1
|
Wang F, Zhang X, Zhang J, Xu Q, Yu X, Xu A, Yi C, Bian X, Shao S. Recent advances in the adjunctive management of diabetic foot ulcer: Focus on noninvasive technologies. Med Res Rev 2024; 44:1501-1544. [PMID: 38279968 DOI: 10.1002/med.22020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 12/15/2023] [Accepted: 01/10/2024] [Indexed: 01/29/2024]
Abstract
Diabetic foot ulcer (DFU) is one of the most costly and serious complications of diabetes. Treatment of DFU is usually challenging and new approaches are required to improve the therapeutic efficiencies. This review aims to update new and upcoming adjunctive therapies with noninvasive characterization for DFU, focusing on bioactive dressings, bioengineered tissues, mesenchymal stem cell (MSC) based therapy, platelet and cytokine-based therapy, topical oxygen therapy, and some repurposed drugs such as hypoglycemic agents, blood pressure medications, phenytoin, vitamins, and magnesium. Although the mentioned therapies may contribute to the improvement of DFU to a certain extent, most of the evidence come from clinical trials with small sample size and inconsistent selections of DFU patients. Further studies with high design quality and adequate sample sizes are necessitated. In addition, no single approach would completely correct the complex pathogenesis of DFU. Reasonable selection and combination of these techniques should be considered.
Collapse
Affiliation(s)
- Fen Wang
- Division of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
| | - Xiaoling Zhang
- Division of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
| | - Jing Zhang
- Division of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
| | - Qinqin Xu
- Division of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
| | - Xuefeng Yu
- Division of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
| | - Anhui Xu
- Division of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chengla Yi
- Division of Trauma Surgery, Tongji Hospital, Tongji Medical College, Wuhan, China
| | - Xuna Bian
- Division of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
| | - Shiying Shao
- Division of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
| |
Collapse
|
2
|
Burciaga-Hernandez LA, Cueto-Villalobos CF, Ortega-Piñon N, Gonzalez-Curiel IE, Godina-Gonzalez S, Mendez-Frausto G, Aguilar-Esquivel AP, Maldonado-Lagunas V, Guerrero-de la Torre LE, Melendez-Zajgla J, Sanchez-Garcia EK, Mitre-Aguilar IB, Mendoza-Almanza G. Gene Expression Behavior of a Set of Genes in Platelet and Tissue Samples from Patients with Breast Cancer. Int J Mol Sci 2023; 24:ijms24098348. [PMID: 37176055 PMCID: PMC10179257 DOI: 10.3390/ijms24098348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/25/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
The tumor microenvironment (TME) is constituted by a great diversity of highly dynamic cell populations, each of which contributes ligands, receptors, soluble proteins, mRNAs, and miRNAs, in order to regulate cellular activities within the TME and even promote processes such as angiogenesis or metastasis. Intravasated platelets (PLT) undergo changes in the TME that convert them into tumor-educated platelets (TEP), which supports the development of cancer, angiogenesis, and metastasis through the degranulation and release of biomolecules. Several authors have reported that the deregulation of PF4, VEGF, PDGF, ANG-1, WASF3, LAPTM4B, TPM3, and TAC1 genes participates in breast cancer progression, angiogenesis, and metastasis. The present work aimed to analyze the expression levels of this set of genes in tumor tissues and platelets derived from breast cancer patients by reverse transcription-quantitative polymerase chain reaction (RTqPCR) assays, in order to determine if there was an expression correlation between these sources and to take advantage of the new information to be used in possible diagnosis by liquid biopsy. Data from these assays showed that platelets and breast cancer tumors present similar expression levels of a subset of these genes' mRNAs, depending on the molecular subtype, comorbidities, and metastasis presence.
Collapse
Affiliation(s)
- Luis A Burciaga-Hernandez
- Maestría en Ciencias Biomédicas, Universidad Autónoma de Zacatecas, Zacatecas 98160, Mexico
- Unidad Académica de Ciencias Biológicas, Universidad Autónoma de Zacatecas, Zacatecas 98068, Mexico
| | | | - Nancy Ortega-Piñon
- Unidad Académica de Ciencias Biológicas, Universidad Autónoma de Zacatecas, Zacatecas 98068, Mexico
| | - Irma E Gonzalez-Curiel
- Laboratorio de InmunotoxicologÍa y Terapéutica Experimental, Unidad Académica de Ciencias QuÍmicas, Universidad Autónoma de Zacatecas, Zacatecas 98160, Mexico
| | - Susana Godina-Gonzalez
- Laboratorio de Biomarcadores, Unidad Académica de Ciencias QuÍmicas, Universidad Autónoma de Zacatecas, Zacatecas 98160, Mexico
| | - Gwendolyne Mendez-Frausto
- Laboratorio de InmunotoxicologÍa y Terapéutica Experimental, Unidad Académica de Ciencias QuÍmicas, Universidad Autónoma de Zacatecas, Zacatecas 98160, Mexico
| | | | - Vilma Maldonado-Lagunas
- Laboratorio de Epigenetica, Instituto Nacional de Medicina Genomica (INMEGEN), Ciudad de México 14610, Mexico
| | - Luis E Guerrero-de la Torre
- Maestría en Ciencias Biomédicas, Universidad Autónoma de Zacatecas, Zacatecas 98160, Mexico
- Hospital General Zacatecas "Luz González Cosío", Zacatecas 98160, Mexico
| | - Jorge Melendez-Zajgla
- Laboratorio de Genomica Funcional del Cancer, Instituto Nacional de Medicina Genomica (INMEGEN), Ciudad de México 14610, Mexico
| | - Erika K Sanchez-Garcia
- Laboratorio de Epigenetica, Instituto Nacional de Medicina Genomica (INMEGEN), Ciudad de México 14610, Mexico
| | - Irma B Mitre-Aguilar
- Unidad de Bioquímica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubiran (INCMNSZ), Ciudad de México 14080, Mexico
| | - Gretel Mendoza-Almanza
- Maestría en Ciencias Biomédicas, Universidad Autónoma de Zacatecas, Zacatecas 98160, Mexico
- Laboratorio de Epigenetica, Instituto Nacional de Medicina Genomica (INMEGEN), Ciudad de México 14610, Mexico
- Consejo Nacional de Ciencia y Tecnología, Ciudad de México 03940, Mexico
| |
Collapse
|
3
|
Iso Y, Usui S, Suzuki H. Mesenchymal Stem/Stromal Cells in Skeletal Muscle Are Pro-Angiogenic, and the Effect Is Potentiated by Erythropoietin. Pharmaceutics 2023; 15:pharmaceutics15041049. [PMID: 37111534 PMCID: PMC10142054 DOI: 10.3390/pharmaceutics15041049] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/11/2023] [Accepted: 03/23/2023] [Indexed: 04/29/2023] Open
Abstract
The aim of this study was to investigate the angiogenic potential of skeletal muscle mesenchymal stem/stromal cells (mMSCs). Platelet-derived growth factor receptor (PDGFR)-α positive mMSCs secreted vascular endothelial growth factor (VEGF) and hepatocyte growth factor when cultured in an ELISA assay. The mMSC-medium significantly induced endothelial tube formation in an in vitro angiogenesis assay. The mMSC implantation promoted capillary growth in rat limb ischemia models. Upon identifying the erythropoietin receptor (Epo-R) in the mMSCs, we examined how Epo affected the cells. Epo stimulation enhanced the phosphorylation of Akt and STAT3 in the mMSCs and significantly promoted cellular proliferation. Next, Epo was directly administered into the rats' ischemic hindlimb muscles. PDGFR-α positive mMSCs in the interstitial area of muscles expressed VEGF and proliferating cell markers. The proliferating cell index was significantly higher in the ischemic limbs of Epo-treated rats than in untreated controls. Investigations by laser Doppler perfusion imaging and immunohistochemistry demonstrated significantly improved perfusion recovery and capillary growth in the Epo-treated groups versus the control groups. Taken together, the results of this study demonstrated that mMSCs possessed a pro-angiogenic property, were activated by Epo, and potentially contributed to capillary growth in skeletal muscle after ischemic injury.
Collapse
Affiliation(s)
- Yoshitaka Iso
- Division of Cardiology, Showa University Fujigaoka Hospital, 1-30 Fujigaoka, Yokohama City 227-8501, Kanagawa, Japan
| | - Sayaka Usui
- Division of Cardiology, Showa University Fujigaoka Hospital, 1-30 Fujigaoka, Yokohama City 227-8501, Kanagawa, Japan
| | - Hiroshi Suzuki
- Division of Cardiology, Showa University Fujigaoka Hospital, 1-30 Fujigaoka, Yokohama City 227-8501, Kanagawa, Japan
| |
Collapse
|
4
|
Menger MM, Nalbach L, Roma LP, Laschke MW, Menger MD, Ampofo E. Erythropoietin exposure of isolated pancreatic islets accelerates their revascularization after transplantation. Acta Diabetol 2021; 58:1637-1647. [PMID: 34254190 PMCID: PMC8542558 DOI: 10.1007/s00592-021-01760-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 06/06/2021] [Indexed: 12/15/2022]
Abstract
AIMS The exposure of isolated pancreatic islets to pro-angiogenic factors prior to their transplantation represents a promising strategy to accelerate the revascularization of the grafts. It has been shown that erythropoietin (EPO), a glycoprotein regulating erythropoiesis, also induces angiogenesis. Therefore, we hypothesized that EPO exposure of isolated islets improves their posttransplant revascularization. METHODS Flow cytometric, immunohistochemical and quantitative real-time (qRT)-PCR analyses were performed to study the effect of EPO on the viability, cellular composition and gene expression of isolated islets. Moreover, islets expressing a mitochondrial or cytosolic H2O2 sensor were used to determine reactive oxygen species (ROS) levels. The dorsal skinfold chamber model in combination with intravital fluorescence microscopy was used to analyze the revascularization of transplanted islets. RESULTS We found that the exposure of isolated islets to EPO (3 units/mL) for 24 h does not affect the viability and the production of ROS when compared to vehicle-treated and freshly isolated islets. However, the exposure of islets to EPO increased the number of CD31-positive cells and enhanced the gene expression of insulin and vascular endothelial growth factor (VEGF)-A. The revascularization of the EPO-cultivated islets was accelerated within the initial phase after transplantation when compared to both controls. CONCLUSION These findings indicate that the exposure of isolated islets to EPO may be a promising approach to improve clinical islet transplantation.
