1
|
Guan Y, Zhou H, Luo B, Hussain S, Xiong L. Research progress of neonatal hypoxic-ischemic encephalopathy in nonhuman primate models. IBRAIN 2023; 9:183-194. [PMID: 37786551 PMCID: PMC10528769 DOI: 10.1002/ibra.12097] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 02/25/2023] [Accepted: 03/02/2023] [Indexed: 10/04/2023]
Abstract
Neonatal hypoxic-ischemic encephalopathy (HIE) is one of the important complications of neonatal asphyxia, which not only leads to neurological disability but also seriously threatens the life of neonates. Over the years, animal models of HIE have been a research hotspot to find ways to cope with HIE and thereby reduce the risk of neonatal death or disability in moderate-to-severe HIE. By reviewing the literature related to HIE over the years, it was found that nonhuman primates share a high degree of homology with human gross neural anatomy. The basic data on nonhuman primates are not yet complete, so it is urgent to mine and develop new nonhuman primate model data. In recent years, the research on nonhuman primate HIE models has been gradually enriched and the content is more novel. Therefore, the purpose of this review is to further summarize the methods for establishing the nonhuman primate HIE model and to better elucidate the relevance of the nonhuman primate model to humans by observing the behavioral manifestations, neuropathology, and a series of biomarkers of HIE in primates HIE. Finally, the most popular and desirable treatments studied in nonhuman primate models in the past 5 years are summarized.
Collapse
Affiliation(s)
- Yi‐Huan Guan
- School of AnesthesiologyZunyi Medical UniversityZunyiChina
| | - Hong‐Su Zhou
- Department of Experimental AnimalsKunming Medical UniversityKunmingChina
| | - Bo‐Yan Luo
- School of PharmacyZunyi Medical UniversityZunyiChina
| | - Sajid Hussain
- NUTECH School of Applied Sciences and HumanitiesNational University of TechnologyIslamabadPakistan
| | - Liu‐Lin Xiong
- School of Pharmacy and Medical Sciences, Faculty of Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| |
Collapse
|
2
|
Knorr DY, Rodriguez Polo I, Pies HS, Schwedhelm-Domeyer N, Pauls S, Behr R, Heinrich R. The cytokine receptor CRLF3 is a human neuroprotective EV-3 (Epo) receptor. Front Mol Neurosci 2023; 16:1154509. [PMID: 37168680 PMCID: PMC10165946 DOI: 10.3389/fnmol.2023.1154509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 03/15/2023] [Indexed: 05/13/2023] Open
Abstract
The evolutionary conserved orphan cytokine receptor-like factor 3 (CRLF3) has been implicated in human disease, vertebrate hematopoiesis and insect neuroprotection. While its specific functions are elusive, experimental evidence points toward a general role in cell homeostasis. Erythropoietin (Epo) is a major regulator of vertebrate hematopoiesis and a general cytoprotective cytokine. Erythropoietic functions mediated by classical Epo receptor are understood in great detail whereas Epo-mediated cytoprotective mechanisms are more complex due to involvement of additional Epo receptors and a non-erythropoietic splice variant with selectivity for certain receptors. In the present study, we show that the human CRLF3 mediates neuroprotection upon activation with the natural Epo splice variant EV-3. We generated CRLF3 knock-out iPSC lines and differentiated them toward the neuronal lineage. While apoptotic death of rotenone-challenged wild type iPSC-derived neurons was prevented by EV-3, EV-3-mediated neuroprotection was absent in CRLF3 knock-out neurons. Rotenone-induced apoptosis and EV-3-mediated neuroprotection were associated with differential expression of pro-and anti-apoptotic genes. Our data characterize human CRLF3 as a receptor involved in Epo-mediated neuroprotection and identify CRLF3 as the first known receptor for EV-3.
Collapse
Affiliation(s)
- Debbra Y. Knorr
- Department of Cellular Neurobiology, Johann-Friedrich-Blumenbach Institute for Zoology and Anthropology, Georg-August University Göttingen, Göttingen, Germany
- *Correspondence: Debbra Y. Knorr,
| | - Ignacio Rodriguez Polo
- Department of Developmental Biology, Göttingen Center for Molecular Biosciences, Georg-August University Göttingen, Göttingen, Germany
- Research Platform Degenerative Diseases, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany
- Developmental Models Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Hanna S. Pies
- Department of Cellular Neurobiology, Johann-Friedrich-Blumenbach Institute for Zoology and Anthropology, Georg-August University Göttingen, Göttingen, Germany
| | - Nicola Schwedhelm-Domeyer
- Department of Cellular Neurobiology, Johann-Friedrich-Blumenbach Institute for Zoology and Anthropology, Georg-August University Göttingen, Göttingen, Germany
| | - Stephanie Pauls
- Department of Cellular Neurobiology, Johann-Friedrich-Blumenbach Institute for Zoology and Anthropology, Georg-August University Göttingen, Göttingen, Germany
| | - Rüdiger Behr
- Research Platform Degenerative Diseases, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany
| | - Ralf Heinrich
- Department of Cellular Neurobiology, Johann-Friedrich-Blumenbach Institute for Zoology and Anthropology, Georg-August University Göttingen, Göttingen, Germany
- Ralf Heinrich,
| |
Collapse
|
3
|
Robinson S, Winer JL, Kitase Y, Brigman JL, Jantzie LL. Neonatal administration of erythropoietin attenuates cognitive deficits in adult rats following placental insufficiency. J Neurosci Res 2022; 100:2112-2126. [PMID: 33611820 PMCID: PMC10097461 DOI: 10.1002/jnr.24815] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 01/29/2021] [Accepted: 02/04/2021] [Indexed: 01/07/2023]
Abstract
Preterm birth is a principal cause of neurological disability later in life, including cognitive and behavioral deficits. Notably, cognitive impairment has greater impact on quality of life than physical disability. Survivors of preterm birth commonly have deficits of executive function. Difficulties with tasks and planning complexity correlate positively with increasing disability. To overcome these barriers for children born preterm, preclinical and clinical studies have emphasized the importance of neurorestoration. Erythropoietin (EPO) is a endogenous cytokine with multiple beneficial mechanisms of action following perinatal brain injury. While most preclinical investigations have focused on pathology and molecular mechanisms, translational studies of repair using clinically viable biobehavioral biomarkers are still lacking. Here, using an established model of encephalopathy of prematurity secondary to placental insufficiency, we tested the hypothesis that administration of EPO in the neonatal period would attenuate deficits in recognition memory and cognitive flexibility in adult rats of both sexes. We assessed cognition and executive function in two ways. First, using the classic test of novel object recognition and second, using a touchscreen platform. Touchscreen testing allows for rigorous testing of cognition and executive function in preclinical and clinical scenarios. Data show that adult rats exhibit deficits in recognition memory and cognitive flexibility following in utero placental insufficiency. Notably, neonatal treatment of EPO attenuates these deficits in adulthood and facilitates functional repair. Together, these data validate EPO neurorestoration using a clinically relevant outcome measure and support the concept that postnatal treatment following in utero injury can improve cognition and executive function through adulthood.
Collapse
Affiliation(s)
- Shenandoah Robinson
- Division of Pediatric Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jesse L Winer
- Division of Pediatric Neurosurgery, Oregon Health and Science University, Portland, OR, USA
| | - Yuma Kitase
- Division of Neonatal-Perinatal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jonathan L Brigman
- Department of Neuroscience, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Lauren L Jantzie
- Division of Pediatric Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Division of Neonatal-Perinatal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Kennedy Krieger Institute, Baltimore, MD, USA
| |
Collapse
|
4
|
Introducing the brain erythropoietin circle to explain adaptive brain hardware upgrade and improved performance. Mol Psychiatry 2022; 27:2372-2379. [PMID: 35414656 PMCID: PMC9004453 DOI: 10.1038/s41380-022-01551-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 03/22/2022] [Accepted: 03/25/2022] [Indexed: 12/30/2022]
Abstract
Executive functions, learning, attention, and processing speed are imperative facets of cognitive performance, affected in neuropsychiatric disorders. In clinical studies on different patient groups, recombinant human (rh) erythropoietin (EPO) lastingly improved higher cognition and reduced brain matter loss. Correspondingly, rhEPO treatment of young rodents or EPO receptor (EPOR) overexpression in pyramidal neurons caused remarkable and enduring cognitive improvement, together with enhanced hippocampal long-term potentiation. The 'brain hardware upgrade', underlying these observations, includes an EPO induced ~20% increase in pyramidal neurons and oligodendrocytes in cornu ammonis hippocampi in the absence of elevated DNA synthesis. In parallel, EPO reduces microglia numbers and dampens their activity and metabolism as prerequisites for undisturbed EPO-driven differentiation of pre-existing local neuronal precursors. These processes depend on neuronal and microglial EPOR. This novel mechanism of powerful postnatal neurogenesis, outside the classical neurogenic niches, and on-demand delivery of new cells, paralleled by dendritic spine increase, let us hypothesize a physiological procognitive role of hypoxia-induced endogenous EPO in brain, which we imitate by rhEPO treatment. Here we delineate the brain EPO circle as working model explaining adaptive 'brain hardware upgrade' and improved performance. In this fundamental regulatory circle, neuronal networks, challenged by motor-cognitive tasks, drift into transient 'functional hypoxia', thereby triggering neuronal EPO/EPOR expression.
Collapse
|
5
|
Newton SS, Sathyanesan M. Erythropoietin and Non-Erythropoietic Derivatives in Cognition. Front Pharmacol 2021; 12:728725. [PMID: 34552490 PMCID: PMC8450392 DOI: 10.3389/fphar.2021.728725] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/19/2021] [Indexed: 01/04/2023] Open
Abstract
Cognitive deficits are widespread in psychiatric disorders, including major depression and schizophrenia. These deficits are known to contribute significantly to the accompanying functional impairment. Progress in the development of targeted treatments of cognitive deficits has been limited and there exists a major unmet need to develop more efficacious treatments. Erythropoietin (Epo) has shown promising procognitive effects in psychiatric disorders, providing support for a neurotrophic drug development approach. Several preclinical studies with non-erythropoietic derivatives have demonstrated that the modulation of behavior is independent of erythropoiesis. In this review, we examine the molecular, cellular and cognitive actions of Epo and non-erythropoietic molecular derivatives by focusing on their neurotrophic, synaptic, myelin plasticity, anti-inflammatory and neurogenic mechanisms in the brain. We also discuss the role of receptor signaling in Epo and non-erythropoietic EPO-mimetic molecules in their procognitive effects.
