51
|
Athanasiadou I, Dokoumetzidis A, Voss SC, El Saftawy W, Al-Maadheed M, Valsami G, Georgakopoulos C. Hyperhydration Effect on Pharmacokinetic Parameters and Detection Sensitivity of Recombinant Human Erythropoietin in Urine and Serum Doping Control Analysis of Males. J Pharm Sci 2019; 108:2162-2172. [DOI: 10.1016/j.xphs.2019.01.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 01/13/2019] [Accepted: 01/15/2019] [Indexed: 10/27/2022]
|
52
|
Macias-Velez RJ, Fukushima-Díaz de León L, Beas-Zárate C, Rivera-Cervantes MC. Intranasal Erythropoietin Protects CA1 Hippocampal Cells, Modulated by Specific Time Pattern Molecular Changes After Ischemic Damage in Rats. J Mol Neurosci 2019; 68:590-602. [PMID: 31054091 DOI: 10.1007/s12031-019-01308-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 03/20/2019] [Indexed: 12/21/2022]
Abstract
Erythropoietin, a multitarget molecule exhibited neuroprotective properties, especially against cerebral ischemia. However, little effort has been made to determinate both the administration pathway and doses that diminishes neuronal damage. In this study, we investigate the effect on CA1 region of different intranasal doses of rHuEPO (500, 1000 and 2500 IU/kg) applied in distinct post-damage times (1, 6, and 24 h) against ischemic cellular damage. Furthermore, most effective dose and time were used to evaluate gen and protein expression changes in 3 key molecules (EPO, EPOR, and βcR). We established that CA1-region present histopathological damage in this ischemia model and that rHuEPO protects cells against damage, particularly at 1000 IU dose. Molecular data shows that EPO and EPOR gene expression are upregulated in a short term after damage treatment with rHuEPO (1 h); oppositely, BcR is upregulated in ischemic and Isc + EPO. Protein expression data displays no changes on EPO expression in evaluated times after treatment, but a tendency to increase 24 h after damage; in the opposite way, EPOR is upregulated significantly 6 h after treatment and this effect last until 24 h. So, our data suggest that a single intranasal dose of rHuEPO (1 h post-injury) provides histological neurorestoration in CA1 hippocampal region, even if we did not observe a dose-dependent dose effect, the medium dose evaluated (1000 UI/kg of b.w.) was more effective and sufficient for induces molecular changes that provides a platform for neuroprotection.
Collapse
Affiliation(s)
- R J Macias-Velez
- Laboratorio de Neurobiología Celular, Departamento de Biología Celular y Molecular, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - L Fukushima-Díaz de León
- Laboratorio de Neurobiología Celular, Departamento de Biología Celular y Molecular, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - C Beas-Zárate
- Laboratorio de Regeneración Neural y Desarrollo Neural, Departamento de Biología Celular y Molecular, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | - M C Rivera-Cervantes
- Laboratorio de Neurobiología Celular, Departamento de Biología Celular y Molecular, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico.
| |
Collapse
|
53
|
Li Y, Zhou Y, Zhang D, Wu W, Kang X, Wu Q, Wang P, Liu X, Gao G, Zhou Y, Wang G, Chang Y. Hypobaric hypoxia regulates iron metabolism in rats. J Cell Biochem 2019; 120:14076-14087. [DOI: 10.1002/jcb.28683] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 01/22/2019] [Accepted: 01/24/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Yaru Li
- Laboratory of Molecular Iron Metabolism, The Key Laboratory of Animal Physiology, Biochemistry, and Molecular Biology of Hebei Province, College of Life Science Hebei Normal University Shijiazhuang Hebei China
| | - Yue Zhou
- Laboratory of Molecular Iron Metabolism, The Key Laboratory of Animal Physiology, Biochemistry, and Molecular Biology of Hebei Province, College of Life Science Hebei Normal University Shijiazhuang Hebei China
| | - Dong Zhang
- Laboratory of Molecular Iron Metabolism, The Key Laboratory of Animal Physiology, Biochemistry, and Molecular Biology of Hebei Province, College of Life Science Hebei Normal University Shijiazhuang Hebei China
| | - Wen‐Yue Wu
- Laboratory of Molecular Iron Metabolism, The Key Laboratory of Animal Physiology, Biochemistry, and Molecular Biology of Hebei Province, College of Life Science Hebei Normal University Shijiazhuang Hebei China
| | - Xiaoxuan Kang
- Laboratory of Molecular Iron Metabolism, The Key Laboratory of Animal Physiology, Biochemistry, and Molecular Biology of Hebei Province, College of Life Science Hebei Normal University Shijiazhuang Hebei China
| | - Qiong Wu
- Laboratory of Molecular Iron Metabolism, The Key Laboratory of Animal Physiology, Biochemistry, and Molecular Biology of Hebei Province, College of Life Science Hebei Normal University Shijiazhuang Hebei China
| | - Peina Wang
- Laboratory of Molecular Iron Metabolism, The Key Laboratory of Animal Physiology, Biochemistry, and Molecular Biology of Hebei Province, College of Life Science Hebei Normal University Shijiazhuang Hebei China
| | - Xiaopeng Liu
- Laboratory of Molecular Iron Metabolism, The Key Laboratory of Animal Physiology, Biochemistry, and Molecular Biology of Hebei Province, College of Life Science Hebei Normal University Shijiazhuang Hebei China
- Department of Neurosurgery The Second Hospital of Hebei Medical University Shijiazhuang Hebei China
| | - Guofen Gao
- Laboratory of Molecular Iron Metabolism, The Key Laboratory of Animal Physiology, Biochemistry, and Molecular Biology of Hebei Province, College of Life Science Hebei Normal University Shijiazhuang Hebei China
| | - Yaru Zhou
- Department of Endocrinology The Third Hospital of Hebei Medical University Shijiazhuang Hebei China
| | - Guangyou Wang
- Department of Neurobiology, Heilongjiang Provincial Key Laboratory of Neurobiology Harbin Medical University Harbin Heilongjiang China
| | - Yan‐Zhong Chang
- Laboratory of Molecular Iron Metabolism, The Key Laboratory of Animal Physiology, Biochemistry, and Molecular Biology of Hebei Province, College of Life Science Hebei Normal University Shijiazhuang Hebei China
| |
Collapse
|
54
|
Martin TD, Green MS, Whitehead MT, Scheett TP, Webster MJ, Hudson GM. Six weeks of oral Echinacea purpurea supplementation does not enhance the production of serum erythropoietin or erythropoietic status in recreationally active males with above-average aerobic fitness. Appl Physiol Nutr Metab 2019; 44:791-795. [PMID: 30608872 DOI: 10.1139/apnm-2018-0783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The purpose of this study was to investigate the effect of 6 weeks of oral Echinacea purpurea supplementation on serum erythropoietin (EPO) and erythropoietic status. Twenty-four males (mean ± SE; age = 25.2 ± 1.4 years, height = 178.1 ± 1.4 cm, body mass = 78.1 ± 1.6 kg, body fat = 12.7 ± 0.9%, maximal oxygen uptake = 52.9 ± 0.9 mL·kg-1·min-1) were randomly grouped using a matched-pair, double-blind design and self-administered 8000 mg·day-1 of either E. purpurea (n = 12) or placebo (n = 12) for 42 consecutive days. Blood samples were collected prior to supplementation (day 0) and every 2 weeks during the supplementation period (days 14, 28, and 42) and were analyzed for EPO, red blood cell count, hemoglobin concentration, hematocrit, mean corpuscular volume, and mean corpuscular hemoglobin concentration. Separate 2 × 4 (group × time) factorial ANOVA with repeated measures on time were used to determine statistical differences with significance set at p ≤ 0.05. There were no significant interaction, group, or time effects observed for EPO or erythropoietic status markers for any of the measurement points (p ≤ 0.05). The present study indicated that 6 weeks of oral E. purpurea supplementation in recreationally active males with above average aerobic fitness did not enhance EPO or erythropoietic status. These findings are in contrast with previous reports of E. purpurea supplementation in untrained participants with average fitness levels, but consistent with observations in trained endurance athletes.