Collapse
Affiliation(s)
- Maximilian M Menger
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg, Saar, Germany
- Department of Trauma and Reconstructive Surgery, Faculty of Medicine, BG Hospital Tuebingen, Eberhard-Karls-University Tuebingen, Tuebingen, Germany
| | - Lisa Nalbach
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg, Saar, Germany
| | - Leticia P Roma
- Biophysics Department, Center for Human and Molecular Biology, Saarland University, 66421, Homburg, Saar, Germany
| | - Matthias W Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg, Saar, Germany
| | - Michael D Menger
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg, Saar, Germany
| | - Emmanuel Ampofo
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg, Saar, Germany.
| |
Collapse
|
5
|
Menger MM, Nalbach L, Roma LP, Körbel C, Wrublewsky S, Glanemann M, Laschke MW, Menger MD, Ampofo E. Erythropoietin accelerates the revascularization of transplanted pancreatic islets. Br J Pharmacol 2020; 177:1651-1665. [PMID: 31721150 DOI: 10.1111/bph.14925] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 10/30/2019] [Accepted: 11/03/2019] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND AND PURPOSE Pancreatic islet transplantation is a promising therapeutic approach for Type 1 diabetes. A major prerequisite for the survival of grafted islets is a rapid revascularization after transplantation. Erythropoietin (EPO), the primary regulator of erythropoiesis, has been shown to promote angiogenesis. Therefore, we investigated in this study whether EPO improves the revascularization of transplanted islets. EXPERIMENTAL APPROACH Islets from FVB/N mice were transplanted into dorsal skinfold chambers of recipient animals, which were daily treated with an intraperitoneal injection of EPO (500 IU·kg-1 ) or vehicle (control) throughout an observation period of 14 days. In a second set of experiments, animals were only pretreated with EPO over a 6-day period prior to islet transplantation. The revascularization of the grafts was assessed by repetitive intravital fluorescence microscopy and immunohistochemistry. In addition, a streptozotocin-induced diabetic mouse model was used to study the effect of EPO-pretreatment on the endocrine function of the grafts. KEY RESULTS EPO treatment slightly accelerated the revascularization of the islet grafts. This effect was markedly more pronounced in EPO-pretreated animals, resulting in significantly higher numbers of engrafted islets and an improved perfusion of endocrine tissue without affecting systemic haematocrit levels when compared with controls. Moreover, EPO-pretreatment significantly accelerated the recovery of normoglycaemia in diabetic mice after islet transplantation. CONCLUSION AND IMPLICATIONS These findings demonstrate that, particularly, short-term EPO-pretreatment represents a promising therapeutic approach to improve the outcome of islet transplantation, without an increased risk of thromboembolic events.
Collapse
Affiliation(s)
- Maximilian M Menger
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Lisa Nalbach
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Leticia P Roma
- Biophysics Department, Center for Human and Molecular Biology, Saarland University, Homburg/Saar, Germany
| | - Christina Körbel
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Selina Wrublewsky
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Matthias Glanemann
- Department for General, Visceral, Vascular and Pediatric Surgery, Saarland University, Homburg/Saar, Germany
| | - Matthias W Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Michael D Menger
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Emmanuel Ampofo
- Institute for Clinical and Experimental Surgery, Saarland University, Homburg/Saar, Germany
| |
Collapse
|
6
|
Cohrs G, Goerden S, Lucius R, Synowitz M, Mehdorn HM, Held-Feindt J, Knerlich-Lukoschus F. Spatial and Cellular Expression Patterns of Erythropoietin-Receptor and Erythropoietin during a 42-Day Post-Lesional Time Course after Graded Thoracic Spinal Cord Impact Lesions in the Rat. J Neurotrauma 2018; 35:593-607. [PMID: 28895456 DOI: 10.1089/neu.2017.4981] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Erythropoietin (Epo) exhibits promising neuroregenerative potential for spinal cord injury (SCI), and might be involved in other long-term sequelae, such as neuropathic pain development. The current studies investigated the time courses and spatial and cellular patterns of Epo and erythropoietin receptor (EpoR) expression along the spinal axis after graded SCI. Male Long Evans rats received 100 kdyn, 150 kdyn, and 200 kdyn thoracic (T9) contusions from an Infinite Horizon impactor. Sham controls received laminectomies. Anatomical and quantitative immunohistochemical analyses of the EpoR/Epo expression along the whole spinal axis were performed 7, 15, and 42 postoperative days (DPO) after the lesioning. Cellular expression was investigated by double- and triple-labeling for EpoR/Epo with cellular markers and proliferating cells in subgroups of 5-bromo-2-deoxyuridine pre-treated animals. Prolonged EpoR/Epo-expression was confirmed by real-time reverse transcriptase polymerase chain reaction (RT-PCR). Quantified EpoR/Epo immunoreactivities in pain-related spinal cord regions and ventrolateral white matter (VLWM) were correlated with the mechanical sensitivity thresholds and locomotor function of the respective animals. EpoR and Epo were constitutively expressed in the ventral horn neurons and vascular and glial cells in the dorsal columns (DC) and the VLWM. After SCI, in addition to expression in the lesion core, EpoR/Epo immunoreactivities exhibited significant time- and lesion grade-dependent induction in the DC and VLWM along the spinal axis. EpoR and Epo immunoreactive cells were co-stained with markers for astroglial, neural precursor cell and vascular markers. In the VLWM, EpoR- and Epo-positive proliferating cells were co-stained with glial fibrillary acidic protein (GFAP) and nestin. The DC EpoR/Epo immunoreactivities exhibited linear relationships with the behavioral correlates of post-lesional chronic pain development at DPO 42. SCI leads to long-lasting multicellular EpoR/Epo induction beyond the lesion core in the spinal cord regions that are involved in central pain development and regenerative processes. Our studies provide a time frame to investigate the effects of Epo application on motor function or pain development, especially in the later time course after lesioning.
Collapse
Affiliation(s)
- Gesa Cohrs
- 1 Department of Neurosurgery, University Hospital of Schleswig-Holstein Campus Kiel , Kiel, Germany
| | - Stephan Goerden
- 1 Department of Neurosurgery, University Hospital of Schleswig-Holstein Campus Kiel , Kiel, Germany
| | - Ralph Lucius
- 2 Anatomical Institute, Christian-Albrechts University Kiel , Kiel, Germany
| | - Michael Synowitz
- 1 Department of Neurosurgery, University Hospital of Schleswig-Holstein Campus Kiel , Kiel, Germany
| | | | - Janka Held-Feindt
- 1 Department of Neurosurgery, University Hospital of Schleswig-Holstein Campus Kiel , Kiel, Germany
| | | |
Collapse
|
7
|
Erythropoietin and Its Angiogenic Activity. Int J Mol Sci 2017; 18:ijms18071519. [PMID: 28703764 PMCID: PMC5536009 DOI: 10.3390/ijms18071519] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 07/10/2017] [Accepted: 07/11/2017] [Indexed: 01/09/2023] Open
Abstract
Erythropoietin (EPO) is the main hematopoietic hormone acting on progenitor red blood cells via stimulation of cell growth, differentiation, and anti-apoptosis. However, its receptor (EPOR) is also expressed in various non-hematopoietic tissues, including endothelium. EPO is a pleiotropic growth factor that exhibits growth stimulation and cell/tissue protection on numerous cells and tissues. In this article we review the angiogenesis potential of EPO on endothelial cells in heart, brain, and leg ischemia, as well as its role in retinopathy protection and tumor promotion. Furthermore, the effect of EPO on bone marrow and adipose tissue is also discussed.
Collapse
|
8
|
McKeever KH, McNally BA, Hinchcliff KW, Lehnhard RA, Poole DC. Effects of erythropoietin on systemic hematocrit and oxygen transport in the splenectomized horse. Respir Physiol Neurobiol 2016; 225:38-47. [PMID: 26853328 DOI: 10.1016/j.resp.2016.02.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 01/30/2016] [Accepted: 02/02/2016] [Indexed: 11/16/2022]
Abstract
To test the hypotheses that erythropoietin (rhuEPO) treatment increases systemic hematocrit, maximal O2 uptake (VO2max, by elevated perfusive and diffusive O2 conductances) and performance five female horses (4-13 years) received 15 IU/kg rhuEPO (erythropoietin) three times per week for three weeks. These horses had been splenectomized over 1 year previously to avoid confounding effects from the mobilization of splenic red blood cell reserves. Each horse performed three maximal exercise tests (one per month) on an inclined (4°) treadmill to the limit of tolerance; two control trials and one following EPO treatment. Measurements of hemoglobin concentration ([Hb] and hematocrit), plasma and blood volume, VO2, cardiac output as well as arterial and mixed venous blood gases were made at rest and during maximal exercise. EPO increased resting [Hb] by 18% from 13.3 ± 0.6 to 15.7 ± 0.8 g/dL (mean ± SD) corresponding to an increased hematocrit from 36 ± 2 to 46 ± 2% concurrent with 23 and 10% reductions in plasma and blood volume, respectively (all P<0.05). EPO elevated VO2max by 20% from 25.7 ± 1.7 to 30.9 ± 3.4 L/min (P<0.05) via a 17% increase in arterial O2 content and 18% greater arteriovenous O2 difference in the face of an unchanged cardiac output. To achieve the greater VO2max after EPO, diffusive O2 conductance increased ∼ 30% (from 580 ± 76 to 752 ± 166 mL O2/mmHg/min, P<0.05) which was substantially greater than the elevation of perfusive O2 conductance. These effects of EPO were associated with an increased exercise performance (total running time: control, 216 ± 72; EPO, 264 ± 48 s, P<0.05). We conclude that EPO substantially increases VO2max and performance in the splenectomized horse via improved perfusive and diffusive O2 transport.
Collapse
Affiliation(s)
- Kenneth H McKeever
- Department of Animal Science, Rutgers the State University of New Jersey, New Brunswick, NJ 08903, United States.
| | - Beth A McNally
- School of Health, Physical Education and Recreation, The Ohio State University, Columbus, OH 43210, United States
| | - Kenneth W Hinchcliff
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Melbourne, Australia
| | - Robert A Lehnhard
- Department of Kinesiology, University of Maine, Orono, ME, United States
| | - David C Poole
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, United States
| |
Collapse
|
9
|
Epoetin Alpha and Epoetin Zeta: A Comparative Study on Stimulation of Angiogenesis and Wound Repair in an Experimental Model of Burn Injury. BIOMED RESEARCH INTERNATIONAL 2015; 2015:968927. [PMID: 26146639 PMCID: PMC4471383 DOI: 10.1155/2015/968927] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 04/02/2015] [Accepted: 04/15/2015] [Indexed: 11/17/2022]
Abstract
Deep second-degree burns are characterized by delayed formation of granulation tissue and impaired angiogenesis. Erythropoietin (EPO) is able to stimulate angiogenesis and mitosis, activating vascularization and cell cycle. The aim of our study was to investigate whether two biosimilar recombinant human erythropoietins, EPO-α and EPO-Z, may promote these processes in an experimental model of burn injury. A total of 84 mice were used and a scald burn was produced on the back after shaving, in 80°C water for 10 seconds. Mice were then randomized to receive EPO-α (400 units/kg/day/sc) or EPO-Z (400 units/kg/day/sc) or their vehicle (100 μL/day/sc 0.9% NaCl solution). After 12 days, both EPO-α and EPO-Z increased VEGF protein expression. EPO-α caused an increased cyclin D1/CDK6 and cyclin E/CDK2 expression compared with vehicle and EPO-Z (p<0.001). Our study showed that EPO-α and EPO-Z accelerated wound closure and angiogenesis; however EPO-α resulted more effectively in achieving complete skin regeneration. Our data suggest that EPO-α and EPO-Z are not biosimilars for the wound healing effects. The higher efficacy of EPO-α might be likely due to its different conformational structure leading to a more efficient cell proliferation and skin remodelling.
