biochemistry, Faculty of Medicine, Tarbiat Modarres University, Tehran, Iran
| | | | - Maryam Ghasemi-Kasman
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Abdolamir Allameh
- Department of Clinical Biochemistry, Faculty of Medicine, Tarbiat Modarres University, Tehran, Iran
| |
Collapse
|
14
|
Pediatric brain repair from endogenous neural stem cells of the subventricular zone. Pediatr Res 2018; 83:385-396. [PMID: 29028220 DOI: 10.1038/pr.2017.261] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 10/05/2017] [Indexed: 12/22/2022]
Abstract
There is great interest in the regenerative potential of the neural stem cells and progenitors that populate the germinal zones of the immature brain. Studies using animal models of pediatric brain injuries have provided a clearer understanding of the responses of these progenitors to injury. In this review, we have compared and contrasted the responses of the endogenous neural stem cells and progenitors of the subventricular zone in animal models of neonatal cerebral hypoxia-ischemia, neonatal stroke, congenital cardiac disease, and pediatric traumatic brain injury. We have reviewed the dynamic shifts that occur within this germinal zone with injury as well as changes in known signaling molecules that affect these progenitors. Importantly, we have summarized data on the extent to which cell replacement occurs in response to each of these injuries, opportunities available, and obstacles that will need to be overcome to improve neurological outcomes in survivors.
Collapse
|
15
|
Fischer HS, Reibel NJ, Bührer C, Dame C. Prophylactic Early Erythropoietin for Neuroprotection in Preterm Infants: A Meta-analysis. Pediatrics 2017; 139:peds.2016-4317. [PMID: 28557760 DOI: 10.1542/peds.2016-4317] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/09/2017] [Indexed: 11/24/2022] Open
Abstract
CONTEXT Recombinant human erythropoietin (rhEPO) is a promising pharmacological agent for neuroprotection in neonates. OBJECTIVE To investigate whether prophylactic rhEPO administration in very preterm infants improves neurodevelopmental outcomes in a meta-analysis of randomized controlled trials (RCTs). DATA SOURCES Medline, Embase, and the Cochrane Central Register of Controlled Trials were searched in December 2016 and complemented by other sources. STUDY SELECTION RCTs investigating the use of rhEPO in preterm infants versus a control group were selected if they were published in a peer-reviewed journal and reported neurodevelopmental outcomes at 18 to 24 months' corrected age. DATA EXTRACTION Data extraction and analysis followed the standard methods of the Cochrane Neonatal Review Group. The primary outcome was the number of infants with a Mental Developmental Index (MDI) <70 on the Bayley Scales of Infant Development. Secondary outcomes included a Psychomotor Development Index <70, cerebral palsy, visual impairment, and hearing impairment. RESULTS Four RCTs, comprising 1133 infants, were included in the meta-analysis. Prophylactic rhEPO administration reduced the incidence of children with an MDI <70, with an odds ratio (95% confidence interval) of 0.51 (0.31-0.81), P < .005. The number needed to treat was 14. There was no statistically significant effect on any secondary outcome. CONCLUSIONS Prophylactic rhEPO improved the cognitive development of very preterm infants, as assessed by the MDI at a corrected age of 18 to 24 months, without affecting other neurodevelopmental outcomes. Current and future RCTs should investigate optimal dosing and timing of prophylactic rhEPO and plan for long-term neurodevelopmental follow-up.
Collapse
Affiliation(s)
- Hendrik S Fischer
- Department of Neonatology, Charité University Medical Center, Berlin, Germany
| | - Nora J Reibel
- Department of Neonatology, Charité University Medical Center, Berlin, Germany
| | - Christoph Bührer
- Department of Neonatology, Charité University Medical Center, Berlin, Germany
| | - Christof Dame
- Department of Neonatology, Charité University Medical Center, Berlin, Germany
| |
Collapse
|
16
|
Zhang Y, Rogers HM, Zhang X, Noguchi CT. Sex difference in mouse metabolic response to erythropoietin. FASEB J 2017; 31:2661-2673. [PMID: 28283542 DOI: 10.1096/fj.201601223rrr] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 02/21/2017] [Indexed: 11/11/2022]
Abstract
Erythropoietin (EPO) is the cytokine that regulates red blood cell production. Less understood is the nonerythroid action of EPO, including metabolic regulation of fat accumulation and glucose homeostasis. Although EPO treatment increased hematocrit and improved glucose tolerance in male and female mice, we observed a gender difference in EPO effects in weight control. EPO treatment reduced diet-induced weight gain from 9.6 ± 1.5 to 4.2 ± 1.4 g in male mice (P < 0.001), while the weight gain in female mice was similar (4.7 ± 2.0 g with PBS treatment and 3.3 ± 2.1 g with EPO treatment). EPO treatment also reduced weight gain in ovariectomized female mice, while the effect was abrogated with estradiol supplementation, suggesting that the sex-differential response to EPO was associated with estrogen. Furthermore, mice with targeted deletion of EPO receptor in white adipose tissue exhibited sex-differential phenotype in weight control and glucose sensitivity, and EPO receptor gene expression was reduced in wild-type female mice, suggesting that white adipose tissue plays an integral role in mediating the metabolic effects of EPO. Our data provide evidence for a sex-differential response to EPO in weight control in mice and underscore the potential for gender specific EPO action beyond erythropoiesis.-Zhang, Y., Rogers, H. M., Zhang, X., Noguchi, C. T. Sex difference in mouse metabolic response to erythropoietin.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Heather M Rogers
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Xiaojie Zhang
- Laboratory of Animal Sciences Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Constance Tom Noguchi
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA;
| |
Collapse
|
17
|
Zhou Y, Zhao X, Tang Y, Zhu L, Jiang J, Lu X. Erythropoietin Inhibits the Increase of Pulmonary Labile Zinc and the Expression of Inflammatory Mediators Following Subarachnoid Hemorrhage in Rats. Neurocrit Care 2015; 24:472-80. [DOI: 10.1007/s12028-015-0219-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
18
|
Fauchère JC, Koller BM, Tschopp A, Dame C, Ruegger C, Bucher HU. Safety of Early High-Dose Recombinant Erythropoietin for Neuroprotection in Very Preterm Infants. J Pediatr 2015; 167:52-7.e1-3. [PMID: 25863661 DOI: 10.1016/j.jpeds.2015.02.052] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 01/20/2015] [Accepted: 02/18/2015] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To investigate the safety and short term outcome of high dose recombinant human erythropoietin (rhEpo) given shortly after birth and subsequently over the first 2 days for neuroprotection to very preterm infants. STUDY DESIGN Randomized, double masked phase II trial. Preterm infants (gestational age 26 0/7-31 6/7 weeks) were given rhEpo (nt = 229; 3000 U/kg body weight) or NaCl 0.9% (nc = 214) intravenously at 3, 12-18, and 36-42 hours after birth. RESULTS There were no relevant differences between the groups for short-term outcomes such as mortality, retinopathy of prematurity, intraventricular hemorrhage, sepsis, necrotizing enterocolitis, and bronchopulmonary dysplasia. At day 7-10, we found significantly higher hematocrit values, reticulocyte, and white blood cell counts, and a lower platelet count in the rhEpo group. CONCLUSIONS Early high-dose rhEpo administration to very premature infants is safe and causes no excess in mortality or major adverse events. TRIAL REGISTRATION ClinicalTrials.gov: NCT00413946.