Collapse
Affiliation(s)
- Samuel S Newton
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States.,Sioux Falls VA Healthcare System, Sioux Falls, SD, United States
| | - Monica Sathyanesan
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States.,Sioux Falls VA Healthcare System, Sioux Falls, SD, United States
| |
Collapse
|
6
|
Webb SD, Orton LD. Microglial peri-somatic abutments classify two novel types of GABAergic neuron in the inferior colliculus. Eur J Neurosci 2020; 54:5815-5833. [PMID: 33278847 DOI: 10.1111/ejn.15075] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 05/14/2020] [Accepted: 05/20/2020] [Indexed: 12/22/2022]
Abstract
Emerging evidence suggests functional roles for microglia in the healthy, mature nervous system. However, we know little of the cellular density and ramified morphology of microglia in sensory systems, and even less of their inter-relationship with inhibitory neurons. We therefore conducted fluorescent multi-channel immunohistochemistry and confocal microscopy in guinea pigs of both sexes for Iba1, GAD67, GFAP, calbindin, and calretinin. We explored these markers in the inferior colliculi (IC), which contain sub-regions specialized for different aspects of auditory processing. First, we found that while the density of Iba1+ somata is similar throughout the IC parenchyma, Iba1+ microglia in dorsal cortex are significantly more ramified than those in the central nucleus or lateral cortex. Conversely, Iba1+ ramifications in ventral central nucleus, a region with the highest density of GAD67+ (putative GABAergic) neurons in IC, are longer with fewer ramifications. Second, we observed extensive abutments of ramified Iba1+ processes onto GAD67+ somata throughout the whole IC and developed novel measures to quantify these. Cluster analyses revealed two novel sub-types of GAD67+ neuron that differ in the quantity of Iba1+ somatic abutments they receive. Unlike previous classification schemes for GAD67+ neurons in IC, these clusters are not related to GAD67+ soma size. Taken together, these data demonstrate that microglial ramifications vary between IC sub-regions in the healthy, adult IC, possibly related to the ongoing demands of their niche. Furthermore, Iba1+ abutments onto neuronal somata are a novel means by which GAD67+ neurons can be classified.
Collapse
Affiliation(s)
- Samuel David Webb
- Department of Life Sciences, Manchester Metropolitan University, Manchester, UK
| | - Llwyd David Orton
- Department of Life Sciences, Manchester Metropolitan University, Manchester, UK.,Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
7
|
Jantzie L, El Demerdash N, Newville JC, Robinson S. Time to reconsider extended erythropoietin treatment for infantile traumatic brain injury? Exp Neurol 2019; 318:205-215. [PMID: 31082389 DOI: 10.1016/j.expneurol.2019.05.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 05/03/2019] [Accepted: 05/08/2019] [Indexed: 01/03/2023]
Abstract
Pediatric traumatic brain injury (TBI) remains a leading cause of childhood morbidity and mortality worldwide. Most efforts to reduce the chronic impact of pediatric TBI involve prevention and minimization of secondary injury. Currently, no treatments are used in routine clinical care during the acute and subacute phases to actively repair injury to the developing brain. The endogenous pluripotent cytokine erythropoietin (EPO) holds promise as an emerging neuroreparative agent in perinatal brain injury (PBI). EPO signaling in the central nervous system (CNS) is essential for multiple stages of neurodevelopment, including the genesis, survival and differentiation of multiple lineages of neural cells. Postnatally, EPO signaling decreases markedly as the CNS matures. Importantly, high-dose, extended EPO regimens have shown efficacy in preclinical controlled cortical impact (CCI) models of infant TBI at two different, early ages by independent research groups. Specifically, extended high-dose EPO treatment after infantile CCI prevents long-term cognitive deficits in adult rats. Because of the striking differences in the molecular and cellular responses to both injury and recovery in the developing and mature CNS, and the excellent safety profile of EPO in infants and children, extended courses of EPO are currently in Phase III trials for neonates with PBI. Extended, high-dose EPO may also warrant testing for infants and young children with TBI.
Collapse
Affiliation(s)
- Lauren Jantzie
- Division of Neonatology, Department of Pediatrics, University of New Mexico School of Medicine, Albuquerque, NM, 87111,United States.; Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, 87111, United States..
| | - Nagat El Demerdash
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, United States
| | - Jessie C Newville
- Division of Neonatology, Department of Pediatrics, University of New Mexico School of Medicine, Albuquerque, NM, 87111,United States.; Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, NM, 87111, United States
| | - Shenandoah Robinson
- Division of Pediatric Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
8
|
Dias RB, Rodrigues TM, Rombo DM, Ribeiro FF, Rodrigues J, McGarvey J, Orcinha C, Henley JM, Sebastião AM. Erythropoietin Induces Homeostatic Plasticity at Hippocampal Synapses. Cereb Cortex 2018; 28:2795-2809. [PMID: 29053799 PMCID: PMC6117472 DOI: 10.1093/cercor/bhx159] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 05/10/2017] [Accepted: 06/08/2017] [Indexed: 01/30/2023] Open
Abstract
The cytokine erythropoietin (EPO) is the master regulator of erythropoiesis. Intriguingly, many studies have shown that the cognitive performance of patients receiving EPO for its hematopoietic effects is enhanced, which prompted the growing interest in the use of EPO-based strategies to treat neuropsychiatric disorders. EPO plays key roles in brain development and maturation, but also modulates synaptic transmission. However, the mechanisms underlying the latter have remained elusive. Here, we show that acute (40-60 min) exposure to EPO presynaptically downregulates spontaneous and afferent-evoked excitatory transmission, without affecting basal firing of action potentials. Conversely, prolonged (3 h) exposure to EPO, if followed by a recovery period (1 h), is able to elicit a homeostatic increase in excitatory spontaneous, but not in evoked, synaptic transmission. These data lend support to the emerging view that segregated pathways underlie spontaneous and evoked neurotransmitter release. Furthermore, we show that prolonged exposure to EPO facilitates a form of hippocampal long-term potentiation that requires noncanonical recruitment of calcium-permeable AMPA receptors for its maintenance. These findings provide important new insight into the mechanisms by which EPO enhances neuronal function, learning, and memory.
Collapse
Affiliation(s)
- Raquel B Dias
- Institute of Pharmacology and Neurosciences, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, Lisboa, Portugal
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, Lisboa, Portugal
| | - Tiago M Rodrigues
- Institute of Pharmacology and Neurosciences, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, Lisboa, Portugal
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, Lisboa, Portugal
| | - Diogo M Rombo
- Institute of Pharmacology and Neurosciences, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, Lisboa, Portugal
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, Lisboa, Portugal
| | - Filipa F Ribeiro
- Institute of Pharmacology and Neurosciences, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, Lisboa, Portugal
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, Lisboa, Portugal
| | - Joana Rodrigues
- Institute of Pharmacology and Neurosciences, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, Lisboa, Portugal
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, Lisboa, Portugal
| | - Jennifer McGarvey
- School of Biochemistry, Centre for Synaptic Plasticity, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, UK
| | - Catarina Orcinha
- Institute of Pharmacology and Neurosciences, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, Lisboa, Portugal
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, Lisboa, Portugal
- Experimental Epilepsy Research, Department of Neurosurgery, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jeremy M Henley
- School of Biochemistry, Centre for Synaptic Plasticity, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, UK
| | - Ana M Sebastião
- Institute of Pharmacology and Neurosciences, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, Lisboa, Portugal
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Avenida Professor Egas Moniz, Lisboa, Portugal
| |
Collapse
|
9
|
Trollmann R, Mühlberger T, Richter M, Boie G, Feigenspan A, Brackmann F, Jung S. Differential regulation of angiogenesis in the developing mouse brain in response to exogenous activation of the hypoxia-inducible transcription factor system. Brain Res 2018; 1688:91-102. [PMID: 29548688 DOI: 10.1016/j.brainres.2018.03.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 03/09/2018] [Accepted: 03/10/2018] [Indexed: 12/18/2022]
Abstract
Angiogenesis due to hypoxic-ischemic (HI) injury represents a crucial compensatory mechanism of the developing brain that is mainly regulated by hypoxia-inducible transcription factors (HIF). Pharmacological stimulation of HIF is suggested as a neuroprotective option, however, studies of its effects on vascular development are limited. We analyzed the influence of the prolyl-4-hydroxylase inhibitor (PHI), FG-4497, and erythropoietin (rhEPO) on post-hypoxic angiogenesis (angiogenic growth factors, vessel structures) in the developing mouse brain (P7) assessed after a regeneration period of 72 h. Exposure to systemic hypoxia (8% O2, 6 h) was followed by treatment (i.p.) with rhEPO (2500/5000 IU/kg) at 0, 24 and 48 h or FG-4497 (60/100 mg/kg) compared to controls. In response to FG-4497 treatment cortical and hippocampal vessel area and branching were significantly increased compared to controls. This was associated with elevated ANGPT-2 as well as decreased ANGPT-1 and TIE-2 mRNA levels. In response to rhEPO, mildly increased angiogenesis was associated with elevated ANGPT-2 but also TIE-2 mRNA levels in comparison to controls. In conclusion, present data demonstrate a differential regulation of the angiopoietin/TIE-2 system in response to PHI and rhEPO in the post-hypoxic developing brain pointing to potential functional consequences for vascular regeneration and vessel development.