Collapse
|
10
|
Feder D, Rugollini M, Santomauro A, Oliveira LP, Lioi VP, Santos RD, Ferreira LG, Nunes MT, Carvalho MH, Delgado PO, Carvalho AAS, Fonseca FLA. Erythropoietin reduces the expression of myostatin in mdx dystrophic mice. ACTA ACUST UNITED AC 2014. [PMID: 25296358 PMCID: PMC4230286 DOI: 10.1590/1414-431x20143858] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Erythropoietin (EPO) has been well characterized as a renal glycoprotein hormone
regulating red blood cell production by inhibiting apoptosis of erythrocyte
progenitors in hematopoietic tissues. EPO exerts regulatory effects in cardiac and
skeletal muscles. Duchenne muscular dystrophy is a lethal degenerative disorder of
skeletal and cardiac muscle. In this study, we tested the possible therapeutic
beneficial effect of recombinant EPO (rhEPO) in dystrophic muscles in mdx mice. Total
strength was measured using a force transducer coupled to a computer. Gene expression
for myostatin, transforming growth factor-β1 (TGF-β1), and tumor necrosis factor-α
(TNF-α) was determined by quantitative real time polymerase chain reaction. Myostatin
expression was significantly decreased in quadriceps from mdx mice treated with rhEPO
(rhEPO=0.60±0.11, control=1.07±0.11). On the other hand, rhEPO had no significant
effect on the expression of TGF-β1 (rhEPO=0.95±0.14, control=1.05±0.16) and TNF-α
(rhEPO=0.73±0.20, control=1.01±0.09). These results may help to clarify some of the
direct actions of EPO on skeletal muscle.
Collapse
Affiliation(s)
- D Feder
- Faculdade de Medicina do ABC, Santo André, SP, Brasil
| | - M Rugollini
- Faculdade de Medicina do ABC, Santo André, SP, Brasil
| | - A Santomauro
- Faculdade de Medicina do ABC, Santo André, SP, Brasil
| | - L P Oliveira
- Faculdade de Medicina do ABC, Santo André, SP, Brasil
| | - V P Lioi
- Faculdade de Medicina do ABC, Santo André, SP, Brasil
| | - R dos Santos
- Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brasil
| | - L G Ferreira
- Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brasil
| | - M T Nunes
- Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brasil
| | - M H Carvalho
- Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brasil
| | - P O Delgado
- Faculdade de Medicina do ABC, Santo André, SP, Brasil
| | | | - F L A Fonseca
- Faculdade de Medicina do ABC, Santo André, SP, Brasil
| |
Collapse
|
11
|
Guadalupe-Grau A, Plenge U, Helbo S, Kristensen M, Andersen PR, Fago A, Belhage B, Dela F, Helge JW. Effects of an 8-weeks erythropoietin treatment on mitochondrial and whole body fat oxidation capacity during exercise in healthy males. J Sports Sci 2014; 33:570-8. [DOI: 10.1080/02640414.2014.951872] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
12
|
Fatemi MJ, Emami AH, Ghiasi S, Seyed Jafari SM, Mohammadi AA. Effects of recombinant human erythropoietin on revascularization of full thickness skin grafts in rat. IRANIAN RED CRESCENT MEDICAL JOURNAL 2014; 16:e8867. [PMID: 25031867 PMCID: PMC4082527 DOI: 10.5812/ircmj.8867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2012] [Accepted: 07/24/2013] [Indexed: 11/16/2022]
Abstract
Background: Autologous skin graft is frequently used in the field of plastic, and reconstructive surgery. The engraftment is dependent upon revascularization and angiogenesis, which can be regulated by different factors. In addition to its hematopoietic effects, erythropoietin is shown to positively affect the wound healing process. Objectives: We studied effects of human erythropoietin on revascularization of full thickness skin grafts in rat. Materials and Methods: Forty adult Albino male rats were selected for this study. Full thickness skin graft was performed for them, and the effects of systemic, and localized administration of erythropoietin on vascularization of the graft area were evaluated in four groups as following: inverse group underwent full thickness skin graft; in normal saline group normal saline was injected under the fascia of grafted area for seven days; systemic EPO group received systemic erythropoietin for seven days after the surgery; and in graft EPO group, erythropoietin was injected under the fascia of grafted area after full thickness skin grafting for seven days. Results: Forty adult Albino male rats (n = 40), with weights ranging from 356 to 469 g (mean 391.5 ± 29.6 g) were included. The vascular densities of central margins were significantly different between inverse group and graft EPO groups (P value = 0.01), and vascular density of central margins of normal saline group and graft EPO groups were significantly different too (P value = 0.04). Conclusions: EPO can stimulate angiogenesis which has an important role in wound healing. So, local administration of EPO seems to be beneficial in engraftment.
Collapse
Affiliation(s)
- Mohammad Javad Fatemi
- Department of Plastic and Reconstructive Surgery, Fatemeh Zahra Hospital, Tehran University of Medical Sciences, Tehran, IR Iran
| | - Abol Hasan Emami
- Department of Plastic and Reconstructive Surgery, Fatemeh Zahra Hospital, Tehran University of Medical Sciences, Tehran, IR Iran
| | - Sina Ghiasi
- Shiraz Burn Research Centre, Division of Plastic Surgery, Department of General Surgery, Shiraz University of Medical Sciences, Shiraz, IR Iran
- Corresponding Author: Sina Ghiasi, Shiraz Burn Research Centre, Division of Plastic Surgery, Department of General Surgery, Shiraz University of Medical Sciences, Shiraz, IR Iran. Tel: +98-7118219640; Ext: 2, Fax: +98-7118217090, E-mail:
| | - Seyed Morteza Seyed Jafari
- Shiraz Burn Research Centre, Division of Plastic Surgery, Department of General Surgery, Shiraz University of Medical Sciences, Shiraz, IR Iran
| | - Ali Akbar Mohammadi
- Shiraz Burn Research Centre, Division of Plastic Surgery, Department of General Surgery, Shiraz University of Medical Sciences, Shiraz, IR Iran
| |
Collapse
|
13
|
Al-Shabrawey M, Elsherbiny M, Nussbaum J, Othman A, Megyerdi S, Tawfik A. Targeting Neovascularization in Ischemic Retinopathy: Recent Advances. EXPERT REVIEW OF OPHTHALMOLOGY 2014; 8:267-286. [PMID: 25598837 DOI: 10.1586/eop.13.17] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pathological retinal neovascularization (RNV) is a common micro-vascular complication in several retinal diseases including retinopathy of prematurity, diabetic retinopathy, age-related macular degeneration and central vein occlusion. The current therapeutic modalities of RNV are invasive and although they may slow or halt the progression of the disease they are unlikely to restore normal acuity. Therefore, there is an urgent need to develop treatment modalities, which are less invasive and therefore associated with fewer procedural complications and systemic side effects. This review article summarizes our understanding of the pathophysiology and current treatment of RNV in ischemic retinopathies; lists potential therapeutic targets; and provides a framework for the development of future treatment modalities.
Collapse
Affiliation(s)
- Mohamed Al-Shabrawey
- Oral Biology/Anatomy, College of Dental Medicine, GeorgiaRegentsUniversity (GRU), Augusta GA, USA ; Ophthalmology and Vision Discovery Institute, Medical College of Georgia, GRU ; Anatomy, Mansoura Faculty of Medicine, Mansoura University-Egypt ; Vascular Biology Center, Medical College of Georgia, GRU
| | - Mohamed Elsherbiny
- Oral Biology/Anatomy, College of Dental Medicine, GeorgiaRegentsUniversity (GRU), Augusta GA, USA ; Ophthalmology and Vision Discovery Institute, Medical College of Georgia, GRU ; Anatomy, Mansoura Faculty of Medicine, Mansoura University-Egypt
| | - Julian Nussbaum
- Ophthalmology and Vision Discovery Institute, Medical College of Georgia, GRU
| | - Amira Othman
- Anatomy, Mansoura Faculty of Medicine, Mansoura University-Egypt
| | - Sylvia Megyerdi
- Oral Biology/Anatomy, College of Dental Medicine, GeorgiaRegentsUniversity (GRU), Augusta GA, USA
| | - Amany Tawfik
- Ophthalmology and Vision Discovery Institute, Medical College of Georgia, GRU ; Cellular Biology and Anatomy, Medical College of Georgia, GRU
| |
Collapse
|
14
|
Hamed S, Bennett CL, Demiot C, Ullmann Y, Teot L, Desmoulière A. Erythropoietin, a novel repurposed drug: an innovative treatment for wound healing in patients with diabetes mellitus. Wound Repair Regen 2013; 22:23-33. [PMID: 24471742 DOI: 10.1111/wrr.12135] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 10/24/2013] [Indexed: 12/22/2022]
Abstract
Developing a new drug is expensive: the cost of going from bench to bedside is about $US1 billion. Therefore, the repurposing of an approved drug is potentially rewarding because it expands the drug's existing therapeutic profile and preempts additional development costs. As the safety profile of a repurposed drug is already well known, any new investigations could then focus on its efficacy and other therapeutic benefits. Recombinant erythropoietin (EPO) is a potential candidate for repurposing because the results of numerous studies have shown that systemic and topical EPO is therapeutically beneficial when it is administered to healthy and diabetic animals with acute and chronic skin wounds and burns. Moreover, the molecular mechanisms of EPO's actions have been elucidated: EPO acts on those nonhematopoietic cells which are involved in the innate immune response where it promotes cellular proliferation and differentiation, exerts its cytoprotective actions, and inhibits apoptosis. In this review, the mechanism of EPO's action in skin wound healing is reviewed, and its potential for treating acute and chronic skin wounds and stimulating tissue regeneration in diabetic patients is discussed.
Collapse
|
15
|
Hong JP, Park SW. The combined effect of recombinant human epidermal growth factor and erythropoietin on full-thickness wound healing in diabetic rat model. Int Wound J 2012; 11:373-8. [PMID: 23078553 DOI: 10.1111/j.1742-481x.2012.01100.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Diabetic wound is a chronic wound in which normal process of wound healing is interrupted. Lack of blood supply, infection and lack of functional growth factors are assumed as some of the conditions that lead to non-healing environment. Epidermal growth factor (EGF) acts primarily to stimulate epithelial cell growth across wound. Erythropoietin (EPO) is a haematopoietic factor, which stimulates the production, differentiation and maturation of erythroid precursor cells. This study hypothesised combining these two factors, non-healing process of diabetic wound will be compensated and eventually lead to acceleration of wound healing compared with single growth factor treatment. A total of 30 diabetic Sprague-Dawley rats were divided into three treatment groups (single treatment of rh-EPO or rh-EGF or combined treatment on a full-thickness skin wound). To assess the wound healing effects of the components, the wound size and the healing time were measured in each treatment groups. The skin histology was examined by light microscopy and immunohistochemical analysis of proliferating markers was performed. The combined treatment with rh-EPO and rh-EGF improved full-thickness wound significantly (P < 0·05) accelerating 50% healing time with higher expression of Ki-67 compared with single growth factor-treated groups. The combined treatment failed to accelerate the total healing time when compared with single growth factor treatments. However, the significant improvement were found in wound size reduction in the combined treatment group on day 4 against single growth factor-treated groups (P < 0·05). This study demonstrated that the combined treatment of rh-EPO and rh-EGF improved the wound healing possibly through a synergistic action of each growth factor. This application provides further insight into combined growth factor therapy on non-healing diabetic wounds.