Collapse
Affiliation(s)
- Tyler D Martin
- a Department of Kinesiology and Health Promotion, Troy University, Troy, AL 36082, USA
| | - Michael S Green
- a Department of Kinesiology and Health Promotion, Troy University, Troy, AL 36082, USA
| | - Malcolm T Whitehead
- b Department of Kinesiology and Health Science, Stephen F. Austin State University, Nacogdoches, TX 75962, USA
| | - Timothy P Scheett
- c Department of Health and Human Performance, College of Charleston, Charleston, SC 29424, USA
| | - Michael J Webster
- d College of Nursing and Health Sciences, Valdosta State University, Valdosta, GA 31698, USA
| | - Geoffrey M Hudson
- e Department of Health, Kinesiology, & Sport, The University of South Alabama, Mobile, AL 36688, USA
| |
Collapse
|
55
|
Baig MA, Panchal SS. Streptozotocin-Induced Diabetes Mellitus in Neonatal Rats: An Insight into its Applications to Induce Diabetic Complications. Curr Diabetes Rev 2019; 16:26-39. [PMID: 30973111 DOI: 10.2174/1573399815666190411115829] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 03/04/2019] [Accepted: 04/05/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Diabetic complications are the major contributor in the mortality of diabetic patients despite controlling blood glucose level. In the journey of new drug discovery, animal models have to play a major role. A large number of chemical-induced and genetically modified animal models have been investigated to induce diabetic complications but none of them was found to be mimicking the pathophysiology of the human. Therefore, the search and identification of the appropriate animal model become essential. OBJECTIVE In the present review, we have made an attempt to understand the pathophysiology of diabetic complication in the neonatal streptozotocin-diabetic rat model and tried to identify the targets for therapeutic agents. The review will help the researchers to explore the animal model to induce diabetic complications, to identify targets and further to find lead molecules for treatment or prevention of diabetic complications. METHODS We have compiled the available research work from 1974 by using prominent databases, organized the available information and analyzed the data to improve the understanding of the pathophysiology of streptozotocin-induced diabetic complications in neonates of rats. RESULTS The neonatal streptozotocin-diabetic rat model is frequently used and well-established animal model for type 2 diabetes mellitus. We have found that this model has been used to study the pathogenesis of various micro and macrovascular diabetic complications and also investigated for its effects on the liver, thymus gland, and brain. The underlying pathophysiology for complications had a resemblance to the human. CONCLUSION The neonatal streptozotocin-diabetic rat model may demonstrate symptomatic diabetic complications due to persistent hyperglycemia at the age of approximately 18-24 weeks. Critical interpretations of available research work showed that the researcher can explore split dose STZ (90- 100mg/kg b.w) model to induce Type 2 DM in neonates of rats at 2nd or 3rd postnatal day.
Collapse
Affiliation(s)
- Mirza Anwar Baig
- Department of Pharmacology, AI's Kalsekar Technical Campus, School of Pharmacy, Navi Mumbai, Maharashtra, India
- Department of Pharmcology, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| | - Shital Sharad Panchal
- Department of Pharmcology, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| |
Collapse
|
56
|
Fuchs O. Treatment of Lymphoid and Myeloid Malignancies by Immunomodulatory Drugs. Cardiovasc Hematol Disord Drug Targets 2019; 19:51-78. [PMID: 29788898 DOI: 10.2174/1871529x18666180522073855] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 05/05/2018] [Accepted: 05/14/2018] [Indexed: 06/08/2023]
Abstract
Thalidomide and its derivatives (lenalidomide, pomalidomide, avadomide, iberdomide hydrochoride, CC-885 and CC-90009) form the family of immunomodulatory drugs (IMiDs). Lenalidomide (CC5013, Revlimid®) was approved by the US FDA and the EMA for the treatment of multiple myeloma (MM) patients, low or intermediate-1 risk transfusion-dependent myelodysplastic syndrome (MDS) with chromosome 5q deletion [del(5q)] and relapsed and/or refractory mantle cell lymphoma following bortezomib. Lenalidomide has also been studied in clinical trials and has shown promising activity in chronic lymphocytic leukemia (CLL) and non-Hodgkin lymphoma (NHL). Lenalidomide has anti-inflammatory effects and inhibits angiogenesis. Pomalidomide (CC4047, Imnovid® [EU], Pomalyst® [USA]) was approved for advanced MM insensitive to bortezomib and lenalidomide. Other IMiDs are in phases 1 and 2 of clinical trials. Cereblon (CRBN) seems to have an important role in IMiDs action in both lymphoid and myeloid hematological malignancies. Cereblon acts as the substrate receptor of a cullin-4 really interesting new gene (RING) E3 ubiquitin ligase CRL4CRBN. This E3 ubiquitin ligase in the absence of lenalidomide ubiquitinates CRBN itself and the other components of CRL4CRBN complex. Presence of lenalidomide changes specificity of CRL4CRBN which ubiquitinates two transcription factors, IKZF1 (Ikaros) and IKZF3 (Aiolos), and casein kinase 1α (CK1α) and marks them for degradation in proteasomes. Both these transcription factors (IKZF1 and IKZF3) stimulate proliferation of MM cells and inhibit T cells. Low CRBN level was connected with insensitivity of MM cells to lenalidomide. Lenalidomide decreases expression of protein argonaute-2, which binds to cereblon. Argonaute-2 seems to be an important drug target against IMiDs resistance in MM cells. Lenalidomide decreases also basigin and monocarboxylate transporter 1 in MM cells. MM cells with low expression of Ikaros, Aiolos and basigin are more sensitive to lenalidomide treatment. The CK1α gene (CSNK1A1) is located on 5q32 in commonly deleted region (CDR) in del(5q) MDS. Inhibition of CK1α sensitizes del(5q) MDS cells to lenalidomide. CK1α mediates also survival of malignant plasma cells in MM. Though, inhibition of CK1α is a potential novel therapy not only in del(5q) MDS but also in MM. High level of full length CRBN mRNA in mononuclear cells of bone marrow and of peripheral blood seems to be necessary for successful therapy of del(5q) MDS with lenalidomide. While transfusion independence (TI) after lenalidomide treatment is more than 60% in MDS patients with del(5q), only 25% TI and substantially shorter duration of response with occurrence of neutropenia and thrombocytopenia were achieved in lower risk MDS patients with normal karyotype treated with lenalidomide. Shortage of the biomarkers for lenalidomide response in these MDS patients is the main problem up to now.
Collapse
Affiliation(s)
- Ota Fuchs
- Institute of Hematology and Blood Transfusion, U Nemocnice 1, 128 20 Prague 2, Czech Republic
| |
Collapse
|
57
|
Leigh J, Juriasingani S, Akbari M, Shao P, Saha MN, Lobb I, Bachtler M, Fernandez B, Qian Z, van Goor H, Pasch A, Feelisch M, Wang R, Sener A. Endogenous H 2S production deficiencies lead to impaired renal erythropoietin production. Can Urol Assoc J 2018; 13:E210-E219. [PMID: 30472982 DOI: 10.5489/cuaj.5658] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Patients suffering from chronic kidney disease (CKD) experience a number of associated comorbidities, including anemia. Relative deficiency in renal erythropoietin (EPO) production is thought to be a primary cause of anemia. Interestingly, CKD patients display low levels of hydrogen sulfide (H2S), an endogenously derived renal oxygen sensor. Previous in vitro experiments have revealed that H2S-deficient renal cell lines produce less EPO than wild-type renal cell lines during hypoxia. METHODS We postulated that H2S might be a primary mediator of EPO synthesis during hypoxia, which was tested using an in vivo murine model of whole-body hypoxia and in clinical samples obtained from CKD patients. RESULTS Following a 72-hour period of hypoxia (11% O2), partial H2S knockout mice (lacking the H2S biosynthetic enzyme cystathionine γ-lyase [CSE]) displayed lower levels of hemoglobin, EPO and cystathionine-β-synthase (CBS) (another H2S biosynthetic enzyme) compared to wild-type mice, all of which was rescued by exogenous H2S supplementation. We also found that anemic CKD patients requiring exogenous EPO exhibited lower urinary thiosulfate levels compared to non-anemic CKD patients of similar CKD classification. CONCLUSIONS Together, our results confirm an interplay between the actions of H2S during hypoxia and EPO production.