Collapse
Affiliation(s)
- Regina Trollmann
- Department of Pediatrics, Division of Neuropediatrics, Friedrich-Alexander University of Erlangen-Nürnberg, Loschgestrasse 15, 91054 Erlangen, Germany.
| | - Theresa Mühlberger
- Department of Pediatrics, Division of Neuropediatrics, Friedrich-Alexander University of Erlangen-Nürnberg, Loschgestrasse 15, 91054 Erlangen, Germany.
| | - Mandy Richter
- Department of Pediatrics, Division of Neuropediatrics, Friedrich-Alexander University of Erlangen-Nürnberg, Loschgestrasse 15, 91054 Erlangen, Germany.
| | - Gudrun Boie
- Department of Pediatrics, Division of Neuropediatrics, Friedrich-Alexander University of Erlangen-Nürnberg, Loschgestrasse 15, 91054 Erlangen, Germany.
| | - Andreas Feigenspan
- Institute of Animal Physiology, Friedrich-Alexander University of Erlangen-Nürnberg, Staudtstrasse 5, 91058 Erlangen, Germany.
| | - Florian Brackmann
- Department of Pediatrics, Division of Neuropediatrics, Friedrich-Alexander University of Erlangen-Nürnberg, Loschgestrasse 15, 91054 Erlangen, Germany.
| | - Susan Jung
- Department of Pediatrics, Division of Neuropediatrics, Friedrich-Alexander University of Erlangen-Nürnberg, Loschgestrasse 15, 91054 Erlangen, Germany.
| |
Collapse
|
10
|
Zhu L, Huang L, Wen Q, Wang T, Qiao L, Jiang L. Recombinant human erythropoietin offers neuroprotection through inducing endogenous erythropoietin receptor and neuroglobin in a neonatal rat model of periventricular white matter damage. Neurosci Lett 2017; 650:12-17. [PMID: 28359933 DOI: 10.1016/j.neulet.2017.03.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 02/10/2017] [Accepted: 03/15/2017] [Indexed: 01/25/2023]
Abstract
Recombinant human erythropoietin (rh-EPO) has been reported to have protective effects against brain injury. The purpose of this study was to evaluate the levels of erythropoietin receptor (EPOR) and neuroglobin (Ngb) in a neonatal rat model of periventricular white matter damage (PWMD), and to identify the relationship between the two proteins. On postnatal day 3 (P3), rats underwent permanent ligation of the right common carotid artery followed by 6% O2 for 4h (HI) or sham operation and normoxic exposure (sham). Immediately after HI, rats received a single intraperitoneal injection of rh-EPO (5U/g) or saline. We assessed the expression level of Ngb and EPOR on postnatal days 5, 7, 10 and 14. EPOR in the HI rats was initially increased as compared to the sham rats at P5. Subsequently, EPOR expression decreased, but was maintained at a higher level than in sham rats from P7 to P14. In rh-EPO treated rats, the increase in EPOR was greater than in HI rats at P5. However, EPOR levels decreased sharply from P7 to P14. In HI rats, Ngb was increased compared to the sham rats from P5 to P14. Ngb levels were further upregulated after rh-EPO administration from P5 to P10 compared to HI rats. However, this upregulation decreased at P14. In conclusion, this study shows that EPOR and Ngb were upregulated, and both of them act as important coordinated neuroprotectors in rh-EPO treatment of PWMD. However, the two proteins exhibit different expression patterns.
Collapse
Affiliation(s)
- Lihua Zhu
- Institute of Clinical and Nursing, Jiangsu Jiankang Vocational College, 69 Huangshan Ling Road, Pukou District, Nanjing 211800, Jiangsu, China
| | - Li Huang
- Department of Pediatrics, Zhongda Hospital, Southeast University, 87 Dingjia Qiao, Gulou District, Nanjing 210009, Jiangsu, China
| | - Quan Wen
- Department of Pediatrics, Zhongda Hospital, Southeast University, 87 Dingjia Qiao, Gulou District, Nanjing 210009, Jiangsu, China
| | - Ting Wang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipai Lou, Xuanwu District, Nanjing 210096, Jiangsu, China
| | - Lixing Qiao
- Department of Pediatrics, Zhongda Hospital, Southeast University, 87 Dingjia Qiao, Gulou District, Nanjing 210009, Jiangsu, China.
| | - Li Jiang
- Department of Pediatrics, Zhongda Hospital, Southeast University, 87 Dingjia Qiao, Gulou District, Nanjing 210009, Jiangsu, China.
| |
Collapse
|
11
|
Retinal pigment epithelium-secretome: A diabetic retinopathy perspective. Cytokine 2017; 95:126-135. [PMID: 28282610 DOI: 10.1016/j.cyto.2017.02.013] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 01/12/2017] [Accepted: 02/14/2017] [Indexed: 12/31/2022]
Abstract
Diabetic retinopathy is a major complication of diabetes mellitus that can lead to retinal vascular abnormalities and visual impairment. While retinal endothelial pathology is well studied, retinal pigment epithelium (RPE) layer modifications and the patho-physiological regulations are not widely understood. The RPE is a highly specialized pigmented layer regulating not only physiological functions such as transport of nutrients, ions, absorption of light, phagocytosis of photoreceptor membranes, but also secretion of a number of cytokines, chemokines, angiogenic and anti-angiogenic factors. The RPE secretome, though crucial in health and disease, remains elusive in diabetic retinopathy. A knowledge of these secreted factors would help explain and correlate the clinical phase of the disease aiding in improved disease management. A comprehensive knowledge of the secreted factors of the RPE is a potential tool for understanding the differential treatment regime of early diabetic retinopathy, diabetic proliferative retinopathy and diabetic macular edema. In this review, we have delineated the importance of factors secreted by the retinal pigment epithelium and its regulation in the pathogenesis of diabetic retinopathy.
Collapse
|
12
|
Miljus N, Massih B, Weis MA, Rison JV, Bonnas CB, Sillaber I, Ehrenreich H, Geurten BRH, Heinrich R. Neuroprotection and endocytosis: erythropoietin receptors in insect nervous systems. J Neurochem 2017; 141:63-74. [DOI: 10.1111/jnc.13967] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 12/23/2016] [Accepted: 01/24/2017] [Indexed: 12/18/2022]
Affiliation(s)
- Natasa Miljus
- Department of Cellular Neurobiology; Institute for Zoology; University of Goettingen; Goettingen Germany
| | - Bita Massih
- Department of Cellular Neurobiology; Institute for Zoology; University of Goettingen; Goettingen Germany
| | - Marissa A. Weis
- Department of Cellular Neurobiology; Institute for Zoology; University of Goettingen; Goettingen Germany
| | - Jan Vincent Rison
- Department of Cellular Neurobiology; Institute for Zoology; University of Goettingen; Goettingen Germany
| | | | | | - Hannelore Ehrenreich
- Clinical Neuroscience; Max Planck Institute of Experimental Medicine; Goettingen Germany
- DFG Center for Nanoscale Microscopy & Molecular Physiology of the Brain (CNMPB); Goettingen Germany
| | - Bart R. H. Geurten
- Department of Cellular Neurobiology; Institute for Zoology; University of Goettingen; Goettingen Germany
| | - Ralf Heinrich
- Department of Cellular Neurobiology; Institute for Zoology; University of Goettingen; Goettingen Germany
| |
Collapse
|
13
|
Ogier M, Belmeguenai A, Lieutaud T, Georges B, Bouvard S, Carré E, Canini F, Bezin L. Cognitive Deficits and Inflammatory Response Resulting from Mild-to-Moderate Traumatic Brain Injury in Rats Are Exacerbated by Repeated Pre-Exposure to an Innate Stress Stimulus. J Neurotrauma 2017; 34:1645-1657. [PMID: 27901414 DOI: 10.1089/neu.2016.4741] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Traumatic brain injury (TBI) is common in both military and civilian populations, and often results in neurobehavioral sequelae that impair quality of life in both patients and their families. Although individuals who are chronically exposed to stress are more likely to experience TBI, it is still unknown whether pre-injury stress influences the outcome after TBI. The present study tested whether behavioral and cognitive long-term outcome after TBI in rats is affected by prior exposure to an innate stress stimulus. Young adult male Sprague-Dawley rats were exposed to the predator odor 2,5-dihydro-2,4,5-trimethylthiazoline (TMT) or to water (WAT); exposure was repeated eight times at irregular intervals over a 2-week period. Rats were subsequently subjected to either mild-to-moderate bilateral brain injury (lateral fluid percussion [LFP]) or sham surgery (Sham). Four experimental groups were studied: Sham-WAT, Sham-TMT, LFP-WAT and LFP-TMT. Compared with Sham-WAT rats, LFP-WAT rats exhibited transient locomotor hyperactivity without signs of anxiety, minor spatial learning acquisition and hippocampal long-term potentiation deficits, and lower baseline activity of the hypothalamic-pituitary-adrenal axis with slightly stronger reactivity to restraint stress. Exposure to TMT had only negligible effects on Sham rats, whereas it exacerbated all deficits in LFP rats except for locomotor hyperactivity. Early brain inflammatory response (8 h post-trauma) was aggravated in rats pre-exposed to TMT, suggesting that increased brain inflammation may sustain functional deficits in these rats. Hence, these data suggest that pre-exposure to stressful conditions can aggravate long-term deficits induced by TBI, leading to severe stress response deficits, possibly due to dysregulated inflammatory response.