Collapse
Affiliation(s)
- Joon Pio Hong
- Department of Plastic Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | | |
Collapse
|
16
|
Sorg H, Harder Y, Krueger C, Reimers K, Vogt PM. The nonhematopoietic effects of erythropoietin in skin regeneration and repair: from basic research to clinical use. Med Res Rev 2012; 33:637-64. [PMID: 22430919 DOI: 10.1002/med.21259] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Erythropoietin (EPO) is the main regulator of red blood cell production but there exists also a variety of nonhematopoietic properties. More recent data show that EPO is also associated with the protection of tissues suffering from ischemia and reperfusion injury as well as with improved regeneration in various organ systems, in particular the skin. This review highlights the mechanisms of EPO in the different stages of wound healing and the reparative processes in the skin emphasizing pathophysiological mechanisms and potential clinical applications. There is clear evidence that EPO effectively influences all wound-healing phases in a dose-dependent manner. This includes inflammation, tissue, and blood vessel formation as well as the remodeling of the wound. The molecular mechanism is predominantly based on an increased expression of the endothelial and inducible nitric oxide (NO) synthase with a consecutive rapid supply of NO as well as an increased content of vascular endothelial growth factor (VEGF) in the wound. The improved understanding of the functions and regulatory mechanisms of EPO in the context of wound-healing problems and ischemia/reperfusion injury, especially during flap surgery, may lead to new considerations of this growth hormone for its regular clinical application in patients.
Collapse
Affiliation(s)
- Heiko Sorg
- Department of Plastic, Hand- and Reconstructive Surgery, Hannover Medical School, 30625, Hannover, Germany.
| | | | | | | | | |
Collapse
|
17
|
Su KH, Shyue SK, Kou YR, Ching LC, Chiang AN, Yu YB, Chen CY, Pan CC, Lee TS. β Common receptor integrates the erythropoietin signaling in activation of endothelial nitric oxide synthase. J Cell Physiol 2011; 226:3330-9. [DOI: 10.1002/jcp.22678] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
18
|
Hamed S, Ullmann Y, Egozi D, Daod E, Hellou E, Ashkar M, Gilhar A, Teot L. Fibronectin potentiates topical erythropoietin-induced wound repair in diabetic mice. J Invest Dermatol 2011; 131:1365-74. [PMID: 21326299 DOI: 10.1038/jid.2011.15] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Diabetes mellitus disrupts all phases of the wound repair cascade and leads to development of chronic wounds. We previously showed that topical erythropoietin (EPO) can promote wound repair in diabetic rats. Fibronectin (FN) has a critical role throughout the process of wound healing, yet it is deficient in wound tissues of diabetic patients. Therefore, we investigated the effect of topical treatment of both EPO and FN (EPO/FN) on wound repair in diabetic mice. Full-thickness excisional skin wounds in diabetic and nondiabetic mice were treated with a cream containing vehicle, EPO, FN, or EPO/FN. We assessed the rate of wound closure, angiogenesis, apoptosis, and expression of inflammatory cytokines, endothelial nitric oxide synthase (eNOS) and β1-integrin, in the wound tissues. We also investigated the effect of EPO, FN, and EPO/FN on human dermal microvascular endothelial cells and fibroblasts cultured on fibrin-coated plates, or in high glucose concentrations. EPO/FN treatment significantly increased the rate of wound closure and this effect was associated with increased angiogenesis, increased eNOS and β1-integrin expression, and reduced expression of inflammatory cytokines and apoptosis. Our findings show that EPO and FN have an additive effect on wound repair in diabetic mice.
Collapse
Affiliation(s)
- Saher Hamed
- The Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Bader A, Lorenz K, Richter A, Scheffler K, Kern L, Ebert S, Giri S, Behrens M, Dornseifer U, Macchiarini P, Machens HG. Interactive Role of Trauma Cytokines and Erythropoietin and Their Therapeutic Potential for Acute and Chronic Wounds. Rejuvenation Res 2011; 14:57-66. [DOI: 10.1089/rej.2010.1050] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Augustinus Bader
- University of Leipzig, Centre for Biotechnology and Biomedicine, Department of Applied Stem Cell Biology and Cell Techniques, Leipzig, Germany
| | - Katrin Lorenz
- University of Leipzig, Centre for Biotechnology and Biomedicine, Department of Applied Stem Cell Biology and Cell Techniques, Leipzig, Germany
| | - Anja Richter
- University of Leipzig, Centre for Biotechnology and Biomedicine, Department of Applied Stem Cell Biology and Cell Techniques, Leipzig, Germany
| | - Katja Scheffler
- University of Leipzig, Centre for Biotechnology and Biomedicine, Department of Applied Stem Cell Biology and Cell Techniques, Leipzig, Germany
| | - Larissa Kern
- University of Leipzig, Centre for Biotechnology and Biomedicine, Department of Applied Stem Cell Biology and Cell Techniques, Leipzig, Germany
| | - Sabine Ebert
- University of Leipzig, Centre for Biotechnology and Biomedicine, Department of Applied Stem Cell Biology and Cell Techniques, Leipzig, Germany
| | - Shibashish Giri
- University of Leipzig, Centre for Biotechnology and Biomedicine, Department of Applied Stem Cell Biology and Cell Techniques, Leipzig, Germany
| | | | - Ulf Dornseifer
- Klinikum Bogenhausen, Zentrum für Schwerbrandverletzte, München, Germany
| | - Paolo Macchiarini
- Hospital Clinico de Barcelona, Department of General Thoracic Surgery, Barcelona, Spain
| | - Hans-Günther Machens
- Klinik für Plastische Chirurgie, Klinikum Rechts der Isar, Technische Universität München, Germany
| |
Collapse
|
20
|
Köllensperger M, Krismer F, Pallua A, Stefanova N, Poewe W, Wenning GK. Erythropoietin is neuroprotective in a transgenic mouse model of multiple system atrophy. Mov Disord 2011; 26:507-515. [PMID: 21462262 DOI: 10.1002/mds.23474] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2009] [Revised: 06/16/2010] [Accepted: 09/13/2010] [Indexed: 11/05/2022] Open
Abstract
Multiple system atrophy is a rapidly progressive neurodegenerative disorder with a markedly reduced life expectancy. Failure of symptomatic treatment raises an urgent need for disease-modifying strategies. We have investigated the neuroprotective potential of erythropoietin in (proteolipid protein)-α-synuclein transgenic mice exposed to 3-nitropropionic acid featuring multiple system atrophy-like pathology including oligodendroglial α-synuclein inclusions and selective neuronal degeneration. Mice were treated with erythropoietin starting before (early erythropoietin) and after (late erythropoietin) intoxication with 3-nitropropionic acid. Nonintoxicated animals receiving erythropoietin and intoxicated animals treated with saline served as control groups. Behavioral tests included pole test, open field activity, and motor behavior scale. Immunohistochemistry for tyrosine hydroxylase and dopamine and cyclic adenosine monophosphate-regulated phosphoprotein (DARPP-32) was analyzed stereologically. Animals receiving erythropoietin before and after 3-nitropropionic acid intoxication scored significantly lower on the motor behavior scale and they performed better in the pole test than controls with no significant difference between early and late erythropoietin administration. Similarly, rearing scores were worse in 3-nitropropionic acid-treated animals with no difference between the erythropoietin subgroups. Immunohistochemistry revealed significant attenuation of 3-nitropropionic acid-induced loss of tyrosine hydroxylase and DARPP-32 positive neurons in substantia nigra pars compacta and striatum, respectively, in both erythropoietin-treated groups without significant group difference in the substantia nigra. However, at striatal level, a significant difference between early and late erythropoietin administration was observed. In the combined (proteolipid protein)-α-synuclein 3-nitropropionic acid multiple system atrophy mouse model, erythropoietin appears to rescue dopaminergic and striatal gabaergic projection neurons. This effect is associated with improved motor function. Further studies are warranted to develop erythropoietin as a potential interventional therapy in multiple system atrophy.
Collapse
Affiliation(s)
- Martin Köllensperger
- Division of Clinical Neurobiology, Department of Neurology, Medical University, Innsbruck, Austria
| | - Florian Krismer
- Division of Clinical Neurobiology, Department of Neurology, Medical University, Innsbruck, Austria
| | - Anton Pallua
- Division of Clinical Neurobiology, Department of Neurology, Medical University, Innsbruck, Austria
| | - Nadia Stefanova
- Division of Clinical Neurobiology, Department of Neurology, Medical University, Innsbruck, Austria
| | - Werner Poewe
- Division of Clinical Neurobiology, Department of Neurology, Medical University, Innsbruck, Austria
| | - Gregor K Wenning
- Division of Clinical Neurobiology, Department of Neurology, Medical University, Innsbruck, Austria
| |
Collapse
|
21
|
Erythropoetin as a novel agent with pleiotropic effects against acute lung injury. Eur J Clin Pharmacol 2010; 67:1-9. [PMID: 21069520 DOI: 10.1007/s00228-010-0938-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2010] [Accepted: 09/28/2010] [Indexed: 12/14/2022]
Abstract
Current pharmacotherapy for acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) is not optimal, and the biological and physiological complexity of these severe lung injury syndromes requires consideration of combined-agent treatments or agents with pleiotropic action. In this regard, exogenous erythropoietin (EPO) represents a possible candidate since a number of preclinical studies have revealed beneficial effects of EPO administration in various experimental models of ALI. Taken together, this treatment strategy is not a single mediator approach, but it rather provides protection by modulating multiple levels of early signaling pathways involved in apoptosis, inflammation, and peroxidation, potentially restoring overall homeostasis. Furthermore, EPO appears to confer vascular protection by promoting angiogenesis. However, only preliminary studies exist and more experimental and clinical studies are necessary to clarify the efficacy and potentially cytoprotective mechanisms of EPO action. In addition to the attempts to optimize the dose and timing of EPO administration, it would be of great value to minimize any potential toxicity, which is essential for EPO to fulfill its role as a potential candidate for the treatment of ALI in routine clinical practice. The present article reviews recent advances that have elucidated biological and biochemical activities of EPO that may be potentially applicable for ALI/ARDS management.