Collapse
Affiliation(s)
- Jean-Claude Fauchère
- Division of Neonatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| | - Brigitte M Koller
- Division of Neonatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Alois Tschopp
- Division of Biostatistics, Institute of Social and Preventive Medicine, University of Zurich, Zurich, Switzerland
| | - Christof Dame
- Department of Neonatology, Charité-Universitätsmedizin, Berlin, Germany
| | - Christoph Ruegger
- Division of Neonatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Hans Ulrich Bucher
- Division of Neonatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | | |
Collapse
|
19
|
Erythropoietin Attenuates the Apoptosis of Adult Neurons After Brachial Plexus Root Avulsion by Downregulating JNK Phosphorylation and c-Jun Expression and Inhibiting c-PARP Cleavage. J Mol Neurosci 2015; 56:917-925. [PMID: 25877688 DOI: 10.1007/s12031-015-0543-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 03/09/2015] [Indexed: 01/05/2023]
Abstract
In the present study, the effects of erythropoietin (EPO) on preventing adult neurons from apoptosis (introduced by brachial plexus avulsion) were examined, and the mechanism was analyzed. Fifty injury rat models were established in this study by using micro-hemostat forceps to pull out brachial plexus root from the intervertebral foramen in supine position. These models were divided into EPO group (avulsion + 1000 U/kg subcutaneously on alternate days) and control group (avulsion + normal saline). C5-T1 spinal cord was harvested at days 1, 2, 4, 7, and 14. Compared with the control group, the apoptosis of spinal motoneurons was significantly decreased on days 4 and 7 in the EPO group, which was also approved by TUNEL examination results. The detection of p-JNK and expression of c-Jun and cleavage of cleaved PARP (c-PARP) were also examined by immunohistochemistry and were increased immediately at day 1, and peaked at day 2, day 2, and day 4 in control group, respectively. However, the amounts were decreased and delayed by EPO treatment significantly at the same time points. In conclusion, the apoptosis of adult spinal motorneurons was associated with JNK phosphorylation, c-Jun expression, and caspase activity, and EPO-mediated neuronal protective effect is proved by downregulating the JNK phosphorylation and c-Jun expression and inhibiting of c-PARP cleavage.
Collapse
|
20
|
Ballot O, Joseph V, Soliz J. Endogenous brain erythropoietin is a potent sex-specific respiratory stimulant in adult and newborn mice. J Appl Physiol (1985) 2015; 118:1386-95. [PMID: 25792712 DOI: 10.1152/japplphysiol.00143.2015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 03/17/2015] [Indexed: 12/31/2022] Open
Abstract
We tested the hypothesis that endogenous brain Epo is a respiratory stimulant. Adult (3 mo) and newborn (10 days) male and female mice received an intracisternal (cisterna magna) injection of soluble Epo receptor (sEpoR; competes with EpoR to bind Epo; 50 μg/ml) or vehicle (0.1% BSA in PBS). Twenty-four hours after injection, we used whole body plethysmography to record minute ventilation (V̇e) tidal volume (VT), respiratory frequency (fR), O2 consumption (V̇o2), and CO2 production (V̇co2) under normoxia and progressive exposure to hypoxia (12-10-6% O2; 10 min each). In adult male and female mice sEpoR decreased normoxic V̇e (-25%), due to a decrease of VT in males and fR in females. Moreover, sEpoR injection decreased the ventilatory response to 12% O2, assessed as V̇e/V̇o2 or V̇e/V̇co2, in male but not in female mice. In newborn male and female mice sEpoR decreased V̇e (-37% in males, -59% in females) and VT (-38% in males, -47% in females) in normoxia and fR in females. During hypoxia, sEpoR decreased V̇e/V̇o2 and V̇e/V̇co2 in mice of both sexes. Upon extreme hypoxia (6% O2), the newborn mice treated with sEpoR showed respiratory depression, signs of asphyxia (gasping) and a high mortality rate in males and females. We concluded that endogenous brain Epo is a potent respiratory stimulant under normoxia and hypoxia in adult and newborn mice. Because sex-specific effects are different in newborn male and female, sex steroids secreted at different ages mice appear to modulate the effects of Epo on respiratory regulation in normoxia and in response to hypoxia.
Collapse
Affiliation(s)
- Orlane Ballot
- Centre de Recherche du CHU de Québec, Pavillon St François d'Assise, Département de Pédiatrie, Faculté de Médecine, Université Laval, Québec, Quebec, Canada
| | - Vincent Joseph
- Centre de Recherche du CHU de Québec, Pavillon St François d'Assise, Département de Pédiatrie, Faculté de Médecine, Université Laval, Québec, Quebec, Canada
| | - Jorge Soliz
- Centre de Recherche du CHU de Québec, Pavillon St François d'Assise, Département de Pédiatrie, Faculté de Médecine, Université Laval, Québec, Quebec, Canada
| |
Collapse
|
21
|
Zhang L, Wang H, Wang T, Jiang N, Yu P, Chong Y, Fu F. Ferulic acid ameliorates nerve injury induced by cerebral ischemia in rats. Exp Ther Med 2014; 9:972-976. [PMID: 25667662 PMCID: PMC4316951 DOI: 10.3892/etm.2014.2157] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 10/23/2014] [Indexed: 01/04/2023] Open
Abstract
This study was designed to investigate the protective effect of ferulic acid (FA) on nerve injury induced by cerebral ischemia. Focal cerebral ischemia was induced by occlusion of the right middle cerebral artery and reperfusion 90 min later in male Sprague-Dawley rats. Daily treatment of the rats with FA was initiated 30 min after the surgery, and was continued for 7 days. The efficacy of FA against nerve injury was assessed by neurological deficit scores as well as pathohistological observation. The expression levels in the brain and level in the peripheral blood of erythropoietin (EPO) and granulocyte colony-stimulating factor (G-CSF) were analyzed by immunohistochemistry and enzyme-linked immunosorbent assay (ELISA), respectively. The results showed that FA attenuated nerve injury of the hippocampus, significantly ameliorated neurological deficits, and increased EPO but not G-CSF expression in the hippocampus and the peripheral blood of ischemic rats. The findings indicate that FA has certain protective effects on the nerve injury of cerebral ischemia, and suggest that promoting EPO in the brain and peripheral blood may be one of the neuroprotective mechanisms of FA.