Collapse
Affiliation(s)
- Jennifer Leigh
- Department of Microbiology and Immunology, Western University, London, ON, Canada.,Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, London, ON, Canada
| | - Smriti Juriasingani
- Department of Microbiology and Immunology, Western University, London, ON, Canada.,Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, London, ON, Canada
| | - Masoud Akbari
- Department of Microbiology and Immunology, Western University, London, ON, Canada.,Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, London, ON, Canada
| | - Peng Shao
- Department of Physiology, Western University, London, ON, Canada
| | - Manujendra N Saha
- Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, London, ON, Canada
| | - Ian Lobb
- Department of Microbiology and Immunology, Western University, London, ON, Canada.,Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, London, ON, Canada
| | - Matthias Bachtler
- Department of Clinical Research, University of Bern, Bern, Switzerland
| | - Bernadette Fernandez
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Zhongming Qian
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, Shanghai, China
| | - Harry van Goor
- Department of Pathology and Medical Biology, University of Groningen, Netherlands.,University Medical Center Groningen, Netherlands
| | - Andreas Pasch
- Department of Clinical Research, University of Bern, Bern, Switzerland.,Department of Clinical Chemistry, University Hospital Bern (Inselspital), Bern, Switzerland
| | - Martin Feelisch
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Rui Wang
- The Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, ON, Canada
| | - Alp Sener
- Department of Microbiology and Immunology, Western University, London, ON, Canada.,Department of Surgery, Western University, London, ON, Canada.,Multi-Organ Transplant Program, London Health Sciences Center, London, ON, Canada.,Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, London, ON, Canada
| |
Collapse
|
58
|
Deregulated iron metabolism in bone marrow from adenine-induced mouse model of chronic kidney disease. Int J Hematol 2018; 109:59-69. [DOI: 10.1007/s12185-018-2531-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 09/04/2018] [Accepted: 09/05/2018] [Indexed: 12/11/2022]
|
59
|
EPO does not promote interaction between the erythropoietin and beta-common receptors. Sci Rep 2018; 8:12457. [PMID: 30127368 PMCID: PMC6102255 DOI: 10.1038/s41598-018-29865-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 07/19/2018] [Indexed: 12/13/2022] Open
Abstract
A direct interaction between the erythropoietin (EPOR) and the beta-common (βc) receptors to form an Innate Repair Receptor (IRR) is controversial. On one hand, studies have shown a functional link between EPOR and βc receptor in tissue protection while others have shown no involvement of the βc receptor in tissue repair. To date there is no biophysical evidence to confirm a direct association of the two receptors either in vitro or in vivo. We investigated the existence of an interaction between the extracellular regions of EPOR and the βc receptor in silico and in vitro (either in the presence or absence of EPO or EPO-derived peptide ARA290). Although a possible interaction between EPOR and βc was suggested by our computational and genomic studies, our in vitro biophysical analysis demonstrates that the extracellular regions of the two receptors do not specifically associate. We also explored the involvement of the βc receptor gene (Csf2rb) under anaemic stress conditions and found no requirement for the βc receptor in mice. In light of these studies, we conclude that the extracellular regions of the EPOR and the βc receptor do not directly interact and that the IRR is not involved in anaemic stress.
Collapse
|
60
|
Kudenchuk PJ. Erythropoietin for Out-of-Hospital Cardiac Arrest: Growing Together or Apart? J Am Coll Cardiol 2018; 68:50-2. [PMID: 27364050 DOI: 10.1016/j.jacc.2016.03.598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 03/02/2016] [Indexed: 11/18/2022]
Affiliation(s)
- Peter J Kudenchuk
- Department of Medicine, Division of Cardiology/Arrhythmia Services and King County Emergency Medical Services, Seattle, Washington.
| |
Collapse
|
61
|
Günal MY, Ozansoy M, Kılıç Ü, Keskin İ, Özdemir EM, Aslan İ, Eren Z, Ersavaş C, Kılıç E. Role of erythropoietin and its receptor in the development of endometriosis in rats. J Turk Ger Gynecol Assoc 2018; 20:41-46. [PMID: 29916217 PMCID: PMC6501872 DOI: 10.4274/jtgga.galenos.2018.2018.0039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Objective: Besides its hematopoietic function, erythropoietin (EPO) may protect tissues from degenerative disorders. As such, EPO and its receptors were revealed in nonhematopoietic cells, including stromal and endometrial epithelial cells. However, the role of EPO in endometrial disorders is still unknown. Here, we aimed to examine the role of EPO and its receptor activation in the development of endometriosis in rats. Material and Methods: Animals were treated with EPO, darbepoietin (the synthetic form of EPO) or EPO’s receptor activator, methoxy polyethylene glycol-epoetin beta (MIRCERA), after development of endometriosis. Endometriosis was induced by estrogen-administration following surgical attachment of endometrial surface on the inner abdominal wall. Treatments were started 3 weeks after induction of endometriosis and continued for the following 3 weeks. For the analysis of recurrence of endometriosis, additional analyses were conducted 3 weeks after cessation of treatments. Results: As compared with vehicle-treated animals, lesion size was reduced significantly and recurrence of endometriosis was not observed in all treatment groups. Histopathologic examination revealed that EPO and darbepoietin were more effective than MIRCERA- and vehicle-treated animals. Conclusion: Here we provide evidence that EPO is a promising candidate for the treatment of endometriosis. Our histopathologic results in particular indicate that EPO is more effective than its receptor activator MIRCERA in the development endometriosis.
Collapse
Affiliation(s)
- Mehmet Yalçın Günal
- Department of Physiology, Alanya Alaaddin Keykubat University School of Medicine, Antalya, Turkey,Regenerative and Restorative Medical Research Center (REMER), İstanbul Medipol University, İstanbul, Turkey
| | - Mehmet Ozansoy
- Department of Physiology, İstanbul Medipol University School of Medicine, İstanbul, Turkey,Regenerative and Restorative Medical Research Center (REMER), İstanbul Medipol University, İstanbul, Turkey
| | - Ülkan Kılıç
- Department of Medical Biology, University of Health Sciences School of Medicine, İstanbul, Turkey,Regenerative and Restorative Medical Research Center (REMER), İstanbul Medipol University, İstanbul, Turkey
| | - İlknur Keskin
- Department of Histology and Embryology, İstanbul Medipol University School of Medicine, İstanbul, Turkey,Regenerative and Restorative Medical Research Center (REMER), İstanbul Medipol University, İstanbul, Turkey
| | - Ekrem Musa Özdemir
- Experimental Animal Center, İstanbul Medipol University, İstanbul, Turkey
| | - İsmail Aslan
- Department of Pharmaceutical Technology, Yeditepe University School of Pharmacy, İstanbul, Turkey
| | - Zehra Eren
- Department of Nephrology, Yeditepe University School of Medicine, İstanbul, Turkey
| | - Cenk Ersavaş
- Department of General Surgery, İstanbul Medipol University School of Medicine, İstanbul, Turkey,Regenerative and Restorative Medical Research Center (REMER), İstanbul Medipol University, İstanbul, Turkey
| | - Ertuğrul Kılıç
- Department of Physiology, İstanbul Medipol University School of Medicine, İstanbul, Turkey,Regenerative and Restorative Medical Research Center (REMER), İstanbul Medipol University, İstanbul, Turkey
| |
Collapse
|
62
|
Different effects of granulocyte colony-stimulating factor and erythropoietin on erythropoiesis. Stem Cell Res Ther 2018; 9:119. [PMID: 29720275 PMCID: PMC5930863 DOI: 10.1186/s13287-018-0877-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 04/08/2018] [Accepted: 04/16/2018] [Indexed: 11/24/2022] Open
Abstract
Background Red blood cells are the most abundant cells in the blood that deliver oxygen to the whole body. Erythropoietin (EPO), a positive regulator of erythropoiesis, is currently the major treatment for chronic anemia. Granulocyte colony-stimulating factor (G-CSF) is a multifunctional cytokine and a well-known regulator of hematopoietic stem cell proliferation, differentiation, and mobilization. The use of EPO in combination with G-CSF has been reported to synergistically improve erythroid responses in a group of patients with myelodysplastic syndromes who did not respond to EPO treatment alone; however, the mechanism remains unclear. Methods C57BL/6 J mice injected with G-CSF or EPO were used to compare the erythropoiesis status and the efficiency of erythroid mobilization by flow cytometry. Results In this study, we found that G-CSF induced more orthochromatophilic erythroblast production than did EPO in the bone marrow and spleen. In addition, in contrast to EPO treatments, G-CSF treatments enhanced the efficiency of the mobilization of newly synthesized reticulocytes into peripheral blood. Our results demonstrated that the effects of G-CSF on erythropoiesis and erythrocytic mobilization were independent of EPO secretion and, in contrast to EPO, G-CSF promoted progression of erythropoiesis through transition of early stage R2 (basophilic erythroblasts) to late stage R4 (orthochromatophilic erythroblasts). Conclusions We demonstrate for the first time that G-CSF treatments induce a faster erythropoiesis-enhancing response than that of EPO. These findings suggest an alternative approach to treating acute anemia, especially when patients are experiencing a clinical emergency in remote areas without proper blood bank supplies.