Collapse
|
22
|
Erythropoietin ameliorates the reduced migration of human fibroblasts during in vitro hypoxia. J Physiol Biochem 2010; 67:1-13. [DOI: 10.1007/s13105-010-0043-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Accepted: 08/21/2010] [Indexed: 12/11/2022]
|
23
|
Olgar S, Kara A, Hicyilmaz H, Balta N, Canatan D. Evaluation of angiogenesis with vascular endothelial growth factor in patients with thalassemia major. Pediatr Int 2010; 52:247-51. [PMID: 19744226 DOI: 10.1111/j.1442-200x.2009.02956.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Thalassemia major (TM) is an important cause of severe anemia that necessitates regular blood transfusion to prevent the profound weakness and cardiac decompensation caused by the anemia. However, iron overloading is an inevitable consequence of prolonged transfusion therapy. In addition, extramedullary hematopoiesis and hemosiderosis cause spleen, liver and marrow enlargement. In recent years the role of angiogenesis has been investigated in physiological and pathological conditions. However, it is known that angiogenetic factors, especially the vascular endothelial growth factor (VEGF), cause differentiation of the hemangioblast. METHODS The effect of angiogenesis hasn't been investigated in TM patients yet, and in this study, angiogenesis was researched in 43 thalassemic patients by serum VEGF measurement. RESULTS VEGF levels were not affected by hemoglobin levels, ferritin levels, or chelation type (P > 0.05). However, VEGF was positively affected by chelation starting age and negatively affected by yearly transfusion requirement of TM patients (P < 0.05). In addition, VEGF of patients who underwent splenectomy were higher than those who didn't undergo splenectomy (P < 0.05). CONCLUSION Early chelating age will negatively influence the VEGF level, which increases angiogenesis, however, early starting transfusion age and regular blood transfusion will positively influence the VEGF level, which decreases angiogenesis in thalassemic patients.
Collapse
Affiliation(s)
- Seref Olgar
- Suleyman Demirel University, Faculty of Medicine, Department of Pediatric Hematology, Isparta, Turkey
| | | | | | | | | |
Collapse
|
24
|
Kondyli M, Gatzounis G, Kyritsis A, Varakis J, Assimakopoulou M. Immunohistochemical detection of phosphorylated JAK-2 and STAT-5 proteins and correlation with erythropoietin receptor (EpoR) expression status in human brain tumors. J Neurooncol 2010; 100:157-64. [PMID: 20336349 DOI: 10.1007/s11060-010-0156-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2009] [Accepted: 03/08/2010] [Indexed: 12/18/2022]
Abstract
Phosphorylated (activated) forms of Janus Kinase 2 (pJAK-2) and STAT-5 transcription factor (pSTAT-5), which are preferentially expressed after binding of erythropoietin (Epo) to its receptor EpoR, are known to be implicated in the molecular mechanisms controlling brain development. The purpose of this study was to investigate the expression of these proteins (pJAK-2, pSTAT-5, and EpoR) in human brain tumors compared with normal brain. Using specific antibodies and immunohistochemistry on formalin-fixed, paraffin-embedded semi-serial tissue sections a total of 87 human brain tumors and samples from normal brain tissue were studied. pJAK-2/pSTAT-5 nuclear co-expression was detected in 39% of astrocytomas, 43% of oligodendrogliomas, 50% of ependymomas, and in all (100%) of the medulloblastomas examined. In contrast, most of the meningiomas showed weak or no immunoreactivity for pJAK-2/pSTAT-5 proteins. A significant percentage of tumors exhibited pSTAT-5 immunoreactivity, being pJAK-2 immunonegative. EpoR/pJAK-2/pSTAT-5 co-expression was detected in a small percentage of astrocytomas (18%) and ependymomas (33%). Oligodendrogliomas and medulloblastomas were EpoR immunonegative. Tumor vessels exhibited EpoR, pJAK-2, and pSTAT-5 immunoreactivity. In normal brain tissue, EpoR immunoreactivity was detected in neurons and vessels whereas pSTAT-5 and pJAK-2 immunoreactivity was limited to some neurons and a few glial cells, respectively. These results indicate the existence of ligand (other than Epo)-dependent or independent JAK-2 activation that leads to constitutive activation of STAT-5 in most human brain tumors. Given the oncogenic potential of the JAK/STAT pathway, detection of different pJAK-2 and pSTAT-5 expression profiles between groups of tumors may reflect differences in the biological behavior of the various human brain tumors.
Collapse
Affiliation(s)
- M Kondyli
- Department of Anatomy, School of Medicine, University of Patras, Patras, Greece
| | | | | | | | | |
Collapse
|
25
|
Lundby C, Robach P, Boushel R, Thomsen JJ, Rasmussen P, Koskolou M, Calbet JAL. Does recombinant human Epo increase exercise capacity by means other than augmenting oxygen transport? J Appl Physiol (1985) 2008; 105:581-7. [DOI: 10.1152/japplphysiol.90484.2008] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This study was performed to test the hypothesis that administration of recombinant human erythropoietin (rHuEpo) in humans increases maximal oxygen consumption by augmenting the maximal oxygen carrying capacity of blood. Systemic and leg oxygen delivery and oxygen uptake were studied during exercise in eight subjects before and after 13 wk of rHuEpo treatment and after isovolemic hemodilution to the same hemoglobin concentration observed before the start of rHuEpo administration. At peak exercise, leg oxygen delivery was increased from 1,777.0 ± 102.0 ml/min before rHuEpo treatment to 2,079.8 ± 120.7 ml/min after treatment. After hemodilution, oxygen delivery was decreased to the pretreatment value (1,710.3 ± 138.1 ml/min). Fractional leg arterial oxygen extraction was unaffected at maximal exercise; hence, maximal leg oxygen uptake increased from 1,511.0 ± 130.1 ml/min before treatment to 1,793.0 ± 148.7 ml/min with rHuEpo and decreased after hemodilution to 1,428.0 ± 111.6 ml/min. Pulmonary oxygen uptake at peak exercise increased from 3,950.0 ± 160.7 before administration to 4,254.5 ± 178.4 ml/min with rHuEpo and decreased to 4,059.0 ± 161.1 ml/min with hemodilution ( P = 0.22, compared with values before rHuEpo treatment). Blood buffer capacity remained unaffected by rHuEpo treatment and hemodilution. The augmented hematocrit did not compromise peak cardiac output. In summary, in healthy humans, rHuEpo increases maximal oxygen consumption due to augmented systemic and muscular peak oxygen delivery.
Collapse
|
26
|
Deudero JJP, Caramelo C, Castellanos MC, Neria F, Fernández-Sánchez R, Calabia O, Peñate S, González-Pacheco FR. Induction of hypoxia-inducible factor 1alpha gene expression by vascular endothelial growth factor. J Biol Chem 2008; 283:11435-44. [PMID: 18305118 DOI: 10.1074/jbc.m703875200] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Transcriptional regulation of vascular endothelial growth factor (VEGF) is critically dependent on hypoxia-inducible factor 1 (HIF-1). However, not only hypoxia, but selected growth factors can induce HIF-1. High levels of both VEGF and HIF-1 coexist in certain conditions, e.g. tumors. Nonetheless, the possibility that the stimulatory relationship between HIF-1 and VEGF may be bi-directional has not been addressed up to date. The present study in endothelial cells analyzed whether HIF-1 is regulated by a product of its own transcriptionally activated genes, namely, VEGF. As a main finding, VEGF-A(165) induced the increase of HIF-1alpha mRNA and HIF-1alpha protein and nuclear translocation. Autologous endothelial cell VEGF mRNA and protein were also increased upon exposure to exogenous VEGF. The signaling implication of reactive oxygen species was examined by comparison with H(2)O(2) and hypoxanthine/xanthine oxidase and by the superoxide dismutase mimetic, MnTMPyP, the Rac1-NAD(P)H oxidase complex inhibitor, apocynin, transfection of a dominant negative Rac1 mutant, and transfection of a p67phox antisense oligonucleotide. Superoxide anion, largely dependent on Rac1-NAD(P)H oxidase complex activity, was the critical signaling element. The transductional functionality of the pathway was confirmed by means of a reporter gene flanked by a transcription site-related VEGF sequence and by quantitative PCR. In summary, the present results reveal a previously undescribed action of VEGF on the expression of its own transcription factor, HIF-1, and on VEGF itself. This effect is principally mediated by superoxide anion, therefore identifying a new, potentially relevant role of reactive oxygen species in VEGF signaling.
Collapse
Affiliation(s)
- Juan José P Deudero
- Laboratorio de Nefrología-Hipertensión, Fundación Jiménez Díaz-Capio, Hospital de la Princesa, Universidad Autónoma de Madrid, 28040 Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Mori S, Sawada T, Kubota K. Asialoerythropoietin is a strong modulator of angiogenesis by bone-marrow cells. J INVEST SURG 2008; 20:357-62. [PMID: 18097877 DOI: 10.1080/08941930701772181] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
The angiogenetic effect of asialoerythropoietin (asialoEPO) was investigated in a murine (BALB/c) ischemic hind limb model created by ligating the right femoral artery, and severe ischemia was created by ligating two points on the femoral artery. The mice were allocated to six groups: 1, Control (n = 15), no treatment; 2, BMC (n = 15), Injected with 1 x 10(6) bone marrow cells (BMC); 3, EPO (n = 15), EPO(500 U/kg for 2 weeks); 4, BMC + EPO (n = 15), BMC + erythropoietin (EPO); 5, asialoEPO (n = 15), asialoEPO (500 U/kg for 2 weeks); 6, BMC + asialoEPO (n = 15); BMC + asialoEPO. Blood flow in the ischemic and normal limbs was measured using a Doppler flowmeter on days 7 and 14. Vessel density was evaluated by immunohistochemical staining for Von Willebrand Factor (vWF). In the severe ischemia model, limb survival was investigated. Blood flow was significantly higher in the BMC + asialoEPO group than in the Control, BMC, EPO, BMC + EPO, or asialoEPO group on day 7 (p = .007) and on day 14 (p = .002). Vascular density was also significantly higher in the BMC + asialoEPO (0.067 +/- 0.022) group than in the Control (0.026 +/- 0.007), BMC (0.027 +/- 0.012) EPO (0.029 +/- 0.002), BMC + EPO (0.048 +/- 0.015), and asialoEPO (0.031 +/- 0.001) groups (p = .006). Finally, limb survival at day 14 in the severe ischemia model was significantly better in the BMC + asialoEPO group (83.3%) than in the Control (40.0%), BMC (52.9%), EPO (44.4%), BMC + EPO (64.7%), or asialoEPO (36.4%) group (p = .02). This provides the conclusions that asialoEPO promotes angiogenesis by BMC and that its action is significantly more potent than that of EPO.