Collapse
Affiliation(s)
- Leiming Zhang
- Department of Pharmacology, School of Pharmacy, Yantai University, Yantai, Shandong 264005, P.R. China
| | - Hongsheng Wang
- Department of Pharmacology, School of Pharmacy, Yantai University, Yantai, Shandong 264005, P.R. China
| | - Tian Wang
- Department of Pharmacology, School of Pharmacy, Yantai University, Yantai, Shandong 264005, P.R. China
| | - Na Jiang
- Department of Pharmacology, School of Pharmacy, Yantai University, Yantai, Shandong 264005, P.R. China
| | - Pengfei Yu
- Department of Pharmacology, School of Pharmacy, Yantai University, Yantai, Shandong 264005, P.R. China
| | - Yating Chong
- Department of Pharmacology, School of Pharmacy, Yantai University, Yantai, Shandong 264005, P.R. China
| | - Fenghua Fu
- Department of Pharmacology, School of Pharmacy, Yantai University, Yantai, Shandong 264005, P.R. China
| |
Collapse
|
22
|
Wang L, Di L, Noguchi CT. Erythropoietin, a novel versatile player regulating energy metabolism beyond the erythroid system. Int J Biol Sci 2014; 10:921-39. [PMID: 25170305 PMCID: PMC4147225 DOI: 10.7150/ijbs.9518] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Accepted: 06/04/2014] [Indexed: 12/12/2022] Open
Abstract
Erythropoietin (EPO), the required cytokine for promoting the proliferation and differentiation of erythroid cells to stimulate erythropoiesis, has been reported to act as a pleiotropic cytokine beyond hematopoietic system. The various activities of EPO are determined by the widespread distribution of its cell surface EPO receptor (EpoR) in multiple tissues including endothelial, neural, myoblasts, adipocytes and other cell types. EPO activity has been linked to angiogenesis, neuroprotection, cardioprotection, stress protection, anti-inflammation and especially the energy metabolism regulation that is recently revealed. The investigations of EPO activity in animals and the expression analysis of EpoR provide more insights on the potential of EPO in regulating energy metabolism and homeostasis. The findings of crosstalk between EPO and some important energy sensors and the regulation of EPO in the cellular respiration and mitochondrial function further provide molecular mechanisms for EPO activity in metabolic activity regulation. In this review, we will summarize the roles of EPO in energy metabolism regulation and the activity of EPO in tissues that are tightly associated with energy metabolism. We will also discuss the effects of EPO in regulating oxidative metabolism and mitochondrial function, the interactions between EPO and important energy regulation factors, and the protective role of EPO from stresses that are related to metabolism, providing a brief overview of previously less appreciated EPO biological function in energy metabolism and homeostasis.
Collapse
Affiliation(s)
- Li Wang
- 1. Faculty of Health Sciences, University of Macau, SAR of People's Republic of China
| | - Lijun Di
- 1. Faculty of Health Sciences, University of Macau, SAR of People's Republic of China
| | - Constance Tom Noguchi
- 2. Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, U.S.A
| |
Collapse
|
23
|
Abstract
Testicular torsion is a urological emergency most commonly seen in adolescence, involving a decrease in blood flow in the testis resulting from torsion of the spermatic cord that can result in gonad injury or even loss if not treated in time. Testicular ischaemia-reperfusion injury represents the principle pathophysiology of testicular torsion, with ischaemia caused by twisting of the spermatic cord, and reperfusion on its subsequent release. Many cellular and molecular mechanisms are involved in ischaemia-reperfusion injury following testicular torsion. Studies have investigated the use of pharmacological agents as supportive therapy to surgical repair in order to prevent the adverse effects of testicular torsion. Numerous substances have been proposed as important in the prevention of post-ischaemia-reperfusion testicular injury. A range of chemicals and drugs has been successfully tested in animal models for the purpose of mitigating the dangerous effects of ischaemia-reperfusion in testis torsion.
Collapse
|
24
|
Zhang Y, Wang L, Dey S, Alnaeeli M, Suresh S, Rogers H, Teng R, Noguchi CT. Erythropoietin action in stress response, tissue maintenance and metabolism. Int J Mol Sci 2014; 15:10296-333. [PMID: 24918289 PMCID: PMC4100153 DOI: 10.3390/ijms150610296] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 05/23/2014] [Accepted: 05/28/2014] [Indexed: 12/20/2022] Open
Abstract
Erythropoietin (EPO) regulation of red blood cell production and its induction at reduced oxygen tension provides for the important erythropoietic response to ischemic stress. The cloning and production of recombinant human EPO has led to its clinical use in patients with anemia for two and half decades and has facilitated studies of EPO action. Reports of animal and cell models of ischemic stress in vitro and injury suggest potential EPO benefit beyond red blood cell production including vascular endothelial response to increase nitric oxide production, which facilitates oxygen delivery to brain, heart and other non-hematopoietic tissues. This review discusses these and other reports of EPO action beyond red blood cell production, including EPO response affecting metabolism and obesity in animal models. Observations of EPO activity in cell and animal model systems, including mice with tissue specific deletion of EPO receptor (EpoR), suggest the potential for EPO response in metabolism and disease.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Li Wang
- Faculty of Health Sciences, University of Macau, Macau SAR, China.
| | - Soumyadeep Dey
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Mawadda Alnaeeli
- Department of Biological Sciences, Ohio University, Zanesville, OH 43701, USA.