Collapse
|
63
|
Güven Bağla A, Içkin Gülen M, Ercan F, Aşgün F, Ercan E, Bakar C. Changes in kidney tissue and effects of erythropoietin after acute heart failure. Biotech Histochem 2018; 93:340-353. [PMID: 29671622 DOI: 10.1080/10520295.2018.1443347] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
Abstract
Impairment of cardiac function causes renal damage. Renal failure after heart failure is attributed to hemodynamic derangement including reduced renal perfusion and increased venous pressure. One mechanism involves apoptosis and is defined as cardiorenal syndrome type 1. Erythropoietin (EPO) is a cytokine that induces erythropoiesis under hypoxic conditions. Hypoxia inducible factor 1 alpha (HIF-1α) plays a regulatory role in cellular response to hypoxia. Protective effects of EPO on heart, kidney and nervous system are unrelated to red blood cell production. We investigated early changes in and effects of EPO on renal tissues of rats with myocardial infarction by morphology and immunohistochemistry. Coronary artery ligation was used to induce myocardial infarction in Wistar rats. Group 1 comprised sham operated rats; groups 2, 3 and 4 included rats after coronary artery ligation that were sacrificed 6 h after ligation and that were treated with saline, 5,000 U/kg EPO or 10,000 U/kg EPO, respectively; group 5 included rats sacrificed 1 h after ligation. Group 2 showed increased renal tubule damage. Significantly less tubule damage was observed in EPO treated groups. EPO and EPO receptor (EPO-R) immunostaining intensities increased slightly for group 5 and became more intense for group 2. EPO and EPO-R immunostaining was observed in the interstitial area, glomerular cells and tubule epithelial cells of EPO treated groups. HIF-1α immunostaining was observed in collecting tubules in the medulla only in group 2. Caspase-3 immunostaining is an indicator of apoptosis. Caspase-3 staining intensity decreased in renal medulla of EPO treated groups. EPO treatment may exert a protective effect on the renal tissues of patients with cardiorenal syndrome.
Collapse
Affiliation(s)
- A Güven Bağla
- a Çanakkale Onsekiz Mart University , School of Medicine, Department of Histology and Embryology , Çanakkale
| | - M Içkin Gülen
- a Çanakkale Onsekiz Mart University , School of Medicine, Department of Histology and Embryology , Çanakkale
| | - F Ercan
- b Marmara University , School of Medicine, Department of Histology and Embryology , Istanbul
| | - F Aşgün
- c Çanakkale Onsekiz Mart University , School of Medicine, Department of Cardiovascular Surgery , Çanakkale
| | - E Ercan
- d Department of Cardiology , Medical Park Hospital , Izmir
| | - C Bakar
- e Çanakkale Onsekiz Mart University , School of Medicine, Department of Public Health , Çanakkale , Turkey
| |
Collapse
|
64
|
Castillo C, Zaror S, Gonzalez M, Hidalgo A, Burgos CF, Cabezas OI, Hugues F, Jiménez SP, González-Horta E, González-Chavarría I, Gavilán J, Montesino R, Sánchez O, Lopez MG, Fuentealba J, Toledo JR. Neuroprotective effect of a new variant of Epo nonhematopoietic against oxidative stress. Redox Biol 2018; 14:285-294. [PMID: 28987867 PMCID: PMC5975214 DOI: 10.1016/j.redox.2017.09.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/05/2017] [Accepted: 09/18/2017] [Indexed: 12/19/2022] Open
Abstract
Human erythropoietin is mainly recognized for its hematopoietic function; however, by binding to its receptor (EpoR), it can activate different signaling pathways as STAT, PI3K, MAPK and RAS to increase cellular differentiation or provide neuroprotective effects, among others. A recombinant human erythropoietin variant with low glycosylation and without hematopoietic effect (EpoL) was purified from skimmed goat milk. Recombinant human erythropoietin (Epo) was obtained from CHO cell line and used as control to compare EpoL effects. Neuroprotection studies were performed in PC12 cells and rat hippocampal slices. Cells were pretreated during 1h with EpoL or Epo and exposed to oxidative agents (H2O2 or FCCP); cell viability was assayed at the end of the experiment by the MTT method. Hippocampal slices were exposed to 15min of oxygen and glucose deprivation (OGD) and the neuroprotective drugs EpoL or Epo were incubated for 2h post-OGD in re-oxygenated medium. Cell cultures stressed with oxidative agents, and pretreated with EpoL, showed neuroprotective effects of 30% at a concentration 10 times lower than that of Epo. Moreover, similar differences were observed in OGD ex vivo assays. Neuroprotection elicited by EpoL was lost when an antibody against EpoR was present, indicating that its effect is EpoR-dependent. In conclusion, our results suggest that EpoL has a more potent neuroprotective profile than Epo against oxidative stress, mediated by activation of EpoR, thus EpoL represents an important target to develop a potential biopharmaceutical to treat different central nervous system pathologies related to oxidative stress such as stroke or neurodegenerative diseases.
Collapse
Affiliation(s)
- C Castillo
- Biotechnology and Biopharmaceutical Laboratory, Pathophysiology Department, Universidad de Concepción, Victor Lamas 1290, P.O. Box 160-C, Concepción, Chile
| | - S Zaror
- Biotechnology and Biopharmaceutical Laboratory, Pathophysiology Department, Universidad de Concepción, Victor Lamas 1290, P.O. Box 160-C, Concepción, Chile
| | - M Gonzalez
- Biotechnology and Biopharmaceutical Laboratory, Pathophysiology Department, Universidad de Concepción, Victor Lamas 1290, P.O. Box 160-C, Concepción, Chile
| | - A Hidalgo
- Biotechnology and Biopharmaceutical Laboratory, Pathophysiology Department, Universidad de Concepción, Victor Lamas 1290, P.O. Box 160-C, Concepción, Chile
| | - C F Burgos
- Laboratory of Screening of Neuroactive Compound, Physiology Department. School of Biological Sciences, Universidad de Concepción, Victor Lamas 1290, P.O. Box 160-C, Concepción, Chile
| | - O I Cabezas
- Clinical Sciences Department, School of Veterinary Sciences, Universidad de Concepción, Avenida Vicente Méndez 595, Chillan, Chile
| | - F Hugues
- Clinical Sciences Department, School of Veterinary Sciences, Universidad de Concepción, Avenida Vicente Méndez 595, Chillan, Chile
| | - S P Jiménez
- Biotechnology and Biopharmaceutical Laboratory, Pathophysiology Department, Universidad de Concepción, Victor Lamas 1290, P.O. Box 160-C, Concepción, Chile
| | - E González-Horta
- Biotechnology and Biopharmaceutical Laboratory, Pathophysiology Department, Universidad de Concepción, Victor Lamas 1290, P.O. Box 160-C, Concepción, Chile
| | - I González-Chavarría
- Biotechnology and Biopharmaceutical Laboratory, Pathophysiology Department, Universidad de Concepción, Victor Lamas 1290, P.O. Box 160-C, Concepción, Chile
| | - J Gavilán
- Laboratory of Screening of Neuroactive Compound, Physiology Department. School of Biological Sciences, Universidad de Concepción, Victor Lamas 1290, P.O. Box 160-C, Concepción, Chile
| | - R Montesino
- Biotechnology and Biopharmaceutical Laboratory, Pathophysiology Department, Universidad de Concepción, Victor Lamas 1290, P.O. Box 160-C, Concepción, Chile
| | - O Sánchez
- Biotechnology and Biopharmaceutical Laboratory, Pathophysiology Department, Universidad de Concepción, Victor Lamas 1290, P.O. Box 160-C, Concepción, Chile
| | - Manuela G Lopez
- Department of Pharmacology and Therapeutics, "Instituo Teófilo Hernando", Universidad Autónoma de Madrid, Spain
| | - J Fuentealba
- Laboratory of Screening of Neuroactive Compound, Physiology Department. School of Biological Sciences, Universidad de Concepción, Victor Lamas 1290, P.O. Box 160-C, Concepción, Chile
| | - J R Toledo
- Biotechnology and Biopharmaceutical Laboratory, Pathophysiology Department, Universidad de Concepción, Victor Lamas 1290, P.O. Box 160-C, Concepción, Chile.
| |
Collapse
|
65
|
Broadgate S, Kiire C, Halford S, Chong V. Diabetic macular oedema: under-represented in the genetic analysis of diabetic retinopathy. Acta Ophthalmol 2018; 96 Suppl A111:1-51. [PMID: 29682912 DOI: 10.1111/aos.13678] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 11/21/2017] [Indexed: 12/15/2022]
Abstract
Diabetic retinopathy, a complication of both type 1 and type 2 diabetes, is a complex disease and is one of the leading causes of blindness in adults worldwide. It can be divided into distinct subclasses, one of which is diabetic macular oedema. Diabetic macular oedema can occur at any time in diabetic retinopathy and is the most common cause of vision loss in patients with type 2 diabetes. The purpose of this review is to summarize the large number of genetic association studies that have been performed in cohorts of patients with type 2 diabetes and published in English-language journals up to February 2017. Many of these studies have produced positive associations with gene polymorphisms and diabetic retinopathy. However, this review highlights that within this large body of work, studies specifically addressing a genetic association with diabetic macular oedema, although present, are vastly under-represented. We also highlight that many of the studies have small patient numbers and that meta-analyses often inappropriately combine patient data sets. We conclude that there will continue to be conflicting results and no meaningful findings will be achieved if the historical approach of combining all diabetic retinopathy disease states within patient cohorts continues in future studies. This review also identifies several genes that would be interesting to analyse in large, well-defined cohorts of patients with diabetic macular oedema in future candidate gene association studies.