Collapse
Affiliation(s)
- Shozo Mori
- Second Department of Surgery, Dokkyo University School of Medicine, Shimotsuga, Tochigi, Japan
| | | | | |
Collapse
|
28
|
Lundby C, Hellsten Y, Jensen MBF, Munch AS, Pilegaard H. Erythropoietin receptor in human skeletal muscle and the effects of acute and long-term injections with recombinant human erythropoietin on the skeletal muscle. J Appl Physiol (1985) 2008; 104:1154-60. [PMID: 18218911 DOI: 10.1152/japplphysiol.01211.2007] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The presence and potential physiological role of the erythropoietin receptor (Epo-R) were examined in human skeletal muscle. In this study we demonstrate that Epo-R is present in the endothelium, smooth muscle cells, and in fractions of the sarcolemma of skeletal muscle fibers. To study the potential effects of Epo in human skeletal muscle, two separate studies were conducted: one to study the acute effects of a single Epo injection on skeletal muscle gene expression and plasma hormones and another to study the effects of long-term (14 wk) Epo treatment on skeletal muscle structure. Subjects (n = 11) received a single Epo injection of 15,000 IU (double blinded, cross over, placebo). A single Epo injection reduced myoglobin and increased transferrin receptor and MRF-4 mRNA content within 10 h after injection. Plasma hormones remained unaltered. Capillarization and fiber hypertrophy was studied in subjects (n = 8) who received long-term Epo administration, and muscle biopsies were obtained before and after. Epo treatment did not alter mean fiber area (0.84 +/- 0.2 vs. 0.72 +/- 0.3 mm(2)), capillaries per fiber (4.3 +/- 0.5 vs. 4.4 +/- 1.3), or number of proliferating endothelial cells. In conclusion, the Epo-R is present in the vasculature and myocytes in human skeletal muscle, suggesting a role in both cell types. In accordance, a single injection of Epo regulates myoglobin, MRF-4, and transferrin receptor mRNA levels. However, in contrast to our hypothesis, prolonged Epo administration had no apparent effect on capillarization or muscle fiber hypertrophy.
Collapse
Affiliation(s)
- Carsten Lundby
- Copenhagen Muscle Research Centre, Rigshospitalet, section 7652, Blegdamsvej 3, 2100 Copenhagen Ø, Denmark.
| | | | | | | | | |
Collapse
|
29
|
Imamura R, Okumi M, Isaka Y, Ichimaru N, Moriyama T, Imai E, Nonomura N, Takahara S, Okuyama A. Carbamylated Erythropoietin Improves Angiogenesis and Protects the Kidneys from Ischemia-Reperfusion Injury. Cell Transplant 2008; 17:135-41. [DOI: 10.3727/000000008783907044] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Several studies have shown that erythropoietin (EPO) can protect the kidneys from ischemia-reperfusion injury and can raise the hemoglobin (Hb) concentration. Recently, the EPO molecule modified by carbamylation (CEPO) has been identified and was demonstrated to be able to protect several organs without increasing the Hb concentration. We hypothesized that treatment with CEPO would protect the kidneys, partly due to the increased peritubular capillaries. The therapeutic effect of CEPO was evaluated using an endothelial tube formation assay in vitro, and a rat ischemia-reperfusion injury model in vivo. EPO treatment showed the tendency of increased tube formation, while CEPO treatment induced more capillary-like formation than EPO. Ischemia-reperfusion-induced kidneys exhibited characteristic nuclei of apoptosis in tubular epithelial cells with decreased peritubular capillaries, while EPO treatment inhibited tubular apoptosis with preserved endothelial cells. Moreover, CEPO-treated kidneys showed minimal tubular apoptosis with increased peritubular capillary endothelial cells. In conclusion, we identified a new therapeutic approach using CEPO to protect kidneys from ischemia-reperfusion injury by promoting angiogenesis.
Collapse
Affiliation(s)
- Ryoichi Imamura
- Department of Urology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Masayoshi Okumi
- Department of Urology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Yoshitaka Isaka
- Department of Advanced Technology for Transplantation, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Naotsugu Ichimaru
- Department of Urology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Toshiki Moriyama
- Health Care Center, Osaka University, Osaka 560-0043, Japan
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Enyu Imai
- Department of Nephrology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Norio Nonomura
- Department of Urology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Shiro Takahara
- Department of Advanced Technology for Transplantation, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Akihiko Okuyama
- Department of Urology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| |
Collapse
|
30
|
Caramelo C, Justo S, Gil P. Anemia en la insuficiencia cardiaca: fisiopatología, patogenia, tratamiento e incógnitas. Rev Esp Cardiol 2007. [DOI: 10.1157/13108999] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
31
|
Nakano M, Satoh K, Fukumoto Y, Ito Y, Kagaya Y, Ishii N, Sugamura K, Shimokawa H. Important role of erythropoietin receptor to promote VEGF expression and angiogenesis in peripheral ischemia in mice. Circ Res 2007; 100:662-9. [PMID: 17293480 DOI: 10.1161/01.res.0000260179.43672.fe] [Citation(s) in RCA: 139] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
We have recently demonstrated that endogenous erythropoietin (Epo)/Epo receptor (EpoR) system plays an important protective role in hypoxia-induced pulmonary hypertension. However, it remains to be examined whether vascular EpoR system contributes to angiogenesis in response to ischemia. We examined angiogenesis in EpoR(-/-)-rescued mice that lack EpoR in most organs including cardiovascular system except erythroid-lineage cells. Two weeks after femoral artery ligation, blood flow recovery, activation of VEGF/VEGF receptor system, and mobilization of endothelial progenitor cells were all impaired in EpoR(-/-)-rescued mice as compared with wild-type (WT) mice. Bone marrow (BM) transplantation with WT-BM cells in EpoR(-/-)-rescued mice partially but significantly improved blood flow recovery after hindlimb ischemia. The extent of VEGF upregulation and the number of BM-derived cells in ischemic tissue were significantly less in EpoR(-/-)-rescued mice compared with WT mice even after BM reconstitution with WT-BM cells. Similarly, the recovery of blood flow was significantly impaired in recipient EpoR(-/-)-rescued mice that had been transplanted with WT-BM or EpoR(-/-)-rescued-BM as compared with recipient WT mice. Furthermore, the Matrigel implantation assay and aortic ring assay showed that microvessel growth in vitro was significantly reduced in EpoR(-/-)-rescued mice as compared with WT mice. These results indicate that vascular EpoR system also plays an important role in angiogenesis in response to hindlimb ischemia through upregulation of VEGF/VEGF receptor system, both directly by enhancing neovascularization and indirectly by recruiting endothelial progenitor cells and BM-derived proangiogenic cells.
Collapse
Affiliation(s)
- Makoto Nakano
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Sinclair AM, Todd MD, Forsythe K, Knox SJ, Elliott S, Begley CG. Expression and function of erythropoietin receptors in tumors. Cancer 2007; 110:477-88. [PMID: 17582631 DOI: 10.1002/cncr.22832] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Safety concerns surrounding the use of recombinant human erythropoietin (Epo) to treat anemia in cancer patients were raised after 2 recent clinical studies reported a worse survival outcome in patients who received epoetin alpha or epoetin beta compared with patients who received placebo. Although those findings contrasted with previous clinical studies, which demonstrated no difference in survival for cancer patients who received erythropoiesis-stimulating agents (ESAs), some investigators have suggested a potential role for ESAs in promoting tumor growth through 1) stimulation of Epo receptors (EpoR) expressed in tumors, 2) stimulation and formation of tumor vessels, and/or 3) enhanced tumor oxygenation. The first and second hypotheses appeared to be supported by some EpoR expression and ESA in vitro studies. However, these conclusions have been challenged because of poor specificity of EpoR-detection methodologies, conflicting data from different groups, and the lack of correlation between in vitro data and in vivo findings in animal tumor models. For this report, the authors reviewed the biology of EpoR in erythropoiesis and compared and contrasted the reported findings on the role of ESAs and EpoR in tumors.
Collapse
|
33
|
Zhang SX, Ma JX. Ocular neovascularization: Implication of endogenous angiogenic inhibitors and potential therapy. Prog Retin Eye Res 2007; 26:1-37. [PMID: 17074526 DOI: 10.1016/j.preteyeres.2006.09.002] [Citation(s) in RCA: 153] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Ocular neovascularization (NV) is the primary cause of blindness in a wide range of ocular diseases, such as diabetic retinopathy (DR) and age-related macular degeneration (AMD). The exact mechanism underlying the pathogenesis of ocular NV is not yet well understood, and as a consequence, there is no satisfactory therapy for ocular NV. In the last 10 years, a number of studies provided increasing evidence demonstrating that the imbalance between angiogenic stimulating factors and angiogenic inhibitors is a major contributor to the angiogenesis induced by various insults, such as hypoxia or ischemia, inflammation and tumor. The angiogenic inhibitors alone or in combination with other existing therapies are, therefore, believed to be promising in the treatment of ocular NV in the near future. This article reviews recent progress in studies on the mechanisms and treatment of ocular NV, focusing on the implication and therapeutic potential of endogenous angiogenic inhibitors in ocular NV.
Collapse
Affiliation(s)
- Sarah X Zhang
- Department of Medicine Endocrinology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | | |
Collapse
|
34
|
Abstract
Promising results have been obtained with bevacizumab (Avestin; Genentech, Inc.; South San Francisco, CA) in clinical trials in patients with a range of solid tumors; however, to maximize the potential of this agent, further research is needed to clarify a number of important issues. These include the optimization of bevacizumab dosage and schedule of administration, the potential value of this agent in combination with other treatment modalities like chemotherapy and radiation, the management of toxicities, and the selection of patients most likely to benefit from treatment. Intriguing results from two recent phase III trials highlight the need for a better understanding of the best ways to incorporate bevacizumab into clinical practice. Ultimately, maximizing the potential value of this agent may require a more thorough understanding of bevacizumab's mechanism of action and the pathways mediating resistance.
Collapse
Affiliation(s)
- Emily Bergsland
- Department of Medicine, University of California, San Francisco, California 94115, USA.
| | | |
Collapse
|
35
|
Galeano M, Altavilla D, Bitto A, Minutoli L, Calò M, Lo Cascio P, Polito F, Giugliano G, Squadrito G, Mioni C, Giuliani D, Venuti FS, Squadrito F. Recombinant human erythropoietin improves angiogenesis and wound healing in experimental burn wounds. Crit Care Med 2006; 34:1139-46. [PMID: 16484928 DOI: 10.1097/01.ccm.0000206468.18653.ec] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVE Erythropoietin interacts with vascular endothelial growth factor (VEGF) and stimulates endothelial cell mitosis and motility; thus it may be of importance in the complex phenomenon of wound healing. The purpose of this study was to investigate the effect of recombinant human erythropoietin (rHuEPO) on experimental burn wounds. DESIGN Randomized experiment. SETTING Research laboratory. SUBJECTS C57BL/6 male mice weighing 25-30 g. INTERVENTIONS Mice were immersed in 80 degrees C water for 10 secs to achieve a deep-dermal second degree burn. Animals were randomized to receive either rHuEPO (400 units/kg/day for 14 days in 100 microL subcutaneously) or its vehicle alone (100 microl/day distilled water for 14 days subcutaneously). On day 14 the animals were killed. Burn areas were used for histologic examination, evaluation of neoangiogenesis by immunohistochemistry, and expression (Western blot) of the specific endothelial marker CD31 as well as quantification of microvessel density, measurement of VEGF wound content (enzyme-linked immunosorbent assay), expression (Western blot) of endothelial and inducible nitric oxide synthases, and determination of wound nitric oxide (NO) products. MEASUREMENTS AND MAIN RESULTS rHuEPO increased burn wound reepithelialization and reduced the time to final wound closure. These effects were completely abated by a passive immunization with specific antibodies against erythropoietin. rHuEPO improved healing of burn wound through increased epithelial proliferation, maturation of the extracellular matrix, and angiogenesis. The hematopoietic factor augmented neoangiogenesis as suggested by the marked increase in microvessel density and by the robust expression of the specific endothelial marker CD31. Furthermore, rHuEPO enhanced the wound content of VEGF caused a marked expression of endothelial and inducible nitric oxide synthases and increased wound content of nitric oxide products. CONCLUSIONS Our study suggests that rHuEPO may be an effective therapeutic approach to improve clinical outcomes after thermal injury.