Collapse
Affiliation(s)
- Michaël Ogier
- 1 Institut de Recherche Biomédicale des Armées , Brétigny-sur-Orge, France .,2 Université Claude Bernard Lyon 1 , Bron, France .,3 Institute for Epilepsy , IDÉE, Bron, France
| | - Amor Belmeguenai
- 2 Université Claude Bernard Lyon 1 , Bron, France .,3 Institute for Epilepsy , IDÉE, Bron, France
| | - Thomas Lieutaud
- 2 Université Claude Bernard Lyon 1 , Bron, France .,3 Institute for Epilepsy , IDÉE, Bron, France
| | - Béatrice Georges
- 2 Université Claude Bernard Lyon 1 , Bron, France .,3 Institute for Epilepsy , IDÉE, Bron, France
| | - Sandrine Bouvard
- 2 Université Claude Bernard Lyon 1 , Bron, France .,3 Institute for Epilepsy , IDÉE, Bron, France
| | - Emilie Carré
- 1 Institut de Recherche Biomédicale des Armées , Brétigny-sur-Orge, France
| | - Frédéric Canini
- 1 Institut de Recherche Biomédicale des Armées , Brétigny-sur-Orge, France .,4 Ecole du Val de Grâce , Paris, France
| | - Laurent Bezin
- 2 Université Claude Bernard Lyon 1 , Bron, France .,3 Institute for Epilepsy , IDÉE, Bron, France
| |
Collapse
|
14
|
Carbamylated erythropoietin enhances mice ventilatory responses to changes in O2 but not CO2 levels. Respir Physiol Neurobiol 2016; 232:1-12. [PMID: 27317882 DOI: 10.1016/j.resp.2016.06.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 06/14/2016] [Accepted: 06/15/2016] [Indexed: 11/21/2022]
Abstract
Erythropoietin (EPO) has beneficial tissue-protective effects in several diseases but erythrocytosis may cause deleterious effects in EPO-treated patients. Thus carbamylated-EPO (C-EPO) and other derivatives retaining tissue-protective but lacking bone marrow-stimulating actions have been developed. Although EPO modulates ventilatory responses, the effects of C-EPO on ventilation have not been investigated. Here, basal breathing and respiratory chemoreflexes were measured by plethysmography after acute and chronic treatments with recombinant human C-EPO (rhC-EPO; 15,000 IU/kg during 5days) or saline (control group). Hematocrit, plasma and brainstem rhC-EPO levels were also quantified. Chronic rhC-EPO significantly elevated tissue rhC-EPO levels but not hematocrit. None of the drug regimen altered basal ventilation (normoxia). Chronic but not acute rhC-EPO enhanced hyperoxic ventilatory depression, and sustained the hypoxic ventilatory response mainly via a reduction of the roll-off phase. By contrast, rhC-EPO did not blunt the ventilatory response to hypercapnia. Thus, chronic C-EPO may be a promising therapy to improve breathing during hypoxia while minimizing adverse effects on cardiovascular function.
Collapse
|
15
|
Erythropoietin Pathway: A Potential Target for the Treatment of Depression. Int J Mol Sci 2016; 17:ijms17050677. [PMID: 27164096 PMCID: PMC4881503 DOI: 10.3390/ijms17050677] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Revised: 04/05/2016] [Accepted: 04/27/2016] [Indexed: 12/21/2022] Open
Abstract
During the past decade, accumulating evidence from both clinical and experimental studies has indicated that erythropoietin may have antidepressant effects. In addition to the kidney and liver, many organs have been identified as secretory tissues for erythropoietin, including the brain. Its receptor is expressed in cerebral and spinal cord neurons, the hypothalamus, hippocampus, neocortex, dorsal root ganglia, nerve axons, and Schwann cells. These findings may highlight new functions for erythropoietin, which was originally considered to play a crucial role in the progress of erythroid differentiation. Erythropoietin and its receptor signaling through JAK2 activate multiple downstream signaling pathways including STAT5, PI3K/Akt, NF-κB, and MAPK. These factors may play an important role in inflammation and neuroprogression in the nervous system. This is particularly true for the hippocampus, which is possibly related to learning, memory, neurocognitive deficits and mood alterations. Thus, the influence of erythropoietin on the downstream pathways known to be involved in the treatment of depression makes the erythropoietin-related pathway an attractive target for the development of new therapeutic approaches. Focusing on erythropoietin may help us understand the pathogenic mechanisms of depression and the molecular basis of its treatment.
Collapse
|
16
|
Rangarajan V, Juul SE. Erythropoietin: emerging role of erythropoietin in neonatal neuroprotection. Pediatr Neurol 2014; 51:481-8. [PMID: 25266611 PMCID: PMC4180944 DOI: 10.1016/j.pediatrneurol.2014.06.008] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 06/09/2014] [Accepted: 06/12/2014] [Indexed: 01/27/2023]
Abstract
BACKGROUND In the last two decades, there has been considerable evolution in understanding the role of erythropoietin in neuroprotection. Erythropoietin has both paracrine and autocrine functions in the brain. Erythropoietin binding results in neurogenesis, oligodendrogenesis, and angiogenesis. Erythropoietin and its receptor are upregulated by exposure to hypoxia and proinflammatory cytokines after brain injury. While erythropoietin aids in recovery of locally injured neuronal cells, it provides negative feedback to glial cells in the penumbra, thereby limiting extension of injury. This forms the rationale for use of recombinant erythropoietin and erythropoietin mimetics in neonatal and adult injury models of stroke, traumatic brain injury, spinal cord injury, intracerebral hemorrhage, and neonatal hypoxic ischemia. METHOD Review of published literature (Pubmed, Medline, and Google scholar). RESULTS Preclinical neuroprotective data are reviewed, and the rationale for proceeding to clinical trials is discussed. Results from phase I/II trials are presented, as are updates on ongoing and upcoming clinical trials of erythropoietin neuroprotection in neonatal populations. CONCLUSIONS The scientific rationale and preclinical data for erythropoietin neuroprotection are promising. Phase II and III clinical trials are currently in process to determine the safety and efficacy of neuroprotective dosing of erythropoietin for extreme prematurity and hypoxic-ischemic encephalopathy in neonates.
Collapse
Affiliation(s)
- Vijayeta Rangarajan
- Division of Neonatology, Department of Pediatrics, University of Washington, Seattle, Washington
| | - Sandra E Juul
- Division of Neonatology, Department of Pediatrics, University of Washington, Seattle, Washington.
| |
Collapse
|
17
|
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: 85] [Impact Index Per Article: 7.7] [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
|
18
|
Zhang J, Wang Q, Xiang H, Xin Y, Chang M, Lu H. Neuroprotection with erythropoietin in preterm and/or low birth weight infants. J Clin Neurosci 2014; 21:1283-7. [PMID: 24650681 DOI: 10.1016/j.jocn.2013.10.040] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 10/13/2013] [Accepted: 10/27/2013] [Indexed: 12/01/2022]
Abstract
Neonatal brain injury caused by extreme prematurity remains a great challenge for prevention. Erythropoietin (EPO) has shown neuroprotective effects in a series of neonatal experimental models and recent clinical trials of premature infants. In this meta-analysis of seven clinical trials, EPO was associated with a highly reproducible reduction in the risk of neurodevelopmental disability in preterm infants. However, there was no difference in the risk for morbidity, cerebral palsy, visual deficit, severe hearing deficit, necrotizing enterocolitis, intracranial hemorrhage and patent ductus arteriosus. The use of EPO, to some extent, is associated with reduction in neurodevelopmental disability in preterm infants. More double blind randomized controlled trials are needed to establish the best therapeutic approach for neuroprotection in preterm infants.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang 212001, Jiangsu Province, PR China
| | - Qiuxia Wang
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang 212001, Jiangsu Province, PR China
| | - Hong Xiang
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang 212001, Jiangsu Province, PR China
| | - Yue Xin
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang 212001, Jiangsu Province, PR China
| | - Ming Chang
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang 212001, Jiangsu Province, PR China
| | - Hongyan Lu
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang 212001, Jiangsu Province, PR China.
| |
Collapse
|
19
|
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
|
20
|
Hayley S, Litteljohn D. Neuroplasticity and the next wave of antidepressant strategies. Front Cell Neurosci 2013; 7:218. [PMID: 24312008 PMCID: PMC3834236 DOI: 10.3389/fncel.2013.00218] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 10/29/2013] [Indexed: 12/13/2022] Open
Abstract
Depression is a common chronic psychiatric disorder that is also often co-morbid with numerous neurological and immune diseases. Accumulating evidence indicates that disturbances of neuroplasticity occur with depression, including reductions of hippocampal neurogenesis and cortical synaptogenesis. Improper trophic support stemming from stressor-induced reductions of growth factors, most notably brain derived neurotrophic factor (BDNF), likely drives such aberrant neuroplasticity. We posit that psychological and immune stressors can interact upon a vulnerable genetic background to promote depression by disturbing BDNF and neuroplastic processes. Furthermore, the chronic and commonly relapsing nature of depression is suggested to stem from "faulty wiring" of emotional circuits driven by neuroplastic aberrations. The present review considers depression in such terms and attempts to integrate the available evidence indicating that the efficacy of current and "next wave" antidepressant treatments, whether used alone or in combination, is at least partially tied to their ability to modulate neuroplasticity. We particularly focus on the N-methyl-D-aspartate (NMDA) antagonist, ketamine, which already has well documented rapid antidepressant effects, and the trophic cytokine, erythropoietin (EPO), which we propose as a potential adjunctive antidepressant agent.
Collapse
Affiliation(s)
- Shawn Hayley
- Department of Neuroscience, Carleton University Ottawa, ON, Canada
| | | |
Collapse
|
21
|
Osborn M, Rustom N, Clarke M, Litteljohn D, Rudyk C, Anisman H, Hayley S. Antidepressant-like effects of erythropoietin: a focus on behavioural and hippocampal processes. PLoS One 2013; 8:e72813. [PMID: 24019878 PMCID: PMC3760922 DOI: 10.1371/journal.pone.0072813] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 07/19/2013] [Indexed: 12/11/2022] Open
Abstract
Depression is a chronic and debilitating condition with a significant degree of relapse and treatment resistance that could stem, at least in part, from disturbances of neuroplasticity. This has led to an increased focus on treatment strategies that target brain derived neurotrophic factor (BDNF), synaptic plasticity and adult neurogenesis. In the current study we aimed to assess whether erythropoietin (EPO) would have antidepressant-like effects given its already established pro-trophic actions. In particular, we assessed whether EPO would diminish the deleterious effects of a social stressor in mice. Indeed, EPO induced anxiolytic and antidepressant-like responses in a forced swim test, open field, elevated-plus maze, and a novelty test, and appeared to blunt some of the negative behavioural effects of a social stressor. Furthermore, EPO promoted adult hippocampal neurogenesis, an important feature of effective antidepressants. Finally, a separate study using the mTOR inhibitor rapamycin revealed that antagonizing this pathway prevented the impact of EPO upon forced swim performance. These data are consistent with previous findings showing that the mTOR pathway and its neurogenic and synaptogenic effects might mediate the behavioral consequences of antidepressant agents. Our findings further highlight EPO as a possible adjunct treatment for affective disorders, as well as other stressor associated disorders of impaired neuroplasticity.