| | - Sukanya Suresh
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Heather Rogers
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Ruifeng Teng
- Mouse Metabolism Core Laboratory, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Constance Tom Noguchi
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
25
|
Ratilal BO, Arroja MMC, Rocha JPF, Fernandes AMA, Barateiro AP, Brites DMTO, Pinto RMA, Sepodes BMN, Mota-Filipe HD. Neuroprotective effects of erythropoietin pretreatment in a rodent model of transient middle cerebral artery occlusion. J Neurosurg 2014; 121:55-62. [PMID: 24702327 DOI: 10.3171/2014.2.jns132197] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
UNLABELLED OBJECT.: There is an unmet clinical need to develop neuroprotective agents for neurosurgical and endovascular procedures that require transient cerebral artery occlusion. The aim in this study was to explore the effects of a single dose of recombinant human erythropoietin (rhEPO) before middle cerebral artery (MCA) occlusion in a focal cerebral ischemia/reperfusion model. METHODS Twenty-eight adult male Wistar rats were subjected to right MCA occlusion via the intraluminal thread technique for 60 minutes under continuous cortical perfusion monitoring by laser Doppler flowmetry. Rats were divided into 2 groups: control and treatment. In the treated group, rhEPO (1000 IU/kg intravenously) was administered 10 minutes before the onset of the MCA ischemia. At 24-hour reperfusion, animals were examined for neurological deficits, blood samples were collected, and animals were killed. The following parameters were evaluated: brain infarct volume, ipsilateral hemispheric edema, neuron-specific enolase plasma levels, parenchyma histological features (H & E staining), Fluoro-Jade-positive neurons, p-Akt and total Akt expression by Western blot analysis, and p-Akt-positive nuclei by immunohistochemical investigation. RESULTS Infarct volume and Fluoro-Jade staining of degenerating neurons in the infarct area did not vary between groups. The severity of neurological deficit (p < 0.001), amount of brain edema (78% reduction in treatment group, p < 0.001), and neuron-specific enolase plasma levels (p < 0.001) were reduced in the treatment group. Perivascular edema was histologically less marked in the treatment group. No variations in the expression or localization of p-Akt were seen. CONCLUSIONS Administration of rhEPO before the onset of 60-minute transient MCA ischemia protected the brain from this insult. It is unlikely that rhEPO pretreatment leads to direct neuronal antiapoptotic effects, as supported by the lack of Akt activation, and its benefits are most probably related to an indirect effect on brain edema as a consequence of blood-brain barrier preservation. Although research on EPO derivatives is increasing, rhEPO acts through distinct neuroprotective pathways and its clinical safety profile is well known. Clinically available rhEPO is a potential therapy for prevention of neuronal injury induced by transitory artery occlusion during neurovascular procedures.
Collapse
|
26
|
Kaneko N, Kako E, Sawamoto K. Enhancement of ventricular-subventricular zone-derived neurogenesis and oligodendrogenesis by erythropoietin and its derivatives. Front Cell Neurosci 2013; 7:235. [PMID: 24348331 PMCID: PMC3842008 DOI: 10.3389/fncel.2013.00235] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 11/08/2013] [Indexed: 12/17/2022] Open
Abstract
In the postnatal mammalian brain, stem cells in the ventricular-subventricular zone (V-SVZ) continuously generate neuronal and glial cells throughout life. Genetic labeling of cells of specific lineages have demonstrated that the V-SVZ is an important source of the neuroblasts and/or oligodendrocyte progenitor cells (OPCs) that migrate toward injured brain areas in response to several types of insult, including ischemia and demyelinating diseases. However, this spontaneous regeneration is insufficient for complete structural and functional restoration of the injured brain, so interventions to enhance these processes are sought for clinical applications. Erythropoietin (EPO), a clinically applied erythropoietic factor, is reported to have cytoprotective effects in various kinds of insult in the central nervous system. Moreover, recent studies suggest that EPO promotes the V-SVZ-derived neurogenesis and oligodendrogenesis. EPO increases the proliferation of progenitors in the V-SVZ and/or the migration and differentiation of their progenies in and around injured areas, depending on the dosage, timing, and duration of treatment, as well as the type of animal model used. On the other hand, EPO has undesirable side effects, including thrombotic complications. We recently demonstrated that a 2-week treatment with the EPO derivative asialo-EPO promotes the differentiation of V-SVZ-derived OPCs into myelin-forming mature oligodendrocytes in the injured white matter of neonatal mice without causing erythropoiesis. Here we present an overview of the multifaceted effects of EPO and its derivatives in the V-SVZ and discuss the possible applications of these molecules in regenerative medicine.
Collapse
Affiliation(s)
- Naoko Kaneko
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences Nagoya, Japan
| | - Eisuke Kako
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences Nagoya, Japan ; Department of Anesthesiology and Medical Crisis Management, Nagoya City University Graduate School of Medical Sciences Nagoya, Japan
| | - Kazunobu Sawamoto
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences Nagoya, Japan
| |
Collapse
|
27
|
MacNutt MJ, Rupert JL, Sheel AW. Haematological acclimation and re-acclimation to hypoxia in the mouse. Respir Physiol Neurobiol 2013; 189:153-61. [PMID: 23891710 DOI: 10.1016/j.resp.2013.07.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Revised: 06/20/2013] [Accepted: 07/18/2013] [Indexed: 11/19/2022]
Abstract
Haematological responses throughout 4 w of initial acclimation (IA) and three paradigms of re-acclimation (RA) to hypoxia (FI(O₂)) were examined in female mice. We hypothesised that (i) haematological responses would be increased during re-exposure, resulting in greater O₂-carrying capacity in RA compared to IA; and (ii) further improvements would occur when abbreviating the de-acclimation period to 1 w (RA↓DA) or extending the IA period to 8 w (RA↑IA). The serum [EPO] response was blunted in all RA groups compared to IA but the resulting reticulocyte response was similar in all experimental groups. The [Hb] response was the same in RA and RA↓DA as in IA but was blunted in RA↑IA due to a reduction in mean corpuscular Hb. The sensitivity of EPO-producing cells appears blunted but the sensitivity of erythroid precursors to EPO is enhanced by recent hypoxic exposure. Erythropoietic regulation is altered during RA in a manner that is dependent on the paradigm of initial exposure.
Collapse
Affiliation(s)
- Meaghan J MacNutt
- School of Kinesiology, University of British Columbia, Vancouver, BC, Canada
| | | | | |
Collapse
|
28
|
Abstract
The hematopoietic growth factor erythropoietin (Epo) circulates in plasma and controls the oxygen carrying capacity of the blood (Fisher. Exp Biol Med (Maywood) 228:1-14, 2003). Epo is produced primarily in the adult kidney and fetal liver and was originally believed to play a role restricted to stimulation of early erythroid precursor proliferation, inhibition of apoptosis, and differentiation of the erythroid lineage. Early studies showed that mice with targeted deletion of Epo or the Epo receptor (EpoR) show impaired erythropoiesis, lack mature erythrocytes, and die in utero around embryonic day 13.5 (Wu et al. Cell 83:59-67, 1995; Lin et al. Genes Dev. 10:154-164, 1996). These animals also exhibited heart defects, abnormal vascular development as well as increased apoptosis in the brain suggesting additional functions for Epo signaling in normal development of the central nervous system and heart. Now, in addition to its well-known role in erythropoiesis, a diverse array of cells have been identified that produce Epo and/or express the Epo-R including endothelial cells, smooth muscle cells, and cells of the central nervous system (Masuda et al. J Biol Chem. 269:19488-19493, 1994; Marti et al. Eur J Neurosci. 8:666-676, 1996; Bernaudin et al. J Cereb Blood Flow Metab. 19:643-651, 1999; Li et al. Neurochem Res. 32:2132-2141, 2007). Endogenously produced Epo and/or expression of the EpoR gives rise to autocrine and paracrine signaling in different organs particularly during hypoxia, toxicity, and injury conditions. Epo has been shown to regulate a variety of cell functions such as calcium flux (Korbel et al. J Comp Physiol B. 174:121-128, 2004) neurotransmitter synthesis and cell survival (Velly et al. Pharmacol Ther. 128:445-459, 2010; Vogel et al. Blood. 102:2278-2284, 2003). Furthermore Epo has neurotrophic effects (Grimm et al. Nat Med. 8:718-724, 2002; Junk et al. Proc Natl Acad Sci U S A. 99:10659-10664, 2002), can induce an angiogenic phenotype in cultured endothelial cells and is a potent angiogenic factor in vivo (Ribatti et al. Eur J Clin Invest. 33:891-896, 2003) and might enhance ventilation in hypoxic conditions (Soliz et al. J Physiol. 568:559-571, 2005; Soliz et al. J Physiol. 583, 329-336, 2007). Thus multiple functions have been identified breathing new life and exciting possibilities into what is really an old growth factor.This review will address the function of Epo in non-hematopoietic tissues with significant emphasis on the brain and heart.