Collapse
Affiliation(s)
- Suzanne Broadgate
- Nuffield Laboratory of Ophthalmology; Nuffield Department of Clinical Neurosciences; University of Oxford; Oxford UK
| | - Christine Kiire
- Nuffield Laboratory of Ophthalmology; Nuffield Department of Clinical Neurosciences; University of Oxford; Oxford UK
- Oxford Eye Hospital; John Radcliffe Hospital; Oxford University NHS Foundation Trust; Oxford UK
| | - Stephanie Halford
- Nuffield Laboratory of Ophthalmology; Nuffield Department of Clinical Neurosciences; University of Oxford; Oxford UK
| | - Victor Chong
- Nuffield Laboratory of Ophthalmology; Nuffield Department of Clinical Neurosciences; University of Oxford; Oxford UK
| |
Collapse
|
66
|
Anoshkina Y, Costas-Rodríguez M, Speeckaert M, Van Biesen W, Delanghe J, Vanhaecke F. Iron isotopic composition of blood serum in anemia of chronic kidney disease. Metallomics 2018; 9:517-524. [PMID: 28417130 DOI: 10.1039/c7mt00021a] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Chronic kidney disease (CKD) is a general term for disorders that affect the structure and function of the kidneys. Iron deficiency (ID) and anemia occur in the vast majority of CKD patients, most of whom are elderly. However, establishing the cause of anemia in CKD, and therefore making an informed decision concerning the corresponding therapeutic treatment, is still a challenge. High-precision Fe isotopic analysis of blood serum samples of CKD patients with and without ID/anemia was performed via multi-collector inductively coupled plasma-mass spectrometry (MC-ICP-MS) for such a purpose. Patients with CKD and/or iron disorders showed a heavier serum Fe isotopic composition than controls. Many clinical parameters used for the diagnosis and follow-up of anemia correlated significantly with the serum Fe isotopic composition. In contrast, no relation was observed between the serum Fe isotopic composition and the estimated glomerular filtration rate as a measure of kidney function. Among the CKD patients, the serum Fe isotopic composition was substantially heavier in the occurrence of ID anemia, while erythropoietin-related anemia did not exert this effect. The Fe isotopic composition can thus be useful for distinguishing these different types of anemias in CKD patients, i.e. ID anemia vs. erythropoietin-related anemia.
Collapse
Affiliation(s)
- Yulia Anoshkina
- Department of Analytical Chemistry, Ghent University, Campus Sterre, Krijgslaan 281-S12, BE-9000 Ghent, Belgium.
| | | | | | | | | | | |
Collapse
|
67
|
Singh D, Kaur G, Bakshi D, Sahota J, Thakur A, Grover S. Evaluation of Hemoglobin Concentration and Hematocrit Values in Temporomandibular Joint Ankylosis Patients in Comparison to Nonankylosed Patients. J Contemp Dent Pract 2018; 19:210-213. [PMID: 29422472 DOI: 10.5005/jp-journals-10024-2238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
AIM The aim of this study is to find if there is any correlation between the hematological parameters and temporomandibular joint (TMJ) ankylosis and severity of the disease in such patients when compared with the nonankylosed patients. MATERIALS AND METHODS A total of 70 patients with age ranging from 10 to 40 years were included in the study after excluding the subjects according to the inclusion criteria. We categorized the subjects into two major groups: group I: control (nonankylosed/ healthy subjects) and group II: study group (ankylosed subjects) with each group containing 35 subjects (n = 35) respectively. A detailed personal and medical history was obtained. The pharynx diameter was also recorded for each patient, and blood investigations using venous blood were done, which included hemoglobin concentration and hematocrit values. RESULTS The results of study population showed a mean age of 22 ± 2.2 years. The most common etiology reported was trauma (65.7%) followed by infections, in which Noma was the most common one (80%). The difference of the mean values for hemoglobin and hematocrit concentration, between both the groups, was found to be statistically significant (p < 0.0001). Furthermore, a positive correlation was observed between the hemoglobin concentration and duration of ankylosis. CONCLUSION This study was an attempt to find a relation between the hemoglobin and hematocrit values in TMJ ankylosis patients so that the clinical treatment and management of such patients during surgeries be improved and may be beneficial for the patient. CLINICAL SIGNIFICANCE Temporomandibular joint ankylosis patients have to undergo complex surgical treatment, where the risk of excessive blood loss is high. Therefore, considering the complications of blood transfusions, such as infections and other risk factors, these patients can be good subjects for autologous blood transfusions, which help in improvement of the overall well-being of the patient.
Collapse
Affiliation(s)
- Deepinder Singh
- Private Practitioner, Department of Oral and Maxillofacial Surgery, Ludhiana, Punjab, India
| | - Guneet Kaur
- Private Practitioner, Department of Periodontology, Ludhiana, Punjab, India
| | - Dipanshu Bakshi
- Department of Dentistry, Chintpurni Medical College & Hospital, Pathankot, Punjab, India
| | - Jasjit Sahota
- Department of Periodontics, Institute of Dental Sciences Jammu, Jammu and Kashmir, India
| | - Ambika Thakur
- Department of Dentistry, Chintpurni Medical College & Hospital, Pathankot, Punjab, India
| | - Shekhar Grover
- Department of Public Health Dentistry, Maulana Azad Institute of Dental Sciences, New Delhi, India, Phone: +919501544877, e-mail:
| |
Collapse
|
68
|
Beck J, Henschel C, Chou J, Lin A, Del Balzo U. Evaluation of the Carcinogenic Potential of Roxadustat (FG-4592), a Small Molecule Inhibitor of Hypoxia-Inducible Factor Prolyl Hydroxylase in CD-1 Mice and Sprague Dawley Rats. Int J Toxicol 2017; 36:427-439. [PMID: 29153032 DOI: 10.1177/1091581817737232] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The carcinogenic potential of roxadustat (FG-4592), a novel orally active, heterocyclic small molecule inhibitor of hypoxia-inducible factor prolyl hydroxylase (HIF-PH) enzymes in clinical development for treatment of anemia, was evaluated in CD-1 mice and Sprague Dawley rats. Inhibition of HIF-PH by roxadustat leads to a rapid increase in cytoplasmic HIF-α concentrations, followed by translocation of HIF-α to the nucleus and upregulation of HIF-responsive genes, including erythropoietin. Roxadustat was dosed by oral gavage 3 times weekly (TIW) for up to 104 weeks in mice at 0, 15, 30, and 60 mg/kg and in rats at 0, 2.5, 5, and 10 mg/kg. Treatment-associated changes in hematology parameters were consistent with the pharmacologic activity of roxadustat and included elevations in hematocrit in mice at 30 and 60 mg/kg TIW and elevations in erythrocyte count, hemoglobin, hematocrit, and red cell distribution width in rats at 10 mg/kg TIW. No increase in mortality or neoplastic effects compared with vehicle controls was observed after roxadustat treatment in either species. No treatment-related nonneoplastic findings were observed in mice, whereas nonneoplastic microscopic findings in rats were limited to atrial/aortic thromboses at 10 mg/kg TIW males and bone marrow hypercellularity in all treated male and female groups, consistent with the pharmacology of roxadustat. In conclusion, roxadustat administered by oral gavage to mice and rats TIW for up to 104 weeks resulted in dose-dependent exposure and hematologic effects with no effect on survival or development of neoplastic lesions at up to 60 mg/kg in mice and up to 10 mg/kg in rats.