Collapse
Affiliation(s)
- Meagan Osborn
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Nazneen Rustom
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Melanie Clarke
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Darcy Litteljohn
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Chris Rudyk
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Hymie Anisman
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Shawn Hayley
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
- * E-mail:
| |
Collapse
|
22
|
Chamorro ME, Wenker SD, Vota DM, Vittori DC, Nesse AB. Signaling pathways of cell proliferation are involved in the differential effect of erythropoietin and its carbamylated derivative. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:1960-8. [PMID: 23602701 DOI: 10.1016/j.bbamcr.2013.04.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 03/30/2013] [Accepted: 04/08/2013] [Indexed: 12/20/2022]
Abstract
It is now recognized that in addition to its activity upon erythroid progenitor cells, erythropoietin (Epo) is capable of stimulating survival of different non-erythroid cells. Since stimulation of erythropoiesis is unwanted for neuroprotection, Epo-like compounds with a more selective action are under investigation. Although the carbamylated derivative of erythropoietin (cEpo) has demonstrated non-hematopoietic tissue protection without erythropoietic effect, little is known about differential mechanisms between Epo and cEpo. Therefore, we investigated signaling pathways which play a key role in Epo-induced proliferation. Here we show that cEpo blocked FOXO3a phosphorylation, allowing expression of downstream target p27(kip1) in UT-7 and TF-1 cells capable of erythroid differentiation. This is consistent with the involvement of cEpo in slowing down G1-to-S-phase progression compared with the effect of Epo upon cell cycle. In contrast, similar antiapoptotic actions of cEpo and Epo were observed in neuronal SH-SY5Y cells. Inhibition and competition assays suggest that Epo may act through both, the homodimeric (EpoR/EpoR) and the heterodimeric (EpoR/βcR) receptors in neuronal SH-SY5Y cells and probably in the TF-1 cell type as well. Results also indicate that cEpo needs both the EpoR and βcR subunits to prevent apoptosis of neuronal cells. Based on evidence suggesting that cell proliferation pathways were involved in the differential effect of Epo and cEpo, we went forward to studying downstream signals. Here we provide the first evidence that unlike Epo, cEpo failed to induce FOXO3a inactivation and subsequent p27(kip1) downregulation, which is clearly shown in the incapacity of cEpo to induce erythroid cell growth.
Collapse
|
23
|
Kako E, Kaneko N, Aoyama M, Hida H, Takebayashi H, Ikenaka K, Asai K, Togari H, Sobue K, Sawamoto K. Subventricular zone-derived oligodendrogenesis in injured neonatal white matter in mice enhanced by a nonerythropoietic erythropoietin derivative. Stem Cells 2013; 30:2234-47. [PMID: 22890889 DOI: 10.1002/stem.1202] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Perinatal hypoxia-ischemia (HI) frequently causes white-matter injury, leading to severe neurological deficits and mortality, and only limited therapeutic options exist. The white matter of animal models and human patients with HI-induced brain injury contains increased numbers of oligodendrocyte progenitor cells (OPCs). However, the origin and fates of these OPCs and their potential to repair injured white matter remain unclear. Here, using cell-type- and region-specific genetic labeling methods in a mouse HI model, we characterized the Olig2-expressing OPCs. We found that after HI, Olig2+ cells increased in the posterior part of the subventricular zone (pSVZ) and migrated into the injured white matter. However, their oligodendrocytic differentiation efficiency was severely compromised compared with the OPCs in normal tissue, indicating the need for an intervention to promote their differentiation. Erythropoietin (EPO) treatment is a promising candidate, but it has detrimental effects that preclude its clinical use for brain injury. We found that long-term postinjury treatment with a nonerythropoietic derivative of EPO, asialo-erythropoietin, promoted the maturation of pSVZ-derived OPCs and the recovery of neurological function, without affecting hematopoiesis. These results demonstrate the limitation and potential of endogenous OPCs in the pSVZ as a therapeutic target for treating neonatal white-matter injury.
Collapse
Affiliation(s)
- Eisuke Kako
- Department of Developmental and Regenerative Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Fares RP, Belmeguenai A, Sanchez PE, Kouchi HY, Bodennec J, Morales A, Georges B, Bonnet C, Bouvard S, Sloviter RS, Bezin L. Standardized environmental enrichment supports enhanced brain plasticity in healthy rats and prevents cognitive impairment in epileptic rats. PLoS One 2013; 8:e53888. [PMID: 23342033 PMCID: PMC3544705 DOI: 10.1371/journal.pone.0053888] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Accepted: 12/04/2012] [Indexed: 12/23/2022] Open
Abstract
Environmental enrichment of laboratory animals influences brain plasticity, stimulates neurogenesis, increases neurotrophic factor expression, and protects against the effects of brain insult. However, these positive effects are not constantly observed, probably because standardized procedures of environmental enrichment are lacking. Therefore, we engineered an enriched cage (the Marlau™ cage), which offers: (1) minimally stressful social interactions; (2) increased voluntary exercise; (3) multiple entertaining activities; (4) cognitive stimulation (maze exploration), and (5) novelty (maze configuration changed three times a week). The maze, which separates food pellet and water bottle compartments, guarantees cognitive stimulation for all animals. Compared to rats raised in groups in conventional cages, rats housed in Marlau™ cages exhibited increased cortical thickness, hippocampal neurogenesis and hippocampal levels of transcripts encoding various genes involved in tissue plasticity and remodeling. In addition, rats housed in Marlau™ cages exhibited better performances in learning and memory, decreased anxiety-associated behaviors, and better recovery of basal plasma corticosterone level after acute restraint stress. Marlau™ cages also insure inter-experiment reproducibility in spatial learning and brain gene expression assays. Finally, housing rats in Marlau™ cages after severe status epilepticus at weaning prevents the cognitive impairment observed in rats subjected to the same insult and then housed in conventional cages. By providing a standardized enriched environment for rodents during housing, the Marlau™ cage should facilitate the uniformity of environmental enrichment across laboratories.
Collapse
MESH Headings
- Adaptation, Psychological/physiology
- Animals
- Anxiety/complications
- Body Weight
- Brain/cytology
- Brain/pathology
- Brain/physiology
- Brain/physiopathology
- CA1 Region, Hippocampal/cytology
- CA1 Region, Hippocampal/pathology
- CA1 Region, Hippocampal/physiology
- CA1 Region, Hippocampal/physiopathology
- Cognition
- Eating
- Exploratory Behavior/physiology
- Health
- Housing, Animal/standards
- Lipid Metabolism
- Male
- Neurogenesis/genetics
- Neuronal Plasticity
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Reference Standards
- Reproducibility of Results
- Restraint, Physical/psychology
- Status Epilepticus/pathology
- Status Epilepticus/physiopathology
- Status Epilepticus/psychology
- Stress, Psychological/complications
- Stress, Psychological/pathology
- Stress, Psychological/physiopathology
- Synapses/pathology
Collapse
Affiliation(s)
- Raafat P. Fares
- Université de Lyon, Lyon, France
- Université Lyon 1, Villeurbanne, France
- Inserm, Institut National de la Santé et de la Recherche Médicale, U1028, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- CNRS, Centre National de la Recherche Scientifique, UMR5292, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- IDÉE, Institut Des ÉpilepsiEs, Lyon, France
- IRBA, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| | - Amor Belmeguenai
- Université de Lyon, Lyon, France
- Université Lyon 1, Villeurbanne, France
- Inserm, Institut National de la Santé et de la Recherche Médicale, U1028, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- CNRS, Centre National de la Recherche Scientifique, UMR5292, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- IDÉE, Institut Des ÉpilepsiEs, Lyon, France
| | - Pascal E. Sanchez
- Université de Lyon, Lyon, France
- Université Lyon 1, Villeurbanne, France
- Inserm, Institut National de la Santé et de la Recherche Médicale, U1028, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- CNRS, Centre National de la Recherche Scientifique, UMR5292, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- IDÉE, Institut Des ÉpilepsiEs, Lyon, France
| | - Hayet Y. Kouchi
- Université de Lyon, Lyon, France
- Université Lyon 1, Villeurbanne, France
- Inserm, Institut National de la Santé et de la Recherche Médicale, U1028, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- CNRS, Centre National de la Recherche Scientifique, UMR5292, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- IDÉE, Institut Des ÉpilepsiEs, Lyon, France
| | - Jacques Bodennec
- Université de Lyon, Lyon, France
- Université Lyon 1, Villeurbanne, France
- Inserm, Institut National de la Santé et de la Recherche Médicale, U1028, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- CNRS, Centre National de la Recherche Scientifique, UMR5292, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- IDÉE, Institut Des ÉpilepsiEs, Lyon, France
| | - Anne Morales
- Université de Lyon, Lyon, France
- Université Lyon 1, Villeurbanne, France
- Inserm, Institut National de la Santé et de la Recherche Médicale, U1028, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- CNRS, Centre National de la Recherche Scientifique, UMR5292, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- IDÉE, Institut Des ÉpilepsiEs, Lyon, France
| | - Béatrice Georges
- Université de Lyon, Lyon, France
- Université Lyon 1, Villeurbanne, France
- Inserm, Institut National de la Santé et de la Recherche Médicale, U1028, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- CNRS, Centre National de la Recherche Scientifique, UMR5292, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- IDÉE, Institut Des ÉpilepsiEs, Lyon, France
| | - Chantal Bonnet
- Université de Lyon, Lyon, France
- Université Lyon 1, Villeurbanne, France
- Inserm, Institut National de la Santé et de la Recherche Médicale, U1028, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- CNRS, Centre National de la Recherche Scientifique, UMR5292, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- IDÉE, Institut Des ÉpilepsiEs, Lyon, France
| | - Sandrine Bouvard
- Université de Lyon, Lyon, France
- Université Lyon 1, Villeurbanne, France
- Inserm, Institut National de la Santé et de la Recherche Médicale, U1028, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- CNRS, Centre National de la Recherche Scientifique, UMR5292, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- IDÉE, Institut Des ÉpilepsiEs, Lyon, France
| | - Robert S. Sloviter
- Department of Neurobiology, Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Laurent Bezin
- Université de Lyon, Lyon, France
- Université Lyon 1, Villeurbanne, France
- Inserm, Institut National de la Santé et de la Recherche Médicale, U1028, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- CNRS, Centre National de la Recherche Scientifique, UMR5292, Lyon Neuroscience Research Center, Tiger Team, Lyon, France
- IDÉE, Institut Des ÉpilepsiEs, Lyon, France
- * E-mail:
| |
Collapse
|
25
|
Baudin A, Blot K, Verney C, Estevez L, Santamaria J, Gressens P, Giros B, Otani S, Daugé V, Naudon L. Maternal deprivation induces deficits in temporal memory and cognitive flexibility and exaggerates synaptic plasticity in the rat medial prefrontal cortex. Neurobiol Learn Mem 2012; 98:207-14. [PMID: 22922490 DOI: 10.1016/j.nlm.2012.08.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Revised: 07/27/2012] [Accepted: 08/12/2012] [Indexed: 01/18/2023]
Abstract
Early life adverse events can lead to structural and functional impairments in the prefrontal cortex (PFC). Here, we investigated whether maternal deprivation (MD) alters PFC-dependent executive functions, neurons and astrocytes number and synaptic plasticity in adult male Long-Evans rats. The deprivation protocol consisted of a daily separation of newborn Long-Evans pups from their mothers and littermates 3h/day postnatal day 1-14. Cognitive performances were assessed in adulthood using the temporal order memory task (TMT) and the attentional set-shifting task (ASST) that principally implicates the PFC and the Morris water maze task (WMT) that does not essentially rely on the PFC. The neurons and astrocytes of the prelimbic (PrL) area of the medial PFC (mPFC) were immunolabelled respectively with anti-NeuN and anti-GFAP antibodies and quantified by stereology. The field potentials evoked by electrical stimulation of ventral hippocampus (ventral HPC) were recorded in vivo in the PrL area. In adulthood, MD produced cognitive deficits in two PFC-dependent tasks, the TMT and ASST, but not in the WMT. In parallel, MD induced in the prelimbic area of the medial PFC an upregulation of long-term potentiation (LTP), without any change in the number of neurons and astrocytes. We provide evidence that MD leads in adults to an alteration of the cognitive abilities dependent on the PFC, and to an exaggerated synaptic plasticity in this region. We suggest that this latter phenomenon may contribute to the impairments in the cognitive tasks.