Collapse
Affiliation(s)
- Mariarosaria Galeano
- Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Buemi M, Caccamo C, Nostro L, Cavallaro E, Floccari F, Grasso G. Brain and cancer: the protective role of erythropoietin. Med Res Rev 2005; 25:245-59. [PMID: 15389732 DOI: 10.1002/med.20012] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Erythropoietin (Epo) is a pleiotropic agent, that is to say, it can act on several cell types in different ways. An independent system Epo/Epo receptor (EpoR) was detected in brain, leading to the hypothesis that this hormone could be involved in cerebral functions. Epo/EpoR expression changes during ontogenesis, thus indicating the importance of this system in neurodevelopment. Moreover, the hypoxia-induced production of Epo in the adult brain suggests that it could exert a neurotrophic and neuroprotective effect in case of brain injury. Epo could also influence neurotransmission, inducing neurotransmitters (NT) release. Epo therapy in anemic cancer patients is still a controversial issue, because of its possible action as a growth and an angiogenic factor. In our speculative hypothesis Epo could be involved in a "two steps process" that, after a neovascularization phase, leads to its down regulation. Moreover, Epo-activated signaling pathways could be modulated as possible targets to interfere in neoplastic cells cycle. In conclusion, treatment with rHuEpo could change therapeutical perspectives in different pathological conditions, such as central nervous system (CNS) diseases, but further studies are needed to clarify its physiopathological activities in different clinical fields.
Collapse
Affiliation(s)
- Michele Buemi
- Chair of Nephrology, Department of Internal Medicine, University of Messina, Italy.
| | | | | | | | | | | |
Collapse
|
37
|
Lissoni P, Perego M, Veronese E, Fumagalli G, Brivio F, Colciago M, Messina G, Rovelli F, Brivio R, Gardani G. Endocrine effects of erythropoietin in cancer patients. Hematology 2005; 9:363-7. [PMID: 15763975 DOI: 10.1080/10245330400001876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
The recent advances in the knowledge of the psychoneuroimmunological pathogenesis of human neoplasms have demonstrated the existence of feed-back mechanisms operating between interleukins and endocrine secretions, which play an important role in the regulation of the immune responses, including the anticancer immunity. In contrast, few studies only have been performed to investigate the possible relation between endocrine activities and hematopoietic growth factors. The present study was performed to analyze the acute endocrine effects of erythropoietin-alpha (EPO) on the main endocrine secretions. The study was carried out in 10 advanced solid tumor patients. EPO was injected subcutaneously at a dose of 10,000 U, and venous blood samples were collected before and 2, 4 and 6 h after EPO administration. No significant changes in mean serum levels of FSH, LH and TSH were seen in response to EPO. Cortisol and DHEAS concentrations increased after EPO injection, whereas those of PRL decreased, but none of these differences was statistically significant. Finally, mean serum levels of both growth hormone (GH) and somatomedin-C (IGF-1) significantly decreased after EPO administration. This preliminary study shows that EPO may inhibit GH secretion from the pituitary gland and IGF-1 production. Since GH would stimulate EPO release, the results of this study may suggest the existence of feedback mechanism operating between GH secretion and EPO production, with inhibitory effect of EPO on GH secretion, and stimulatory action of GH on EPO production. Therefore, this study would describe the first example of hemato-endocrine feedback mechanisms. Moreover, this study, by showing an inhibitory effect of EPO on IGF-1 secretion, would suggest a possible use of EPO in the medical oncology not only for the treatment of cancer related anemia, but also to counteract tumor growth by blocking IGF-1 production, which has been proven to be a growth factor for several tumor histotypes. Obviously, IGF-1 is not the only tumor growth factor, but it could play a fundamental role in the regulation of production and activity of several other tumor growth factors. In any case, this study describes the only acute endocrine effects of EPO. Therefore, further studies, by evaluating the endocrine effects of a chronic treatment with EPO, will be required to establish which may be its effect on IGF-1 endogenous production, and its consequence on survival time.
Collapse
Affiliation(s)
- Paolo Lissoni
- Division of Radiation Oncology, San Gerardo Hospital, Milano, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Lenz T, Gauer S, Weich HA, Haak T, Bergner R, Gossmann J. Vascular endothelial growth factor and its soluble receptor, Flt-1, are not correlated to erythropoietin in diabetics with normal or reduced renal function. Nephrology (Carlton) 2005; 10:84-9. [PMID: 15705187 DOI: 10.1111/j.1440-1797.2005.00366.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND AIMS Recombinant erythropoietin upregulates the expression of the vascular endothelial growth factor (VEGF) receptors, Flt-1 (VEGFR-1) and KDR/Flk-1 (VEGFR-2), in endothelial cells. The integrity of the VEGF system seems to be crucial for the regulation of endothelial permeability and thus for the avoidance of renal protein leakage. As albuminuria/proteinuria is a hallmark of diabetic nephropathy, we examined cross-sectionally in 35 type 1 and 37 type 2 diabetic patients with various degrees of renal dysfunction and albuminuria whether there was an interrelationship between intrinsic erythropoietin (EPO) and VEGF/Flt-1. METHODS AND RESULTS In patients with plasma creatinine values < or =1.5 (n = 53) or >1.5 mg/dL (n = 19), the mean serum EPO was 5.6 +/- 4.4 and 10.2 +/- 7.0 mU/mL (P = 0.02), respectively. In the two groups, urinary and serum VEGF(165) concentrations were similarly distributed (mean 94.3 +/- 91.8 vs 108 +/- 72.2 ng/L and 91.7 +/- 76.8 vs 91.9 +/- 74.9 ng/L, respectively; both P = NS). The mean urinary Flt-1 for the two groups amounted to 0.14 +/- 0.35 and 0.51 +/- 0.93 ng/mL (P = 0.045), respectively. No correlation between VEGF or Flt-1 and EPO was apparent. CONCLUSION Our data suggest that in vivo EPO does not affect the functionality and/or production of components of the VEGF/Flt-1 system in diabetics with normal or reduced renal function.
Collapse
Affiliation(s)
- Tomas Lenz
- Medical Clinic IV - Nephrology, University Hospital, Frankfurt am Main, Germany
| | | | | | | | | | | |
Collapse
|
39
|
Galeano M, Altavilla D, Cucinotta D, Russo GT, Calò M, Bitto A, Marini H, Marini R, Adamo EB, Seminara P, Minutoli L, Torre V, Squadrito F. Recombinant human erythropoietin stimulates angiogenesis and wound healing in the genetically diabetic mouse. Diabetes 2004; 53:2509-17. [PMID: 15331568 DOI: 10.2337/diabetes.53.9.2509] [Citation(s) in RCA: 121] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The effects of recombinant human erythropoietin (rHuEPO) in diabetes-related healing defects were investigated by using an incisional skin-wound model produced on the back of female diabetic C57BL/KsJ-m(+/+)Lept(db) mice (db(+)/db(+)) and their normoglycemic littermates (db(+/+)m). Animals were treated with rHuEPO (400 units/kg in 100 microl s.c.) or its vehicle alone (100 microl). Mice were killed on different days (3, 6, and 12 days after skin injury) for measurement of vascular endothelial growth factor (VEGF) mRNA expression and protein synthesis, for monitoring angiogenesis by CD31 expression, and for evaluating histological changes. Furthermore, we evaluated wound-breaking strength at day 12. At day 6, rHuEPO injection in diabetic mice resulted in an increase in VEGF mRNA expression (vehicle = 0.33 +/- 0.1 relative amount of mRNA; rHuEPO = 0.9 +/- 0.09 relative amount of mRNA; P < 0.05) and protein wound content (vehicle = 23 +/- 5 pg/wound; rHuEPO = 92 +/- 12 pg/wound; P < 0.05) and caused a marked increase in CD31 gene expression (vehicle = 0.18 +/- 0.05 relative amount of mRNA; rHuEPO = 0.98 +/- 0.21 relative amount of mRNA; P < 0.05) and protein synthesis. Furthermore, rHuEPO injection improved the impaired wound healing and, at day 12, increased the wound-breaking strength in diabetic mice (vehicle = 12 +/- 2 g/mm; rHuEPO 21 +/- 5 g/mm; P < 0.05). Erythropoietin may have a potential application in diabetes-related wound disorders.
Collapse
Affiliation(s)
- Mariarosaria Galeano
- Department of ClinicalExperimental Medicine, Section of Pharmacology, Azienda Ospedaliera Universitaria "G Martino," Torre Biologica, 5th Floor, Via Consolare Valeria Gazzi, 98125 Messina, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Buemi M, Galeano M, Sturiale A, Ientile R, Crisafulli C, Parisi A, Catania M, Calapai G, Impalà P, Aloisi C, Squadrito F, Altavilla D, Bitto A, Tuccari G, Frisina N. RECOMBINANT HUMAN ERYTHROPOIETIN STIMULATES ANGIOGENESIS AND HEALING OF ISCHEMIC SKIN WOUNDS. Shock 2004; 22:169-73. [PMID: 15257091 DOI: 10.1097/01.shk.0000133591.47776.bd] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Wound healing in ischemic tissues such as flap margins due to inadequate blood supply is still a source of considerable morbidity in surgical practice. Adequate tissue perfusion is particularly important in wound healing. We investigated the effects of recombinant human erythropoietin (rHuEPO) on wound healing in an ischemic skin wound model. Sixty-three Sprague-Dawley rats were used. Normal incisional wound and H-shaped double flaps were used as the wound models. Animals were treated with rHuEPO (400 IU/kg) or its vehicle. Rats were killed on different days (3, 5, and 10 days after skin injury) and the wounded skin tissues were used for immunohistochemistry and for analysis of vascular endothelial growth factor content and collagen content. Tissue transglutaminase immunostaining of histological specimens was used as a vascular marker to determine the level of microvessel density. The results showed a higher level of vascular endothelial growth factor protein and an increased microvessel density in ischemic wounds with rHuEPO treatment than the normal incisional wounds and ischemic control wounds. Collagen content was higher in the incisional wounds and in the ischemic wounds with rHuEPO treatment compared with the ischemic control wounds. Our results suggest that erythropoietin may be an effective therapeutic approach in improving healing in ischemic skin wounds.