Collapse
Affiliation(s)
| | | | | | - Al Lin
- 1 FibroGen, Inc, San Francisco, CA, USA
| | | |
Collapse
|
69
|
Xiao X, Zhang Y, Wu J, Jia L. Poly(norepinephrine)-coated open tubular column for the separation of proteins and recombination human erythropoietin by capillary electrochromatography. J Sep Sci 2017; 40:4636-4644. [DOI: 10.1002/jssc.201700720] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 09/04/2017] [Accepted: 10/01/2017] [Indexed: 01/28/2023]
Affiliation(s)
- Xue Xiao
- Ministry of Education Key Laboratory of Laser Life Science & Institute of Laser Life Science; College of Biophotonics; South China Normal University; Guangzhou China
| | - Yamin Zhang
- Ministry of Education Key Laboratory of Laser Life Science & Institute of Laser Life Science; College of Biophotonics; South China Normal University; Guangzhou China
| | - Jia Wu
- Ministry of Education Key Laboratory of Laser Life Science & Institute of Laser Life Science; College of Biophotonics; South China Normal University; Guangzhou China
| | - Li Jia
- Ministry of Education Key Laboratory of Laser Life Science & Institute of Laser Life Science; College of Biophotonics; South China Normal University; Guangzhou China
| |
Collapse
|
70
|
Zhang J, Zhao D, Na N, Li H, Miao B, Hong L, Huang Z. Renoprotective effect of erythropoietin via modulation of the STAT6/MAPK/NF-κB pathway in ischemia/reperfusion injury after renal transplantation. Int J Mol Med 2017; 41:25-32. [PMID: 29115389 PMCID: PMC5746301 DOI: 10.3892/ijmm.2017.3204] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 10/02/2017] [Indexed: 12/23/2022] Open
Abstract
Ischemia/reperfusion injury (IRI) commonly occurs in renal transplantation. Erythropoietin (EPO) exerts a protective effect in IRI. To investigate the underlying molecular mechanism, rat models of renal IRI were established and treated with EPO and/or lentivirus-mediated EPO-siRNA, the signal transducer and activator of transcription 6 (STAT6) inhibitor AS1517499, the JNK inhibitor SP600125, the p38 mitogen-activated protein kinase (MAPK) inhibitor SB203580, and the nuclear factor (NF)-κB inhibitor lactacystin. Histological examination revealed that EPO protected the kidney from IRI, through decreasing the extent of tissue congestion and inflammatory cell infiltration; however, EPO siRNA did not exert the same protective effect. In addition, the EPO level was inversely associated with renal IRI. EPO downregulated the expression of interferon-γ, interleukin (IL)-4, creatinine and caspase-3, and upregulated the expression of IL-10, thymic stromal lymphopoietin, STAT6, p-JNK and p-p38, while the opposite effects were observed with the administration of EPO-siRNA and the specific respective inhibitors. Further results revealed that MAPK (p-JNK and p-p38) acted upstream of NF-κB, and that NF-κB signaling regulated the expression of caspase-1 and -3, which may be responsible for the cytotoxicity associated with IRI. Taken together, the results of the present study demonstrated that EPO exerted a protective effect in renal IRI via the STAT6/MAPK/NF-κB pathway. This protective effect of EPO may improve reperfusion tolerance in ischemic kidneys and benefit transplant recipients.
Collapse
Affiliation(s)
- Jinhua Zhang
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Daqiang Zhao
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Ning Na
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Heng Li
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Bin Miao
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Liangqing Hong
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Zhengyu Huang
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| |
Collapse
|
71
|
Moransard M, Bednar M, Frei K, Gassmann M, Ogunshola OO. Erythropoietin reduces experimental autoimmune encephalomyelitis severity via neuroprotective mechanisms. J Neuroinflammation 2017; 14:202. [PMID: 29029628 PMCID: PMC5640948 DOI: 10.1186/s12974-017-0976-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 10/03/2017] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Treatment with erythropoietin (Epo) in experimental autoimmune encephalomyelitis (EAE), the rodent model of multiple sclerosis (MS), has consistently been shown to ameliorate disease progression and improve overall outcome. The effect has been attributed to modulation of the immune response and/or preservation of the central nervous system (CNS) tissue integrity. It remains unclear, however, if (a) Epo acts primarily in the CNS or the periphery and if (b) Epo's beneficial effect in EAE is mainly due to maintaining CNS tissue integrity or to modulation of the immune response. If Epo acts primarily by modulating the immune system, where is this modulation required? In the periphery, the CNS or both? METHODS To address these questions, we used two well-characterized transgenic mouse strains that constitutively overexpress recombinant human Epo (rhEpo) either systemically (tg6) or in CNS only (tg21) in a MOG-induced EAE model. We assessed clinical severity, disease progression, immunomodulation, and CNS tissue integrity, including neuronal survival. RESULTS Although disease onset remained unaffected, EAE progression was alleviated in transgenic animals compared to controls with both lines performing equally well showing that expression of Epo in the periphery is not required; Epo expression in the CNS is sufficient. Immunomodulation was observed in both strains but surprisingly the profile of modulation differed substantially between strains. Modulation in the tg21 strain was limited to a reduction in macrophages in the CNS, with no peripheral immunomodulatory effects observed. In contrast, in the tg6 strain, macrophages were upregulated in the CNS, and, in the periphery of this strain, T cells and macrophages were downregulated. The lack of a consistent immunomodulatory profile across both transgenic species suggests that immunomodulation by Epo is unlikely to be the primary mechanism driving amelioration of EAE. Finally, CNS tissue integrity was affected in all strains. Although myelin appeared equally damaged in all strains, neuronal survival was significantly improved in the spinal cord of tg21 mice, indicating that Epo may ameliorate EAE predominantly by protecting neurons. CONCLUSIONS Our data suggests that moderate elevated brain Epo levels provide clinically significant neuroprotection in EAE without modulation of the immune response making a significant contribution.
Collapse
Affiliation(s)
- M Moransard
- Department of Internal Medicine, Section of Clinical Immunology, University Hospital Zürich, Zurich, Switzerland
| | - M Bednar
- Institute of Veterinary Physiology and Zurich Center of Integrative Human Physiology (ZIHP), Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 260, CH-8057, Zurich, Switzerland
| | - K Frei
- Department of Neurosurgery, University Hospital Zurich, CH-8006, Zurich, Switzerland
| | - M Gassmann
- Institute of Veterinary Physiology and Zurich Center of Integrative Human Physiology (ZIHP), Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 260, CH-8057, Zurich, Switzerland
- Universidad Peruana Cayetano Heredia (UPCH), Lima, Peru
| | - O O Ogunshola
- Institute of Veterinary Physiology and Zurich Center of Integrative Human Physiology (ZIHP), Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 260, CH-8057, Zurich, Switzerland.
| |
Collapse
|
72
|
Hasan S, Mosier MJ, Szilagyi A, Gamelli RL, Muthumalaiappan K. Discrete β-adrenergic mechanisms regulate early and late erythropoiesis in erythropoietin-resistant anemia. Surgery 2017; 162:901-916. [PMID: 28716301 PMCID: PMC5675564 DOI: 10.1016/j.surg.2017.06.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 05/07/2017] [Accepted: 06/03/2017] [Indexed: 01/16/2023]
Abstract
BACKGROUND Anemia of critical illness is resistant to exogenous erythropoietin. Packed red blood cells transfusions is the only treatment option, and despite related cost and morbidity, there is a need for alternate strategies. Erythrocyte development can be divided into erythropoietin-dependent and erythropoietin-independent stages. We have shown previously that erythropoietin-dependent development is intact in burn patients and the erythropoietin-independent early commitment stage, which is regulated by β1/β2-adrenergic mechanisms, is compromised. Utilizing the scald burn injury model, we studied erythropoietin-independent late maturation stages and the effect of β1/β2, β-2, or β-3 blockade in burn mediated erythropoietin-resistant anemia. METHODS Burn mice were randomized to receive daily injections of propranolol (nonselective β1/β2 antagonist), nadolol (long-acting β1/β2 antagonist), butoxamine (selective β2 antagonist), or SR59230A (selective β3 antagonist) for 6 days after burn. Total bone marrow cells were characterized as nonerythroid cells, early and late erythroblasts, nucleated orthochromatic erythroblasts and enucleated reticulocyte subsets using CD71, Ter119, and Syto-16 by flow cytometry. Multipotential progenitors were probed for MafB expressing cells. RESULTS Although propranolol improved early and late erythroblasts, only butoxamine and selective β3-antagonist administrations were positively reflected in the peripheral blood hemoglobin and red blood cells count. While burn impeded early commitment and late maturation stages, β1/β2 antagonism increased the early erythroblasts through commitment stages via β2 specific MafB regulation. β3 antagonism was more effective in improving overall red blood cells through late maturation stages. CONCLUSION The study unfolds novel β2 and β3 adrenergic mechanisms orchestrating erythropoietin resistant anemia after burn, which impedes both the early commitment stage and the late maturation stages, respectively.