Collapse
Affiliation(s)
- Aurélie Baudin
- INSERM, UMRs, Physiopathologie des Maladies du Système Nerveux Central, Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Erythropoietin (Epo) is an essential hormone that binds and activates the Epo receptor (EpoR) resident on the surface of erythroid progenitor cells, thereby promoting erythropoiesis. Recombinant human erythropoietin has been used successfully for over 20 years to treat anemia in millions of patients. In addition to erythropoiesis, Epo has also been reported to have other effects, such as tissue protection and promotion of tumor cell growth or survival. This became of significant concern in 2003, when some clinical trials in cancer patients reported increased tumor progression and worse survival outcomes in patients treated with erythropoiesis-stimulating agents (ESAs). One of the potential mechanisms proffered to explain the observed safety issues was that functional EpoR was expressed in tumors and/or endothelial cells, and that ESAs directly stimulated tumor growth and/or antagonized tumor ablative therapies. Since then, numerous groups have performed further research evaluating this potential mechanism with conflicting data and conclusions. Here, we review the biology of endogenous Epo and EpoR expression and function in erythropoiesis, and evaluate the evidence pertaining to the expression of EpoR on normal nonhematopoietic and tumor cells.
Collapse
|
27
|
Pankratova S, Gu B, Kiryushko D, Korshunova I, Køhler LB, Rathje M, Bock E, Berezin V. A new agonist of the erythropoietin receptor, Epobis, induces neurite outgrowth and promotes neuronal survival. J Neurochem 2012; 121:915-23. [DOI: 10.1111/j.1471-4159.2012.07751.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
28
|
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: 30] [Impact Index Per Article: 2.3] [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
|
29
|
Tan H, Kang X, Zhong Y, Shen X, Cheng Y, Jiao Q, Deng L. Erythropoietin upregulates growth associated protein-43 expression and promotes retinal ganglion cell axonal regeneration in vivo after optic nerve crush. Neural Regen Res 2012; 7:295-301. [PMID: 25806072 PMCID: PMC4353103 DOI: 10.3969/j.issn.1673-5374.2012.04.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Accepted: 12/01/2011] [Indexed: 12/22/2022] Open
Abstract
In this study, we established a rat model of optic nerve crush to explore the effects of erythropoietin on retinal ganglion cell axonal regeneration. At 15 days after injury in erythropoietin treated rats, retinal ganglion cell densities in regions corresponding to the 1/6, 3/6 and 5/6 ratios of the retinal radius were significantly increased. In addition, the number of growth associated protein-43 positive axons was significantly increased at different distances (50, 250 and 500 μm) from the crush site after erythropoietin treatment. Erythropoietin significantly increased growth associated protein-43 protein levels in the retina after crush injury, as determined by western blot and immunofluorescence analysis. These results demonstrate that erythropoietin protects injured retinal ganglion cells and promotes axonal regeneration.
Collapse
Affiliation(s)
- Haibo Tan
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xin Kang
- Department of Clinical Pharmacology, Changhai Hospital Affiliated to Second Military Medical University, Shanghai 200433, China
| | - Yisheng Zhong
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xi Shen
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yu Cheng
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qin Jiao
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lianfu Deng
- Shanghai Institute of Traumatology and Orthopedics, Shanghai 200025, China
| |
Collapse
|
30
|
Recombinant human erythropoietin suppresses endothelial cell apoptosis and reduces the ratio of Bax to Bcl-2 proteins in the aortas of apolipoprotein E-deficient mice. J Cardiovasc Pharmacol 2011; 57:424-33. [PMID: 21242808 DOI: 10.1097/fjc.0b013e31820d92fd] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Recent clinical trials have raised concern that therapy with recombinant human erythropoietin (EPO) may increase cardiovascular disease risk, event rate, and mortality. Endothelial cell apoptosis has been implicated in both atherogenesis and in the destabilization and rupture of atheromatous plaques. In the current study, we observed that EPO and the EPO-mimetic peptide EMP-1 markedly suppressed lipopolysaccharide-induced apoptosis in endothelial cell monolayers. Therapeutic concentrations of EPO upregulated Bcl-2 expression and concurrently diminished expression of Bax, resulting in a net decrease in the ratio of Bax to Bcl-2 protein concentrations. In vivo studies demonstrated that EPO receptor is abundantly expressed in murine aorta and that EPO treatment for 10 weeks markedly decreased the ratio of Bax to Bcl-2 protein in the aortas of apolipoprotein E-deficient mice fed a high-fat diet. To our knowledge, these data are the first to reveal a modulation of regulators of the apoptotic pathway in murine aorta by chronic EPO treatment. These observations imply that long-term administration of EPO may have the potential to affect plaque stability.
Collapse
|
31
|
Colella P, Iodice C, Di Vicino U, Annunziata I, Surace EM, Auricchio A. Non-erythropoietic erythropoietin derivatives protect from light-induced and genetic photoreceptor degeneration. Hum Mol Genet 2011; 20:2251-62. [PMID: 21421996 PMCID: PMC3090200 DOI: 10.1093/hmg/ddr115] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Revised: 03/01/2011] [Accepted: 03/15/2011] [Indexed: 11/21/2022] Open
Abstract
Given the high genetic heterogeneity of inherited retinal degenerations (IRDs), a wide applicable treatment would be desirable to halt/slow progressive photoreceptor (PR) cell loss in a mutation-independent manner. In addition to its erythropoietic activity, erythropoietin (EPO) presents neurotrophic characteristics. We have previously shown that adeno-associated viral (AAV) vector-mediated systemic EPO delivery protects from PR degeneration. However, this is associated with an undesired hematocrit increase that could contribute to PR protection. Non-erythropoietic EPO derivatives (EPO-D) are available which allow us to dissect erythropoiesis's role in PR preservation and may be more versatile and safe than EPO as anti-apoptotic agents. We delivered in animal models of light-induced or genetic retinal degeneration either intramuscularly or subretinally AAV vectors encoding EPO or one of the three selected EPO-D: the mutant S100E, the helix A- and B-derived EPO-mimetic peptides. We observed that (i) systemic expression of S100E induces a significantly lower hematocrit increase than EPO and provides similar protection from PR degeneration, and (ii) intraocular expression of EPO-D protects PR from degeneration in the absence of significant hematocrit increase. On the basis of this, we conclude that erythropoiesis is not required for EPO-mediated PR protection. However, the lower efficacy observed when EPO or S100E is expressed intraocularly rather than systemically suggests that hormone systemic effects contribute to PR protection. Unlike S100E, EPO-mimetic peptides preserve PR only when given locally, suggesting that different EPO-D have a different potency or mode of action. In conclusion, our data show that subretinal delivery of AAV vectors encoding EPO-D protects from light-induced and genetic PR degeneration.