Collapse
Affiliation(s)
- Omolara O Ogunshola
- Institute of Veterinary Physiology, Vetsuisse Faculty and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | | |
Collapse
|
29
|
Soliz J. Erythropoietin and respiratory control at adulthood and during early postnatal life. Respir Physiol Neurobiol 2013; 185:87-93. [DOI: 10.1016/j.resp.2012.07.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 07/19/2012] [Accepted: 07/20/2012] [Indexed: 01/10/2023]
|
30
|
Benderro GF, LaManna JC. Kidney EPO expression during chronic hypoxia in aged mice. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 765:9-14. [PMID: 22879008 DOI: 10.1007/978-1-4614-4989-8_2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In order to maintain normal cellular function, mammalian tissue oxygen concentrations must be tightly regulated within a narrow physiological range. The hormone erythropoietin (EPO) is essential for maintenance of tissue oxygen supply by stimulating red blood cell production and promoting their survival. In this study we compared the effects of 290 Torr atmospheric pressure on the kidney EPO protein levels in young (4-month-old) and aged (24-month-old) C57BL/6 mice. The mice were sacrificed after being anesthetized, and kidney samples were collected and processed by Western blot analysis. Relatively low basal expression of EPO during normoxia in young mice showed significant upregulation in hypoxia and stayed upregulated throughout the hypoxic period (threefold compared to normoxic control), showing a slight decline toward the third week. Whereas, a relatively higher normoxic basal EPO protein level in aged mice did not show significant increase until seventh day of hypoxia, but showed significant upregulation in prolonged hypoxia. Hence, we confirmed that there is a progressively increased accumulation of EPO during chronic hypoxia in young and aged mouse kidney, and the EPO upregulation during hypoxia showed a similarity with the pattern of increase in hematocrit, which we have reported previously.
Collapse
Affiliation(s)
- Girriso F Benderro
- Department of Anatomy, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Joseph C LaManna
- Department of Physiology and Biophysics, Case Western Reserve University, 10900 Euclid Avenue, Robbins E 611, Cleveland, OH, 44106-4970, USA.
| |
Collapse
|
31
|
Effect of Recombinant Erythropoietin on Functional Activity of Cultured Human Cells. Bull Exp Biol Med 2012; 153:554-8. [DOI: 10.1007/s10517-012-1765-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
32
|
Lee Y, Nam HY, Kim J, Lee M, Yockman JW, Shin SK, Kim SW. Human erythropoietin gene delivery using an arginine-grafted bioreducible polymer system. Mol Ther 2012; 20:1360-6. [PMID: 22472948 DOI: 10.1038/mt.2012.62] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Erythropoiesis-stimulating agents are widely used to treat anemia for chronic kidney disease (CKD) and cancer, however, several clinical limitations impede their effectiveness. Nonviral gene therapy systems are a novel solution to these problems as they provide stable and low immunogenic protein expression levels. Here, we show the application of an arginine-grafted bioreducible poly(disulfide amine) (ABP) polymer gene delivery system as a platform for in vivo transfer of human erythropoietin plasmid DNA (phEPO) to produce long-term, therapeutic erythropoiesis. A single systemic injection of phEPO/ABP polyplex led to higher hematocrit levels over a 60-day period accompanied with reticulocytosis and high hEPO protein expression. In addition, we found that the distinct temporal and spatial distribution of phEPO/ABP polyplexes contributed to increased erythropoietic effects compared to those of traditional EPO therapies. Overall, our study suggests that ABP polymer-based gene therapy provides a promising clinical strategy to reach effective therapeutic levels of hEPO gene.
Collapse
Affiliation(s)
- Youngsook Lee
- Center for Controlled Chemical Delivery, Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, Utah 84112-5820, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Ponce LL, Navarro JC, Ahmed O, Robertson CS. Erythropoietin neuroprotection with traumatic brain injury. ACTA ACUST UNITED AC 2012; 20:31-8. [PMID: 22421507 DOI: 10.1016/j.pathophys.2012.02.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Numerous experimental studies in recent years have suggested that erythropoietin (EPO) is an endogenous mediator of neuroprotection in various central nervous system disorders, including TBI. Many characteristics of EPO neuroprotection that have been defined in TBI experimental models suggest that it is an attractive candidate for a new treatment of TBI. EPO targets multiple mechanisms known to cause secondary injury after TBI, including anti-excitotoxic, antioxidant, anti-edematous, and anti-inflammatory mechanisms. EPO crosses the blood-brain barrier. EPO has a known dose response and time window for neuroprotection and neurorestoration that would be practical in the clinical setting. However, EPO also stimulates erythropoiesis, which can result in thromboembolic complications. Derivatives of EPO which do not bind to the classical EPO receptor (carbamylated EPO) or that have such a brief half-life in the circulation that they do not stimulate erythropoiesis (asialo EPO and neuro EPO) have the neuroprotective activities of EPO without these potential thromboembolic adverse effects associated with EPO administration. Likewise, a peptide based on the structure of the Helix B segment of the EPO molecule that does not bind to the EPO receptor (pyroglutamate Helix B surface peptide) has promise as another alternative to EPO that may provide neuroprotection without stimulating erythropoiesis.
Collapse
Affiliation(s)
- Lucido L Ponce
- Department of Neurosurgery, Baylor College of Medicine, United States
| | | | | | | |
Collapse
|
34
|
Erythropoietin in brain development and beyond. ANATOMY RESEARCH INTERNATIONAL 2012; 2012:953264. [PMID: 22567318 PMCID: PMC3335485 DOI: 10.1155/2012/953264] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Revised: 10/27/2011] [Accepted: 11/11/2011] [Indexed: 01/17/2023]
Abstract
Erythropoietin is known as the requisite cytokine for red blood cell production. Its receptor, expressed at a high level on erythroid progenitor/precursor cells, is also found on endothelial, neural, and other cell types. Erythropoietin and erythropoietin receptor expression in the developing and adult brain suggest their possible involvement in neurodevelopment and neuroprotection. During ischemic stress, erythropoietin, which is hypoxia inducible, can contribute to brain homeostasis by increasing red blood cell production to increase the blood oxygen carrying capacity, stimulate nitric oxide production to modulate blood flow and contribute to the neurovascular response, or act directly on neural cells to provide neuroprotection as demonstrated in culture and animal models. Clinical studies of erythropoietin treatment in stroke and other diseases provide insight on safety and potential adverse effects and underscore the potential pleiotropic activity of erythropoietin. Herein, we summarize the roles of EPO and its receptor in the developing and adult brain during health and disease, providing first a brief overview of the well-established EPO biology and signaling, its hypoxic regulation, and role in erythropoiesis.