Collapse
Affiliation(s)
- Shirin Hasan
- Department of Surgery, Loyola University Chicago, Health Sciences Division, Maywood, IL; Burn and Shock Trauma Research Institute, Loyola University Chicago, Health Sciences Division, Maywood, IL
| | - Michael J Mosier
- Department of Surgery, Loyola University Chicago, Health Sciences Division, Maywood, IL; Burn and Shock Trauma Research Institute, Loyola University Chicago, Health Sciences Division, Maywood, IL
| | - Andrea Szilagyi
- Burn and Shock Trauma Research Institute, Loyola University Chicago, Health Sciences Division, Maywood, IL
| | - Richard L Gamelli
- Department of Surgery, Loyola University Chicago, Health Sciences Division, Maywood, IL; Burn and Shock Trauma Research Institute, Loyola University Chicago, Health Sciences Division, Maywood, IL
| | - Kuzhali Muthumalaiappan
- Department of Surgery, Loyola University Chicago, Health Sciences Division, Maywood, IL; Burn and Shock Trauma Research Institute, Loyola University Chicago, Health Sciences Division, Maywood, IL.
| |
Collapse
|
73
|
Hernández CC, Burgos CF, Gajardo AH, Silva-Grecchi T, Gavilan J, Toledo JR, Fuentealba J. Neuroprotective effects of erythropoietin on neurodegenerative and ischemic brain diseases: the role of erythropoietin receptor. Neural Regen Res 2017; 12:1381-1389. [PMID: 29089974 PMCID: PMC5649449 DOI: 10.4103/1673-5374.215240] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/26/2017] [Indexed: 12/11/2022] Open
Abstract
Erythropoietin (Epo) is a fundamental hormone in the regulation of hematopoiesis, and other secondary roles mediated by the binding of the hormone to its specific receptor (EpoR), which leads to an activation of key signaling pathways that induce an increase in cell differentiation, apoptosis control and neuroprotection. It has been suggested that their function depends on final conformation of glycosylations, related with affinity to the receptor and its half-life. The presence of EpoR has been reported in different tissues including central nervous system, where it has been demonstrated to exert a neuroprotective function against oxidative stress conditions, such as ischemic injury and neurodegenerative diseases. There is also evidence of an increase in EpoR expression in brain cell lysates of Alzheimer's patients with respect to healthy patients. These results are related with extensive in vitro experimental data of neuroprotection obtained from cell lines, primary cell cultures and hippocampal slices. Additionally, this data is correlated with in vivo experiments (water maze test) in mouse models of Alzheimer's disease where Epo treatment improved cognitive function. These studies support the idea that receptor activation induces a neuroprotective effect in neurodegenerative disorders including dementias, and especially Alzheimer's disease. Taken together, available evidence suggests that Epo appears to be a central element for EpoR activation and neuroprotective properties in the central nervous system. In this review, we will describe the mechanisms associated with neuroprotection and its relation with the activation of EpoR in order with identify new targets to develop pharmacological strategies.
Collapse
Affiliation(s)
- Carolina Castillo Hernández
- Laboratory of Screening of Neuroactive Compounds, Department of Physiology, School of Biological Sciences, University of Concepción, Concepción, Chile
- Laboratory of Biotechnology and Biopharmaceutical, Department of Pathophysiology, School of Biological Sciences, University of Concepción, Concepción, Chile
| | - Carlos Felipe Burgos
- Laboratory of Screening of Neuroactive Compounds, Department of Physiology, School of Biological Sciences, University of Concepción, Concepción, Chile
| | - Angela Hidalgo Gajardo
- Laboratory of Biotechnology and Biopharmaceutical, Department of Pathophysiology, School of Biological Sciences, University of Concepción, Concepción, Chile
| | - Tiare Silva-Grecchi
- Laboratory of Screening of Neuroactive Compounds, Department of Physiology, School of Biological Sciences, University of Concepción, Concepción, Chile
| | - Javiera Gavilan
- Laboratory of Screening of Neuroactive Compounds, Department of Physiology, School of Biological Sciences, University of Concepción, Concepción, Chile
| | - Jorge Roberto Toledo
- Laboratory of Biotechnology and Biopharmaceutical, Department of Pathophysiology, School of Biological Sciences, University of Concepción, Concepción, Chile
| | - Jorge Fuentealba
- Laboratory of Screening of Neuroactive Compounds, Department of Physiology, School of Biological Sciences, University of Concepción, Concepción, Chile
| |
Collapse
|
74
|
Erythropoietin either Prevents or Exacerbates Retinal Damage from Eye Trauma Depending on Treatment Timing. Optom Vis Sci 2017; 94:20-32. [PMID: 27281679 DOI: 10.1097/opx.0000000000000898] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
PURPOSE Erythropoietin (EPO) is a promising neuroprotective agent and is currently in Phase III clinical trials for the treatment of traumatic brain injury. The goal of this study was to determine if EPO is also protective in traumatic eye injury. METHODS The left eyes of anesthetized DBA/2J or Balb/c mice were exposed to a single 26 psi overpressure air-wave while the rest of the body was shielded. DBA/2J mice were given intraperitoneal injections of EPO or buffer and analyses were performed at 3 or 7 days post-blast. Balb/c mice were given intramuscular injections of rAAV.EpoR76E or rAAV.eGFP either pre- or post-blast and analyses were performed at 1 month post-blast. RESULTS EPO had a bimodal effect on cell death, glial reactivity, and oxidative stress. All measures were increased at 3 days post-blast and decreased at 7-days post-blast. Increased retinal ferritin and NADPH oxygenases were detected in retinas from EPO-treated mice. The gene therapy approach protected against axon degeneration, cell death, and oxidative stress when given after blast, but not before. CONCLUSIONS Systemic, exogenous EPO and EPO-R76E protects the retina after trauma even when initiation of treatment is delayed by up to 3 weeks. Systemic treatment with EPO or EPO-R76E beginning before or soon after trauma may exacerbate protective effects of EPO within the retina as a result of increased iron levels from erythropoiesis and, thus, increased oxidative stress within the retina. This is likely overcome with time as a result of an increase in levels of antioxidant enzymes. Either intraocular delivery of EPO or treatment with non-erythropoietic forms of EPO may be more efficacious.
Collapse
|
75
|
He L, Wu S, Hao Q, Dioum EM, Zhang K, Zhang C, Li W, Zhang W, Zhang Y, Zhou J, Pang Z, Zhao L, Ma X, Li M, Zhang Q. Local blockage of self-sustainable erythropoietin signaling suppresses tumor progression in non-small cell lung cancer. Oncotarget 2017; 8:82352-82365. [PMID: 29137269 PMCID: PMC5669895 DOI: 10.18632/oncotarget.19354] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 06/30/2017] [Indexed: 12/18/2022] Open
Abstract
Functional significance of co-expressed erythropoietin (EPO) and its receptor (EPOR) in non-small cell lung cancer (NSCLC) had been under debate. In this study, co-overexpression of EPO/EPOR was confirmed to be positively associated with poor survival in NSCLC. The serum EPO in 14 of 35 enrolled NSCLC patients were found elevated significantly and decreased to normal level after tumor resection. With primary tumor cell culture and patient-derived tumor xenograft (PDX) mouse model, the EPO secretion from the tumors of these 14 patients was verified. Then, we proved the patient derived serum EPO was functionally active and had growth promotion effect in EPO/EPOR overexpressed but not in EPO/EPOR under-expressed NSCLC cells. We also illustrated EPO promoted NSCLC cell proliferation through an EPOR/Jak2/Stat5a/cyclinD1 pathway. In xenograft mouse model, we proved local application of EPO neutralizing antibody and short hairpin RNA (shRNA) against EPOR effectively inhibited the growth of EPO/EPOR overexpressed NSCLC cells and prolonged survivals of the mice. Finally, EPO/EPOR/Jak2/Stat5a/cyclinD1 signaling was found to be a mediator of hypoxia induced growth in EPO/EPOR overexpressed NSCLC. Our results illustrated a subgroup of NSCLC adapt to hypoxia through self-sustainable EPO/EPOR signaling and suggest local blockage of EPO/EPOR as potential therapeutic method in this distinct NSCLC population.