Collapse
Affiliation(s)
- Pasqualina Colella
- Telethon Institute of Genetics and Medicine (TIGEM), 80131 Naples, Italy
- The Open University, Milton Keynes, UK and
| | - Carolina Iodice
- Telethon Institute of Genetics and Medicine (TIGEM), 80131 Naples, Italy
| | - Umberto Di Vicino
- Telethon Institute of Genetics and Medicine (TIGEM), 80131 Naples, Italy
| | - Ida Annunziata
- Telethon Institute of Genetics and Medicine (TIGEM), 80131 Naples, Italy
| | - Enrico M. Surace
- Telethon Institute of Genetics and Medicine (TIGEM), 80131 Naples, Italy
| | - Alberto Auricchio
- Telethon Institute of Genetics and Medicine (TIGEM), 80131 Naples, Italy
- Medical Genetics, Department of Pediatrics, Federico II University, Naples, Italy
| |
Collapse
|
32
|
Schober ME, Block B, Beachy JC, Statler KD, Giza CC, Lane RH. Early and sustained increase in the expression of hippocampal IGF-1, but not EPO, in a developmental rodent model of traumatic brain injury. J Neurotrauma 2011; 27:2011-20. [PMID: 20822461 DOI: 10.1089/neu.2009.1226] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Pediatric traumatic brain injury (pTBI) is the leading cause of traumatic death and disability in children in the United States. Impaired learning and memory in these young survivors imposes a heavy toll on society. In adult TBI (aTBI) models, cognitive outcome improved after administration of erythropoietin (EPO) or insulin-like growth factor-1 (IGF-1). Little is known about the production of these agents in the hippocampus, a brain region critical for learning and memory, after pTBI. Our objective was to describe hippocampal expression of EPO and IGF-1, together with their receptors (EPOR and IGF-1R, respectively), over time after pTBI in 17-day-old rats. We used the controlled cortical impact (CCI) model and measured hippocampal mRNA levels of EPO, IGF-1, EPOR, IGF-1R, and markers of caspase-dependent apoptosis (bcl2, bax, and p53) at post-injury days (PID) 1, 2, 3, 7, and 14. CCI rats performed poorly on Morris water maze testing of spatial working memory, a hippocampally-based cognitive function. Apoptotic markers were present early and persisted for the duration of the study. EPO in our pTBI model increased much later (PID7) than in aTBI models (12 h), while EPOR and IGF-1 increased at PID1 and PID2, respectively, similar to data from aTBI models. Our data indicate that EPO expression showed a delayed upregulation post-pTBI, while EPOR increased early. We speculate that administration of EPO in the first 1-2 days after pTBI would increase hippocampal neuronal survival and function.
Collapse
Affiliation(s)
- Michelle E Schober
- Department of Pediatrics, Division of Critical Care, University of Utah, Salt Lake City, Utah 84158, USA.
| | | | | | | | | | | |
Collapse
|
33
|
Maiese K, Chong ZZ, Shang YC, Hou J. Novel avenues of drug discovery and biomarkers for diabetes mellitus. J Clin Pharmacol 2011; 51:128-52. [PMID: 20220043 PMCID: PMC3033756 DOI: 10.1177/0091270010362904] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Globally, developed nations spend a significant amount of their resources on health care initiatives that poorly translate into increased population life expectancy. As an example, the United States devotes 16% of its gross domestic product to health care, the highest level in the world, but falls behind other nations that enjoy greater individual life expectancy. These observations point to the need for pioneering avenues of drug discovery to increase life span with controlled costs. In particular, innovative drug development for metabolic disorders such as diabetes mellitus becomes increasingly critical given that the number of diabetic people will increase exponentially over the next 20 years. This article discusses the elucidation and targeting of novel cellular pathways that are intimately tied to oxidative stress in diabetes mellitus for new treatment strategies. Pathways that involve wingless, β-nicotinamide adenine dinucleotide (NAD(+)) precursors, and cytokines govern complex biological pathways that determine both cell survival and longevity during diabetes mellitus and its complications. Furthermore, the role of these entities as biomarkers for disease can further enhance their utility irrespective of their treatment potential. Greater understanding of the intricacies of these unique cellular mechanisms will shape future drug discovery for diabetes mellitus to provide focused clinical care with limited or absent long-term complications.
Collapse
Affiliation(s)
- Kenneth Maiese
- Department of Neurology, 8C-1 UHC, Wayne State University School of Medicine, 4201 St. Antoine, Detroit, MI 48201, USA.
| | | | | | | |
Collapse
|
34
|
Xiong T, Qu Y, Mu D, Ferriero D. Erythropoietin for neonatal brain injury: opportunity and challenge. Int J Dev Neurosci 2011; 29:583-91. [DOI: 10.1016/j.ijdevneu.2010.12.007] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2010] [Revised: 12/03/2010] [Accepted: 12/30/2010] [Indexed: 02/04/2023] Open
Affiliation(s)
- Tao Xiong
- Department of PediatricsWest China Second University HospitalSichuan UniversityChengduChina
- Department of Newborn MedicineAffiliated Hospital of Luzhou Medical CollegeLuzhouChina
| | - Yi Qu
- Department of PediatricsWest China Second University HospitalSichuan UniversityChengduChina
| | - Dezhi Mu
- Department of PediatricsWest China Second University HospitalSichuan UniversityChengduChina
- Department of NeurologyNewborn Brain Research InstituteUniversity of CaliforniaSan FranciscoCAUSA
| | - Donna Ferriero
- Department of NeurologyNewborn Brain Research InstituteUniversity of CaliforniaSan FranciscoCAUSA
- Department of PediatricsUniversity of CaliforniaSan FranciscoCAUSA
| |
Collapse
|
35
|
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
|
36
|
Schober ME, Block B, Beachy JC, Statler KD, Giza CC, Lane RH. Early and Sustained Increase in the Expression of Hippocampal IGF-1, But Not EPO, in a Developmental Rodent Model of Traumatic Brain Injury. J Neurotrauma 2010. [DOI: 10.1089/neu.2010.1226] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
37
|
Comparison of the effects of erythropoietin and its carbamylated derivative on behaviour and hippocampal neurogenesis in mice. Neuropharmacology 2010; 60:354-64. [PMID: 20932982 DOI: 10.1016/j.neuropharm.2010.09.025] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2010] [Revised: 09/28/2010] [Accepted: 09/29/2010] [Indexed: 11/21/2022]
Abstract
Erythropoietin (EPO), a well known haematopoietic growth factor, possesses neuroprotective and neurotrophic effects which have been recently reported to improve cognition and to modulate emotional processing. We investigated the effects of EPO and of its non-erythropoietic carbamylated derivative (CEPO) on memory- and emotion-related behaviour in the adult mouse. Locomotor activity, memory performances (place and object recognition tasks), anxiety- (light/dark transition test) and despair-like behaviours (tail suspension test) were assessed over 6 weeks of repeated EPO or CEPO administration (40 μg/kg, twice a week). Given the potential involvement of hippocampal neurogenesis in memory, we also assessed the effects of EPO and CEPO on neurogenesis in the dentate gyrus. Both treatments improved spatial and non-spatial recognition memory and increased the number of NeuN/BrdU double-labeled cells in the dentate gyrus. These effects seem to be, at least partly, independent from an haematopoietic action since administration of CEPO leads to the similar results. Moreover, CEPO decreased, albeit modestly, despair-related behaviour and tended to decrease anxiety-like behaviour. These results suggest that CEPO is as an attractive molecule for the treatment of neuropsychiatric diseases associating memory and/or emotional disorders.
Collapse
|
38
|
Maggioni D, Nicolini G, Chiorazzi A, Meregalli C, Cavaletti G, Tredici G. Different effects of erythropoietin in cisplatin- and docetaxel-induced neurotoxicity: An in vitro study. J Neurosci Res 2010; 88:3171-9. [DOI: 10.1002/jnr.22465] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
39
|
Maiese K, Shang YC, Chong ZZ, Hou J. Diabetes mellitus: channeling care through cellular discovery. Curr Neurovasc Res 2010; 7:59-64. [PMID: 20158461 DOI: 10.2174/156720210790820217] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2009] [Accepted: 12/29/2009] [Indexed: 12/13/2022]
Abstract
Diabetes mellitus (DM) impacts a significant portion of the world's population and care for this disorder places an economic burden on the gross domestic product for any particular country. Furthermore, both Type 1 and Type 2 DM are becoming increasingly prevalent and there is increased incidence of impaired glucose tolerance in the young. The complications of DM are protean and can involve multiple systems throughout the body that are susceptible to the detrimental effects of oxidative stress and apoptotic cell injury. For these reasons, innovative strategies are necessary for the implementation of new treatments for DM that are generated through the further understanding of cellular pathways that govern the pathological consequences of DM. In particular, both the precursor for the coenzyme beta-nicotinamide adenine dinucleotide (NAD(+)), nicotinamide, and the growth factor erythropoietin offer novel platforms for drug discovery that involve cellular metabolic homeostasis and inflammatory cell control. Interestingly, these agents and their tightly associated pathways that consist of cell cycle regulation, protein kinase B, forkhead transcription factors, and Wnt signaling also function in a broader sense as biomarkers for disease onset and progression.
Collapse
Affiliation(s)
- Kenneth Maiese
- Division of Cellular and Molecular Cerebral Ischemia, Wayne State University School of Medicine, Detroit, Michigan 48201, USA.
| | | | | | | |
Collapse
|
40
|
Sturm B, Helminger M, Steinkellner H, Heidari MM, Goldenberg H, Scheiber-Mojdehkar B. Carbamylated erythropoietin increases frataxin independent from the erythropoietin receptor. Eur J Clin Invest 2010; 40:561-5. [PMID: 20456483 DOI: 10.1111/j.1365-2362.2010.02292.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND Friedreich's ataxia (FRDA) is a neurodegenerative disorder caused by decreased expression of the mitochondrial protein frataxin. Recently we showed in a clinical pilot study in Friedreich's ataxia patients that recombinant human erythropoietin (rhuEPO) significantly increases frataxin-expression. In this in vitro study, we investigated the role of the erythropoietin receptor (EPO-R) in the frataxin increasing effect of rhuEPO and if nonerythropoietic carbamylated erythropoietin (CEPO), which cannot bind to the classical EPO-R increases frataxin expression. MATERIALS AND METHODS In our experiments human erythroleukaemic K562 cells (+ EPO-R), human monocytic leukemia THP-1 cells (- EPO-R) and isolated primary lymphocytes from healthy control and FRDA patients were incubated with different concentrations of rhuEPO or CEPO. Frataxin-expression was detected by an electrochemical luminescence immunoassay (based on the principle of an ELISA). RESULTS We show that rhuEPO increases frataxin-expression in K562 cells (expressing EPO-R) as well as in THP-1 cells (without EPO-R expression). These results were confirmed by the finding that CEPO, which cannot bind to the classical EPO-R increased frataxin expression in the same concentration range as rhuEPO. In addition, we show that both EPO derivatives significantly increase frataxin-expression in vitro in control and Friedreich's ataxia patients primary lymphocytes. CONCLUSION Our results provide a scientific basis for further studies examining the effectiveness of nonerythropoietic derivatives of erythropoietin for the treatment of Friedreich's ataxia patients.