Collapse
|
35
|
|
36
|
Erythropoietin: recent developments in the treatment of spinal cord injury. Neurol Res Int 2011; 2011:453179. [PMID: 21766022 PMCID: PMC3135044 DOI: 10.1155/2011/453179] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2011] [Accepted: 05/09/2011] [Indexed: 01/10/2023] Open
Abstract
Erythropoietin (EPO), originally identified for its critical function in regulating production and survival of erythrocytes, is a member of the type 1 cytokine superfamily. Recent studies have shown that EPO has cytoprotective effects in a wide variety of cells and tissues. Here is presented the analysis of EPO effects on spinal cord injury (SCI), considering both animal experiments concerning to mechanisms of neurodegeneration in SCI and EPO as a neuroprotective agent, and some evidences coming from ongoing clinical trials. The evidences underling that EPO could be a promising therapeutic agent in a variety of neurological insults, including trauma, are mounting. In particular, it is highlighted that administration of EPO or other recently generated EPO analogues such as asialo-EPO and carbamylated-EPO demonstrate interesting preclinical and clinical characteristics, rendering the evaluation of these tissue-protective agents imperative in human clinical trials. Moreover the demonstration of rhEPO and its analogues' broad neuroprotective effects in animal models of cord lesion and in human trial like stroke, should encourage scientists and clinicians to design clinical trials assessing the efficacy of these pharmacological compounds on SCI.
Collapse
|
37
|
Carilho R, de Carvalho M, Kuehl U, Pinto S, Pinto A, Kromminga A, Costa J. Erythropoietin and amyotrophic lateral sclerosis: Plasma level determination. ACTA ACUST UNITED AC 2011; 12:439-43. [PMID: 21722017 DOI: 10.3109/17482968.2011.584881] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
In amyotrophic lateral sclerosis (ALS), respiratory muscle weakness causes ventilatory insufficiency and tissue hypoxia, which induces a number of metabolic pathways, and in particular increases erythropoietin (EPO) synthesis. EPO is a glycoprotein with neuroprotective properties that stimulates erythropoiesis. Here, EPO plasma level in a large population of ALS patients, with and without respiratory failure, was measured. Plasma EPO level of patients with ALS (n = 98), controls with other neuromuscular diseases (n = 58) and healthy controls (n = 20) has been quantified by ELISA. No significant difference was found between ALS patients and the two control groups. EPO level was not different between bulbar- and spinal-onset patients and was not correlated with disease duration or functional impairment. However, in the ALS group EPO level was higher in females (p = 0.0006) and correlated positively with age (p = 0.006). The subgroup of ALS patients with respiratory failure had higher plasma levels of EPO compared with ALS patients with preserved respiratory function (p = 0.016), but short-term non-invasive ventilation did not change EPO level. In conclusion, EPO levels were found to be significantly higher in ALS patients with respiratory impairment representing preservation of this homeostatic mechanism.
Collapse
Affiliation(s)
- Rita Carilho
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | | | | | | | | | | | | |
Collapse
|
38
|
Sargin D, Friedrichs H, El-Kordi A, Ehrenreich H. Erythropoietin as neuroprotective and neuroregenerative treatment strategy: comprehensive overview of 12 years of preclinical and clinical research. Best Pract Res Clin Anaesthesiol 2010; 24:573-94. [PMID: 21619868 DOI: 10.1016/j.bpa.2010.10.005] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Accepted: 10/11/2010] [Indexed: 12/13/2022]
Abstract
Erythropoietin (EPO), originally discovered as hematopoietic growth factor, has direct effects on cells of the nervous system that make it a highly attractive candidate drug for neuroprotection/neuroregeneration. Hardly any other compound has led to so much preclinical work in the field of translational neuroscience than EPO. Almost all of the >180 preclinical studies performed by many independent research groups from all over the world in the last 12 years have yielded positive results on EPO as a neuroprotective drug. The fact that EPO was approved for the treatment of anemia >20 years ago and found to be well tolerated and safe, facilitated the first steps of translation from preclinical findings to the clinic. On the other hand, the same fact, naturally associated with loss of patent protection, hindered to develop EPO as a highly promising therapeutic strategy for application in human brain disease. Therefore, only few clinical neuroprotection studies have been concluded, all with essentially positive and stimulating results, but no further development towards the clinic has occurred thus far. This article reviews the preclinical and clinical work on EPO for the indications neuroprotection/neuroregeneration and cognition, and hopefully will stimulate new endeavours promoting development of EPO for the treatment of human brain diseases.
Collapse
Affiliation(s)
- Derya Sargin
- Division of Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Hermann-Rein Str. 3, 37075 Göttingen, Germany
| | | | | | | |
Collapse
|
39
|
Ergun Y, Koc A, Dolapcioglu K, Akaydin Y, Dogruer G, Kontas T, Kozlu T, Aslan E. The protective effect of erythropoietin and dimethylsulfoxide on ischemia-reperfusion injury in rat ovary. Eur J Obstet Gynecol Reprod Biol 2010; 152:186-90. [DOI: 10.1016/j.ejogrb.2010.05.025] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2010] [Revised: 04/28/2010] [Accepted: 05/26/2010] [Indexed: 11/30/2022]
|
40
|
Iwai M, Stetler RA, Xing J, Hu X, Gao Y, Zhang W, Chen J, Cao G. Enhanced oligodendrogenesis and recovery of neurological function by erythropoietin after neonatal hypoxic/ischemic brain injury. Stroke 2010; 41:1032-7. [PMID: 20360553 PMCID: PMC2919308 DOI: 10.1161/strokeaha.109.570325] [Citation(s) in RCA: 163] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2009] [Accepted: 01/27/2010] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Neuronal replacement has recently gained attention as a potential therapeutic target under ischemic conditions. However, the oligodendrogenic infrastructure is equally critical for restoration of brain function and is also sensitive to ischemic injury. Erythropoietin (EPO) is a neuroprotective molecule that stimulates neuronal replacement after neonatal hypoxia/ischemia (H/I) when delivered soon after the onset of reperfusion. Because EPO can improve recovery of neurological function in the absence of tissue protection, we hypothesize that EPO may improve neurological function via enhancement of white matter recovery after H/I. Thus, we sought to determine the effects of delayed administration of EPO on white matter injury and recovery of neurological function after neonatal H/I. METHODS EPO (1000 U/kg) was injected intraperitoneally at multiple time points beginning 48 hours after H/I in postnatal day 7 rats. The effects of EPO on oligodendrogenesis, white matter injury, and neurogenesis were evaluated using bromodeoxyuridine incorporation and cell-specific immunohistochemistry. Neurological function was assessed by sensorimotor behavioral tests. RESULTS Delayed administration of EPO was incapable of reducing brain volume loss but significantly increased oligodendrogenesis and maturation of oligodendrocytes and attenuated white matter injury after H/I. These effects occurred concurrently with enhanced neurogenesis. Delayed EPO treatment improved behavioral neurological outcomes 14 days after H/I injury. CONCLUSIONS Our study demonstrates that delayed administration of EPO promotes oligodendrogenesis and attenuates white matter injury concurrently with increased neurogenesis. These effects likely contribute to the observed improvement in neurological functional outcomes.