Collapse
Affiliation(s)
- Michele Buemi
- Department of Internal Medicine, University of Messina, Messina, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
Erythropoietin (Epo) is a hematopoietic growth factor and cytokine which stimulates erythropoiesis. In recent years, Epo has been shown to have important nonhematopoietic functions in the nervous system. Nonerythropoietic actions of Epo include a critical role in the development, maintenance, protection and repair of the nervous system. A wide variety of experimental studies have shown that Epo and its receptor are expressed in the nervous system and Epo exerts remarkable neuroprotection in cell culture and animal models of nervous system disorders. In this review, we summarize the current knowledge on the neurotrophic and neuroprotective properties of Epo, the mechanisms by which Epo produces neuroprotection and the signal transduction systems regulated by Epo in the nervous system.
Collapse
Affiliation(s)
- Sermin Genc
- Department of Medical Biology and Genetics, School of Medicine, Dokuz Eylul University, Inciralti, 35340, Izmir, Turkey.
| | | | | |
Collapse
|
42
|
Abstract
Many cancer patients suffer from anemia, which has a major detrimental effect on their quality of life. Recombinant human erythropoietin (rHuEPO) is now widely used in cancer patients, as it improves hematocrit, lowers blood transfusion requirements, and improves quality of life. Recent research indicates that EPO has pleiotropic effects on the body well beyond the maintenance of red cell mass, but the mechanisms involved in relieving fatigue and improving quality of life in cancer patients are poorly understood. EPO receptors (EPO-Rs) have been detected in many different cells and tissues, providing evidence for autocrine, paracrine, and endocrine functions of EPO. Apart from its endocrine function, EPO may have a generalized role as an antiapoptotic agent that is associated with enhancement of muscle tone, mucosal status, and gonadal and cognitive function. The recent discovery of EPO-Rs in breast tumor vasculature, while raising important questions about the possible effects of pharmacological doses of rHuEPO on tumor cells, also suggests that the receptors could provide a useful target for drugs attached to EPO.
Collapse
Affiliation(s)
- Terence R Lappin
- Haematology, Cancer Research Centre, Belfast City Hospital, Queen's University, Belfast, Northern Ireland.
| | | | | |
Collapse
|
43
|
Juul S. Erythropoietin in the central nervous system, and its use to prevent hypoxic-ischemic brain damage. ACTA PAEDIATRICA (OSLO, NORWAY : 1992). SUPPLEMENT 2003; 91:36-42. [PMID: 12477263 DOI: 10.1111/j.1651-2227.2002.tb02904.x] [Citation(s) in RCA: 116] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
UNLABELLED A new field of clinical and scientific interest has recently developed based on the discovery that the hematopoietic cytokine erythropoietin (Epo) has important non-hematopoietic functions in the brain and other organs, particularly during development. The biological effects of Epo in the central nervous system (CNS) involve activation of its specific receptor and corresponding signal transduction pathways. Epo receptor expression is abundant in the developing mammalian brain, and decreases as term approaches. Epo has been identified as a neurotrophic and neuroprotective agent in a wide variety of experimental paradigms, from neuronal cell culture to in vivo models of brain injury. Several mechanisms by which Epo produces neuroprotection are recognized. Epo (i) decreases glutamate toxicity, (ii) induces the generation of neuronal anti-apoptotic factors, (iii) reduces inflammation, (iv) decreases nitric oxide-mediated injury, and (v) has direct antioxidant effects. CONCLUSION Collectively, the evidence suggests that Epo may provide a new approach to the treatment of a variety of CNS disorders in adults and children, especially as a possible therapy for perinatal asphyxia. This review summarizes the current knowledge on the neurotrophic and neuroprotective functions of Epo in the developing and injured brain.
Collapse
Affiliation(s)
- S Juul
- Department of Pediatrics, Division of Neonatology, University of Washington, Seattle, Washington 98195, USA.
| |
Collapse
|
44
|
Ashley RA, Dubuque SH, Dvorak B, Woodward SS, Williams SK, Kling PJ. Erythropoietin stimulates vasculogenesis in neonatal rat mesenteric microvascular endothelial cells. Pediatr Res 2002; 51:472-8. [PMID: 11919332 DOI: 10.1203/00006450-200204000-00012] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Human breast milk is a rich source of growth factors, including erythropoietin (Epo), the endogenous hormonal stimulant of erythropoiesis. Recombinant human Epo (rhEpo) has been shown to stimulate 1) angiogenesis, the process of new blood vessel growth from preexisting vessels; 2) vasculogenesis, tubule formation from single-cell suspensions; and 3) endothelial cell proliferation in immortalized endothelial cells and vessel explants. We hypothesized that Epo would induce mitogenesis and stimulate vasculogenesis in primary cultures of microvascular endothelial cells (MVECs) from neonatal rat mesentery. Isolation, purification, characterization, and culture of MVECs were performed. Cell proliferative effects of rhEpo were studied by 3-(4,5 dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay in cultured MVECs. Vasculogenic effects of rhEpo were examined on cultured MVECs plated on either hormone-rich Matrigel substratum or the extracellular matrix protein, type I collagen. Our findings show that MVECs are isolated and purified, and that rhEpo stimulates MVEC proliferation, with maximal proliferation seen with a concentration of 50 IU/mL rhEpo. Tubule formation assays reveal that an rhEpo concentration of 50 IU/mL produces maximal tubule formation after 12 h on both Matrigel and the simple substratum, type I collagen. Our study is the first to examine the effects of rhEpo on the endothelium of the neonatal gastrointestinal tract. These data suggest that Epo may have a trophic effect on the vasculature of the gastrointestinal tract early in development. Furthermore, as Epo has been measured in breast milk, and its receptor has been shown to exist on the mucosa and gastrointestinal vasculature, Epo may be an endogenous stimulant of vessel growth during neonatal gastrointestinal development.
Collapse
Affiliation(s)
- Richard A Ashley
- Mayo Medical School, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | | | | | | | | |
Collapse
|
45
|
Saghizadeh M, Chwa M, Aoki A, Lin B, Pirouzmanesh A, Brown DJ, Ljubimov AV, Kenney MC. Altered expression of growth factors and cytokines in keratoconus, bullous keratopathy and diabetic human corneas. Exp Eye Res 2001; 73:179-89. [PMID: 11446768 DOI: 10.1006/exer.2001.1028] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The purpose of this study was to identify the growth factors and cytokines present in normal and diseased corneas. Total RNA was isolated from normal and diseased corneas. cDNA was synthesized from individual corneas and semiquantitative reverse transcription-polymerase chain reaction (RT-PCR) was performed with primers to IL-1alpha, 1IL-8, PDGF-B, BMP-2, BMP-4, IGF-I, TGF-beta2, FGF-2, and VEGF. After normalization to beta2-microglobulin, several factors were identified that were significantly different from normal. Antibodies to IGF-I, BMP-2, VEGF and TGF-beta2 were used for immunohistochemistry. A total of 93 corneas were used for this study including 31 normal, 20 keratoconus, 19 bullous keratopathy (pseudophakic and aphakic, PBK/ABK), and 23 diabetic corneas. The VEGF RNA levels were significantly decreased in the keratoconus and PBK/ABK corneas but increased in the diabetic corneas. BMP-2 gene expression was lower than normal in the PBK/ABK and diabetic corneas. IGF-I and BMP-4 RNA levels were increased in PBK/ABK. In the immunohistochemical studies, the protein patterns paralleled those found at the mRNA level. The only exception was IGF-I in diabetic corneas that showed increased staining in the epithelium and its basement membrane without a significant increase in mRNA levels. TGF-beta2 mRNA and protein levels were similar to normal in all diseased corneas. Thus, no alterations in the tested growth factors/cytokines were unique to keratoconus corneas. In contrast, PBK/ABK corneas had specific significant elevations of BMP-4 and IGF-I. Diabetic corneas were unique in their increased VEGF mRNA levels. These data suggest that while some growth factor/cytokine alterations are non-specific and can be found in multiple corneal diseases, there are others that are unique to that disease.
Collapse
Affiliation(s)
- M Saghizadeh
- Ophthalmology Research Laboratories, Burns and Allen Research Institute, Cedars-Sinai Medical Center, UCLA Medical School Affiliate, Los Angeles, CA 90048, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Epo was once regarded as a cytokine with only hematopoietic effects. It is now clear that the distributions of Epo and Epo-R are more widespread in the developing human. Epo-R is widely distributed during early fetal development, leading to speculation that Epo acts in concert with other growth factors to optimize growth and development. Areas in which Epo has important recognized effects are on endothelial cells, and in the developing heart, gastrointestinal tract, and brain. It may also be important in the regulation of vascular growth during the menstrual cycle, and in the stimulation of testosterone production in men. Epo and Epo-R are prominent in the brain during fetal development, leading to speculation that they play an important role in neurodevelopment. There are also promising data regarding rEpo as a possible neuroprotective agent in such conditions as hypoxia, because it decreases programmed cell death induced during such adverse conditions. It is unlikely, however, that rEpo crosses the blood-brain barrier in normal premature infants, and it is not clear whether the CNS effects of rEpo, should it cross the blood-brain barrier, are harmful or beneficial in the setting of a developing brain.
Collapse
Affiliation(s)
- S E Juul
- Division of Neonatology, University of Florida College of Medicine, Gainesville, USA.
| |
Collapse
|
47
|
|
48
|
Nitta K, Uchida K, Kimata N, Honda K, Kobayashi H, Kawashima A, Yumura W, Nihei H. Recombinant human erythropoietin stimulates vascular endothelial growth factor release by glomerular endothelial cells. Eur J Pharmacol 1999; 373:121-4. [PMID: 10408258 DOI: 10.1016/s0014-2999(99)00281-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
We designed the present study to address the question of whether recombinant human erythropoietin stimulates DNA synthesis and vascular endothelial growth factor (VEGF) secretion in vitro using cultured bovine glomerular endothelial cells (GENs). Recombinant human erythropoietin dose-dependently stimulated the proliferation of GENs in culture, and this proliferative effect was inhibited by 1 microg/ml anti-VEGF antiserum. The increase in VEGF concentrations in the supernatants containing 10 U/ml rHuEpo was abolished by incubation with 10 microg/ml of anti-human rHuEPO antiserum, 0.2 microg/ml actinomycin D or 10 microg/ml cycloheximide. Taken together, rHuEpo stimulates GEN proliferation in vitro and VEGF release from these cells is associated with stimulation of RNA-dependent DNA and protein synthesis.
Collapse
Affiliation(s)
- K Nitta
- Department of Medicine, Kidney Center, Tokyo Women's Medical University, Japan
| | | | | | | | | | | | | | | |
Collapse
|