Collapse
|
41
|
Pankratova S, Kiryushko D, Sonn K, Soroka V, Køhler LB, Rathje M, Gu B, Gotfryd K, Clausen O, Zharkovsky A, Bock E, Berezin V. Neuroprotective properties of a novel, non-haematopoietic agonist of the erythropoietin receptor. Brain 2010; 133:2281-94. [DOI: 10.1093/brain/awq101] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
42
|
Current world literature. Curr Opin Pediatr 2010; 22:246-55. [PMID: 20299870 DOI: 10.1097/mop.0b013e32833846de] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
43
|
Abstract
PURPOSE OF REVIEW Perinatal asphyxia, intraventricular hemorrhage and stroke are common causes of neonatal brain injury, with hypoxia-ischemia as the final common pathway of injury. Erythropoietin (Epo) has potential to lessen neurologic sequelae due to hypoxia-ischemia. The purpose of this review is to highlight new clinical trials and experimental evidence that expand our understanding of Epo as a potential treatment for perinatal brain injury. RECENT FINDINGS Several trials of Epo treatment are reviewed: two phase I/II trials of high-dose Epo given to preterm infants established pharmacokinetic and safety profiles, and a trial of Epo treatment for term infants with moderate hypoxic-ischemic encephalopathy found reduced disability. Potential risks and benefits of high-dose Epo are discussed. New evidence related to Epo receptor expression, signal transduction pathways, and mechanisms of neuroprotection are reviewed. SUMMARY Cautious optimism is warranted regarding the use of high-dose Epo as a treatment option for neonatal brain injury. To date, Epo has been well tolerated to use in neonatal populations and now studies of neuroprotective efficacy are underway.
Collapse
|
44
|
Fu ZQ, Shao QL, Shen JL, Zhang YJ, Zhao XX, Yao L. Effect of carbamylated erythropoietin on major histocompatibility complex expression and neural differentiation of human neural stem cells. J Neuroimmunol 2010; 221:15-24. [PMID: 20163877 DOI: 10.1016/j.jneuroim.2010.01.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2009] [Revised: 01/13/2010] [Accepted: 01/25/2010] [Indexed: 10/19/2022]
Abstract
The expression of major histocompatibility complex (MHC) on human neural stem cells (hNSCs) is tightly related to the fate of these cells in transplantation, therefore strategies to relieve rejection and promote graft survival are necessary to be applied. This study investigated the effect of carbamylated erythropoietin (CEPO) on MHC expression and differentiation of hNSCs with or without IFN-gamma incubation. Results showed that low levels of MHC molecules were expressed on hNSCs and increased by IFN-gamma. CEPO enhanced MHC-I antigens in both proliferative and differentiated hNSCs, but decreased MHC-II antigens in differentiated hNSCs and those cells exposed to IFN-gamma. Furthermore, CEPO promoted neural differentiation of hNSCs and outgrowth of neurites. Western blot analysis revealed activation of Stat3, Stat5 and Akt during these processes. These results suggest that CEPO may have immunoregulatory function in hNSCs besides its neuroprotection.
Collapse
Affiliation(s)
- Zhong-Qiu Fu
- Department of Pediatrics, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | | | | | | | | | | |
Collapse
|
45
|
Shang YC, Chong ZZ, Hou J, Maiese K. FoxO3a governs early microglial proliferation and employs mitochondrial depolarization with caspase 3, 8, and 9 cleavage during oxidant induced apoptosis. Curr Neurovasc Res 2010; 6:223-38. [PMID: 19807657 DOI: 10.2174/156720209789630302] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2009] [Accepted: 08/31/2009] [Indexed: 12/16/2022]
Abstract
Microglia of the central nervous system have a dual role in the ability to influence the survival of neighboring cells. During inflammatory cell activation, microglia can lead to the disposal of toxic cellular products and permit tissue regeneration, but microglia also may lead to cellular destruction with phagocytic removal. For these reasons, it is essential to elucidate not only the underlying pathways that control microglial activation and proliferation, but also the factors that determine microglial survival. In this regard, we investigated in the EOC 2 microglial cell line with an oxygen-glucose deprivation (OGD) injury model of oxidative stress the role of the "O" class forkhead transcription factor FoxO3a that in some scenarios is closely linked to immune system function. We demonstrate that FoxO3a is a necessary element in the control of early and late apoptotic injury programs that involve membrane phosphatidylserine externalization and nuclear DNA degradation, since transient knockdown of FoxO3a in microglia preserves cellular survival 24 hours following OGD exposure. However, prior to the onset of apoptotic injury, FoxO3a facilitates the activation and proliferation of microglia as early as 3 hours following OGD exposure that occurs in conjunction with the trafficking of the unphosphorylated and active post-translational form of FoxO3a from the cytoplasm to the cell nucleus. FoxO3a also can modulate apoptotic mitochondrial signal transduction pathways in microglia, since transient knockdown of FoxO3a prevents mitochondrial membrane depolarization as well as the release of cytochrome c during OGD. Control of this apoptotic cascade also extends to progressive caspase activation as early as 1 hour following OGD exposure. The presence of FoxO3a is necessary for the expression of cleaved (active) caspase 3, 8, and 9, since loss of FoxO3a abrogates the induction of caspase activity. Interestingly, elimination of FoxO3a reduced caspase 9 activity to a lesser extent than that noted with caspase 3 and 8 activities, suggesting that FoxO3a in relation to caspase 9 may be more reliant upon other signal transduction pathways potentially independent from caspase 3 and 8.
Collapse
Affiliation(s)
- Yan Chen Shang
- Division of Cellular and Molecular Cerebral Ischemia, Wayne State University School of Medicine, Detroit, Michigan 48201, USA
| | | | | | | |
Collapse
|
46
|
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: 91] [Impact Index Per Article: 6.1] [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
|
47
|
Medana IM, Day NPJ, Hien TT, White NJ, Turner GDH. Erythropoietin and its receptors in the brainstem of adults with fatal falciparum malaria. Malar J 2009; 8:261. [PMID: 19930602 PMCID: PMC2785829 DOI: 10.1186/1475-2875-8-261] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2009] [Accepted: 11/22/2009] [Indexed: 12/14/2022] Open
Abstract
Background Facilitation of endogenous neuroprotective pathways, such as the erythropoietin (Epo) pathway, has been proposed as adjuvant treatment strategies in cerebral malaria. Whether different endogenous protein expression levels of Epo or differences in the abundance of its receptor components could account for the extent of structural neuropathological changes or neurological complications in adults with severe malaria was investigated. Methods High sensitivity immunohistochemistry was used to assess the frequency, distribution and concordance of Epo and components of its homodimeric and heteromeric receptors, Epo receptor and CD131, within the brainstem of adults who died of severe malaria. The following relationships with Epo and its receptor components were also defined: (i) sequestration and indicators of hypoxia; (ii) vascular damage in the form of plasma protein leakage and haemorrhage; (iii) clinical complications and neuropathological features of severe malaria disease. Brainstems of patients dying in the UK from unrelated non-infectious causes were examined for comparison. Results The incidence of endogenous Epo in parenchymal brain cells did not greatly differ between severe malaria and non-neurological UK controls at the time of death. However, EpoR and CD131 labelling of neurons was greater in severe malaria compared with non-neurological controls (P = .009). EpoR labelling of vessels was positively correlated with admission peripheral parasite count (P = .01) and cerebral sequestration (P < .0001). There was a strong negative correlation between arterial oxygen saturation and EpoR labelling of glia (P = .001). There were no significant correlations with indicators of vascular damage, neuronal chromatolysis, axonal swelling or vital organ failure. Conclusion Cells within the brainstem of severe malaria patients showed protein expression of Epo and its receptor components. However, the incidence of endogeneous expression did not reflect protection from vascular or neuronal injury, and/or clinical manifestations, such as coma. These findings do not provide support for Epo as an adjuvant neuroprotective agent in adults with severe malaria.
Collapse
Affiliation(s)
- Isabelle M Medana
- Nuffield Department of Clinical Laboratory Sciences, The John Radcliffe Hospital, University of Oxford, Oxford, UK.
| | | | | | | | | |
Collapse
|
48
|
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: 36] [Impact Index Per Article: 2.3] [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
|
49
|
Optimal neuroprotection by erythropoietin requires elevated expression of its receptor in neurons. Proc Natl Acad Sci U S A 2009; 106:9848-53. [PMID: 19497871 DOI: 10.1073/pnas.0901840106] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Erythropoietin receptor (EpoR) binding mediates neuroprotection by endogenous Epo or by exogenous recombinant human (rh)Epo. The level of EpoR gene expression may determine tissue responsiveness to Epo. Thus, harnessing the neuroprotective power of Epo requires an understanding of the Epo-EpoR system and its regulation. We tested the hypothesis that neuronal expression of EpoR is required to achieve optimal neuroprotection by Epo. The ventral limbic region (VLR) in the rat brain was used because we determined that its neurons express minimal EpoR under basal conditions, and they are highly sensitive to excitotoxic damage, such as occurs with pilocarpine-induced status epilepticus (Pilo-SE). We report that (i) EpoR expression is significantly elevated in nearly all VLR neurons when rats are subjected to 3 moderate hypoxic exposures, with each separated by a 4-day interval; (ii) synergistic induction of EpoR expression is achieved in the dorsal hippocampus and neocortex by the combination of hypoxia and exposure to an enriched environment, with minimal increased expression by either treatment alone; and (iii) rhEpo administered after Pilo-SE cannot rescue neurons in the VLR, unless neuronal induction of EpoR is elicited by hypoxia before Pilo-SE. This study thus demonstrates using environmental manipulations in normal rodents, the strict requirement for induction of EpoR expression in brain neurons to achieve optimal neuroprotection. Our results indicate that regulation of EpoR gene expression may facilitate the neuroprotective potential of rhEpo.
Collapse
|