Collapse
Affiliation(s)
- Masanori Iwai
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Turner JD, Mammis A, Prestigiacomo CJ. Erythropoietin for the Treatment of Subarachnoid Hemorrhage: A Review. World Neurosurg 2010; 73:500-7. [DOI: 10.1016/j.wneu.2010.02.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2009] [Accepted: 02/10/2010] [Indexed: 12/20/2022]
|
42
|
Oxidative stress: Biomarkers and novel therapeutic pathways. Exp Gerontol 2010; 45:217-34. [PMID: 20064603 DOI: 10.1016/j.exger.2010.01.004] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2009] [Revised: 12/28/2009] [Accepted: 01/07/2010] [Indexed: 01/12/2023]
Abstract
Oxidative stress significantly impacts multiple cellular pathways that can lead to the initiation and progression of varied disorders throughout the body. It therefore becomes imperative to elucidate the components and function of novel therapeutic strategies against oxidative stress to further clinical diagnosis and care. In particular, both the growth factor and cytokine erythropoietin (EPO) and members of the mammalian forkhead transcription factors of the O class (FoxOs) may offer the greatest promise for new treatment regimens since these agents and the cellular pathways they oversee cover a range of critical functions that directly influence progenitor cell development, cell survival and degeneration, metabolism, immune function, and cancer cell invasion. Furthermore, both EPO and FoxOs function not only as therapeutic targets, but also as biomarkers of disease onset and progression, since their cellular pathways are closely linked and overlap with several unique signal transduction pathways. However, biological outcome with EPO and FoxOs may sometimes be both unexpected and undesirable that can raise caution for these agents and warrant further investigations. Here we present the exciting as well as complicated role EPO and FoxOs possess to uncover the benefits as well as the risks of these agents for cell biology and clinical care in processes that range from stem cell development to uncontrolled cellular proliferation.
Collapse
|
43
|
Kim HW, Kim JL, Lee HK, Hur DY, Yun IH, Kim SD. Enalapril Alters Expression of Key Growth Factors in Experimental Diabetic Retinopathy. Curr Eye Res 2009; 34:976-87. [DOI: 10.3109/02713680903249913] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
44
|
Expression of Erythropoietin mRNA in the Brainstem of Rats Adapted to Intermittent Hypoxia. NEUROPHYSIOLOGY+ 2009. [DOI: 10.1007/s11062-009-9095-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
45
|
Weidemann A, Kerdiles YM, Knaup KX, Rafie CA, Boutin AT, Stockmann C, Takeda N, Scadeng M, Shih AY, Haase VH, Simon MC, Kleinfeld D, Johnson RS. The glial cell response is an essential component of hypoxia-induced erythropoiesis in mice. J Clin Invest 2009; 119:3373-83. [PMID: 19809162 DOI: 10.1172/jci39378] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2009] [Accepted: 08/05/2009] [Indexed: 11/17/2022] Open
Abstract
A key adaptation to environmental hypoxia is an increase in erythropoiesis, driven by the hormone erythropoietin (EPO) through what is traditionally thought to be primarily a renal response. However, both neurons and astrocytes (the largest subpopulation of glial cells in the CNS) also express EPO following ischemic injury, and this response is known to ameliorate damage to the brain. To investigate the role of glial cells as a component of the systemic response to hypoxia, we created astrocyte-specific deletions of the murine genes encoding the hypoxia-inducible transcription factors HIF-1alpha and HIF-2alpha and their negative regulator von Hippel-Lindau (VHL) as well as astrocyte-specific deletion of the HIF target gene Vegf. We found that loss of the hypoxic response in astrocytes does not cause anemia in mice but is necessary for approximately 50% of the acute erythropoietic response to hypoxic stress. In accord with this, erythroid progenitor cells and reticulocytes were substantially reduced in number in mice lacking HIF function in astrocytes following hypoxic stress. Thus, we have demonstrated that the glial component of the CNS is an essential component of hypoxia-induced erythropoiesis.
Collapse
Affiliation(s)
- Alexander Weidemann
- Division of Biology, University of California, San Diego, La Jolla, California 92093, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Maiese K, Hou J, Chong ZZ, Shang YC. Erythropoietin, forkhead proteins, and oxidative injury: biomarkers and biology. ScientificWorldJournal 2009; 9:1072-104. [PMID: 19802503 PMCID: PMC2762199 DOI: 10.1100/tsw.2009.121] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Oxidative stress significantly impacts multiple cellular pathways that can lead to the initiation and progression of varied disorders throughout the body. It therefore becomes imperative to elucidate the components and function of novel therapeutic strategies against oxidative stress to further clinical diagnosis and care. In particular, both the growth factor and cytokine erythropoietin (EPO), and members of the mammalian forkhead transcription factors of the O class (FoxOs), may offer the greatest promise for new treatment regimens, since these agents and the cellular pathways they oversee cover a range of critical functions that directly influence progenitor cell development, cell survival and degeneration, metabolism, immune function, and cancer cell invasion. Furthermore, both EPO and FoxOs function not only as therapeutic targets, but also as biomarkers of disease onset and progression, since their cellular pathways are closely linked and overlap with several unique signal transduction pathways. Yet, EPO and FoxOs may sometimes have unexpected and undesirable effects that can raise caution for these agents and warrant further investigations. Here we present the exciting as well as the complex role that EPO and FoxOs possess to uncover the benefits as well as the risks of these agents for cell biology and clinical care in processes that range from stem cell development to uncontrolled cellular proliferation.
Collapse
Affiliation(s)
- Kenneth Maiese
- Division of Cellular and Molecular Cerebral Ischemia, Wayne State University School of Medicine, Detroit, Michigan, USA.
| | | | | | | |
Collapse
|
47
|
Bakan V, Ciralik H, Tolun FI, Atli Y, Mil A, Oztürk S. Protective effect of erythropoietin on torsion/detorsion injury in rat model. J Pediatr Surg 2009; 44:1988-94. [PMID: 19853760 DOI: 10.1016/j.jpedsurg.2009.02.071] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2008] [Revised: 02/12/2009] [Accepted: 02/13/2009] [Indexed: 11/25/2022]
Abstract
PURPOSE The aim of the study is to investigate the effects of erythropoietin on torsion/detorsion injury in rats. METHODS Forty rats were divided randomly into 5 groups: group I (sham, S), sham operation; group II (torsion/detorsion 1, T/D(1)), 3 hours ischemia and 1 hour reperfusion; group III (torsion/detorsion 2, T/D(2)), 3 hours ischemia and 48 hours reperfusion; group IV (erythropoietin 1, EPO(1)), 3 hours ischemia, 1 hour reperfusion, and a single dose of EPO; and group V (erythropoietin 2, EPO(2)), 3 hours ischemia, 48 hours reperfusion, and 2 doses of EPO. Malondialdehyde (MDA) and nitric oxide (NO) levels and activities of superoxide dismutase and catalase were measured. Tissue damage to ovarian tissue was scored by histologic examination. Data were compared among groups with parametric tests. RESULTS The MDA levels in the S and EPO groups were significantly lower than the T/D groups (P < .001). Catalase and superoxide dismutase activities, and NO levels in the S and EPO groups were significantly higher than in the T/D groups (P < .05). Ovarian tissue damage in the S and EPO groups was significantly less than in the T/D groups (P < .05). Levels of all biochemical markers and ovarian tissue damage scores were similar among the S, EPO(1), and EPO(2) groups (P > .05). CONCLUSION Erythropoietin attenuates ischemia-reperfusion injury when given during the acute phase of ovarian torsion-detorsion in a rat model.
Collapse
Affiliation(s)
- Vedat Bakan
- Faculty of Medicine, Department of Pediatric Surgery, Sutcu Imam University, Kahramanmaras 46100, Turkey.
| | | | | | | | | | | |
Collapse
|
48
|
Rabie T, Marti HH. Brain protection by erythropoietin: a manifold task. Physiology (Bethesda) 2008; 23:263-74. [PMID: 18927202 DOI: 10.1152/physiol.00016.2008] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Many hematopoietic growth factors are produced locally in the brain. Among these, erythropoietin (Epo), has a dominant role for neuroprotection, neurogenesis, and acting as a neurotrophic factor in the central nervous system. These functions make erythropoietin a good candidate for treating diseases associated with neuronal cell death.
Collapse
Affiliation(s)
- Tamer Rabie
- Institute of Physiology and Pathophysiology, University of Heidelberg, Heidelberg, Germany
| | | |
Collapse
|
49
|
Bartels C, Späte K, Krampe H, Ehrenreich H. Recombinant Human Erythropoietin: Novel Strategies for Neuroprotective/Neuro-regenerative Treatment of Multiple Sclerosis. Ther Adv Neurol Disord 2008; 1:193-206. [PMID: 21180577 PMCID: PMC3002551 DOI: 10.1177/1756285608098422] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Treatment of multiple sclerosis (MS) is still unsatisfactory and essentially non-existing for the progressive course of the disease. Recombinant human erythropoietin (EPO) may be a promising neuroprotective/neuroregenerative treatment of MS. In the nervous system, EPO acts anti-apoptotic, antioxidative, anti-inflammatory, neurotrophic and plasticity-modulating. Beneficial effects have been shown in animal models of various neurological and psychiatric diseases, including different models of experimental autoimmune encephalomyelitis. EPO is also effective in human brain disease, as shown in double-blind placebo-controlled clinical studies on ischemic stroke and chronic schizophrenia. An exploratory study on chronic progressive MS yielded lasting improvement in motor and cognitive performance upon high-dose long-term EPO treatment.
Collapse
Affiliation(s)
- Claudia Bartels
- Division of Clinical Neuroscience, Max-Planck-Institute of Experimental
Medicine, Göttingen, Germany
| | - Kira Späte
- Division of Clinical Neuroscience, Max-Planck-Institute of Experimental
Medicine, Göttingen, Germany
| | - Henning Krampe
- Division of Clinical Neuroscience, Max-Planck-Institute of Experimental
Medicine, Göttingen, Germany
| | - Hannelore Ehrenreich
- Ehrenreich Division of Clinical Neuroscience, Max-Planck-Institute of
Experimental Medicine, Göttingen, Germany,
| |
Collapse
|
50
|
Fauchère JC, Dame C, Vonthein R, Koller B, Arri S, Wolf M, Bucher HU. An approach to using recombinant erythropoietin for neuroprotection in very preterm infants. Pediatrics 2008; 122:375-82. [PMID: 18676556 DOI: 10.1542/peds.2007-2591] [Citation(s) in RCA: 113] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE Erythropoietin has been shown to be protective against hypoxic-ischemic and inflammatory injuries in cell culture, animal models of brain injury, and clinical trials of adult humans. The rationale for our study was that early administration of high-dose recombinant human erythropoietin may reduce perinatal brain injury (intraventricular hemorrhage and periventricular leukomalacia) in very preterm infants and improve neurodevelopmental outcome. We investigated whether administration of high-dose recombinant human erythropoietin to very preterm infants shortly after birth and subsequently during the first 2 days is safe in terms of short-term outcome. METHODS This was a randomized, double-masked, single-center trial with a 2:1 allocation in favor of recombinant human erythropoietin. Preterm infants (gestational age: 24 to 31 weeks) were given recombinant human erythropoietin or NaCl 0.9% intravenously 3, 12 to 18, and 36 to 42 hours after birth. RESULTS The percentage of infants who survived without brain injury or retinopathy was 53% in the recombinant human erythropoietin group and 60% in the placebo group. There were no relevant differences regarding short-term outcomes such as intraventricular hemorrhage, retinopathy, sepsis, necrotizing enterocolitis, and bronchopulmonary dysplasia. For 5 infants who were in the recombinant human erythropoietin group and had a gestational age of <26 weeks, withdrawal of intensive care was decided (3 of 5 with severe bilateral intraventricular hemorrhage, 2 of 5 with pulmonary insufficiency); no infant of the control group died. Recombinant human erythropoietin treatment did not result in significant differences in blood pressure, cerebral oxygenation, hemoglobin, leukocyte, and platelet count. CONCLUSIONS No significant adverse effects of early high-dose recombinant human erythropoietin treatment in very preterm infants were identified. These results enable us to embark on a large multicenter trial with the aim of determining whether early high-dose administration of recombinant human erythropoietin to very preterm infants improves neurodevelopmental outcome at 24 months' and 5 years' corrected age.
Collapse
|