Collapse
Affiliation(s)
- Lei He
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Shouzhen Wu
- Shaanxi Institute of Pediatric Diseases, Xi'an Children's Hospital, Xi'an, China
| | - Qiang Hao
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Elhadji M Dioum
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Current/Present address: Diabetes Department, Nestle Institute of Health Science, EPFL Campus, Lausanne, Switzerland
| | - Kuo Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Cun Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Weina Li
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Wei Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Yingqi Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Jiming Zhou
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Zhijun Pang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Lijuan Zhao
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Xiaowen Ma
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Meng Li
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, China
| | - Qiuyang Zhang
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Current/Present address: Center for Esophageal Research, Baylor University Medical Center, Dallas, Texas, USA
| |
Collapse
|
76
|
Eskandani M, Vandghanooni S, Barar J, Nazemiyeh H, Omidi Y. Cell physiology regulation by hypoxia inducible factor-1: Targeting oxygen-related nanomachineries of hypoxic cells. Int J Biol Macromol 2017; 99:46-62. [DOI: 10.1016/j.ijbiomac.2016.10.113] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 10/26/2016] [Indexed: 12/27/2022]
|
77
|
Wilkin T, Baoutina A, Hamilton N. Equine performance genes and the future of doping in horseracing. Drug Test Anal 2017; 9:1456-1471. [DOI: 10.1002/dta.2198] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 03/23/2017] [Accepted: 03/24/2017] [Indexed: 01/20/2023]
Affiliation(s)
- Tessa Wilkin
- Vet Faculty; University of Sydney; Gunn Building, Sydney University, Camperdown NSW Australia
- Bioanalysis; The National Measurement Institute; 36 Bradfield Rd, Lindfield Sydney New South Wales Australia
| | - Anna Baoutina
- School of Life and Environmental Sciences, Faculty of Science; The University of Sydney; Bradfield Rd West Lindfield New South Wales Australia
| | - Natasha Hamilton
- Faculty of Veterinary Science; University of Sydney; Sydney New South Wales Australia
| |
Collapse
|
78
|
Kostakis ID, Zavras N, Damaskos C, Sakellariou S, Korkolopoulou P, Misiakos EP, Tsaparas P, Vaos G, Karatzas T. Erythropoietin and sildenafil protect against ischemia/reperfusion injury following testicular torsion in adult rats. Exp Ther Med 2017; 13:3341-3347. [PMID: 28587411 PMCID: PMC5450555 DOI: 10.3892/etm.2017.4441] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 09/22/2016] [Indexed: 11/06/2022] Open
Abstract
Testicular torsion/detorsion causes severe tissue damage due to ischemia/reperfusion injury. The present study investigated the protective effect of erythropoietin and sildenafil against ischemia/reperfusion injury following unilateral testicular torsion/detorsion in adult rats. A total of 28 adult male rats were included, and were divided into the following groups: Group A (n=5), sham operated; groups B (n=5), C (n=5), D (n=5) and E (n=8), undergoing right testis torsion and detorsion after 90 min. Group B received no drug treatment. Rats in the groups C and D received low-dose (1,000 IU/kg) or high-dose (3,000 IU/kg) erythropoietin, while those in group E received sildenafil (0.7 mg/kg), through intraperitoneal injection after 60 min of torsion. The right testis was extracted 24 h after detorsion, and the tissue was subjected to histopathological examination and immunohistochemical assessment of cleaved caspase-3 expression. Histological alterations and the quality of spermatogenesis were scored according to the Cosentino and the Johnsen scoring systems, respectively. The results demonstrated normal testicular architecture in group A, while the other groups showed ischemic cellular damages, with the worst scores observed in group B. Groups D and E presented better scores compared with group C. Regarding the quality of spermatogenesis, the best scores were observed in group A, and the worst in group B. Groups C, D and E presented similar results, which were improved in comparison to group B, however, not compared to group A. Furthermore, cleaved caspase-3 levels were lower in groups A, D and E, with equal results observed. Group C had higher levels of cleaved caspase-3 compared with these groups, but lower than group B, which presented the highest cleaved caspase-3 levels. In conclusion, erythropoietin and sildenafil protect testis from ischemia/reperfusion injury by decreasing cellular damage and attenuating apoptosis.
Collapse
Affiliation(s)
- Ioannis D Kostakis
- Laboratory of Experimental Surgery and Surgical Research 'N.S. Christeas', National and Kapodistrian University of Athens, Medical School, 11527 Athens, Greece.,Second Department of Propedeutic Surgery, 'Laiko' General Hospital, National and Kapodistrian University of Athens, Medical School, 11527 Athens, Greece
| | - Nick Zavras
- Third Department of Surgery, Attikon University Hospital, National and Kapodistrian University of Athens, Medical School, 12462 Athens, Greece
| | - Christos Damaskos
- Laboratory of Experimental Surgery and Surgical Research 'N.S. Christeas', National and Kapodistrian University of Athens, Medical School, 11527 Athens, Greece.,Second Department of Propedeutic Surgery, 'Laiko' General Hospital, National and Kapodistrian University of Athens, Medical School, 11527 Athens, Greece
| | - Stratigoula Sakellariou
- First Department of Pathology, National and Kapodistrian University of Athens, Medical School, 11527 Athens, Greece
| | - Penelope Korkolopoulou
- First Department of Pathology, National and Kapodistrian University of Athens, Medical School, 11527 Athens, Greece
| | - Evangelos P Misiakos
- Third Department of Surgery, Attikon University Hospital, National and Kapodistrian University of Athens, Medical School, 12462 Athens, Greece
| | - Petros Tsaparas
- Laboratory of Experimental Surgery and Surgical Research 'N.S. Christeas', National and Kapodistrian University of Athens, Medical School, 11527 Athens, Greece.,Second Department of Propedeutic Surgery, 'Laiko' General Hospital, National and Kapodistrian University of Athens, Medical School, 11527 Athens, Greece
| | - George Vaos
- Department of Pediatric Surgery, Alexandroupolis University Hospital, Democritus University of Thrace, Medical School, 68100 Alexandroupolis, Greece
| | - Theodoros Karatzas
- Laboratory of Experimental Surgery and Surgical Research 'N.S. Christeas', National and Kapodistrian University of Athens, Medical School, 11527 Athens, Greece.,Second Department of Propedeutic Surgery, 'Laiko' General Hospital, National and Kapodistrian University of Athens, Medical School, 11527 Athens, Greece
| |
Collapse
|
79
|
Kalemci O, Aydin HE, Kizmazoglu C, Kaya I, Yılmaz H, Arda NM. Effects of Quercetin and Mannitol on Erythropoietin Levels in Rats Following Acute Severe Traumatic Brain Injury. J Korean Neurosurg Soc 2017; 60:355-361. [PMID: 28490163 PMCID: PMC5426445 DOI: 10.3340/jkns.2016.0505.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 10/27/2016] [Accepted: 11/29/2016] [Indexed: 12/31/2022] Open
Abstract
Objective The aim of this study to investigate the normal values of erythropoietin (EPO) and neuroprotective effects of quercetin and mannitol on EPO and hematocrit levels after acute severe traumatic brain injury (TBI) in rat model. Methods A weight-drop impact acceleration model of TBI was used on 40 male Wistar rats. The animals were divided into sham (group I), TBI (group II), TBI+quercetin (50 mg/kg intravenously) (group III), and TBI+mannitol (1 mg/kg intravenously) (group IV) groups. The malondialdehyde, glutathione peroxidase, catalase, EPO, and hematocrit levels were measured 1 and 4 hour after injury. Two-way repeated measures analysis of variance and Tukey’s test were used for statistical analysis. Results The malondialdehyde levels decreased significantly after administration of quercetin and mannitol compared with those in group II. Catalase and glutathione peroxidase levels increased significantly in groups III and IV. Serum EPO levels decreased significantly after mannitol but not after quercetin administration. Serum hematocrit levels did not change significantly after quercetin and mannitol administration 1 hour after trauma. However, mannitol administration decreased serum hematocrit levels significantly after 4 hour. Conclusion This study suggests that quercetin may be a good alternative treatment for TBI, as it did not decrease the EPO levels.
Collapse
Affiliation(s)
- Orhan Kalemci
- Department of Neurosurgery, School of Medicine and Hospital, Dokuz Eylul University, Izmir, Turkey
| | - Hasan Emre Aydin
- Department of Pharmacology, Eskisehir Osmangazi University, Eskisehir, Turkey.,Department of Neurosurgery, School of Medicine and Hospital, Dumlupınar University, Kutahya, Turkey
| | - Ceren Kizmazoglu
- Department of Neurosurgery, School of Medicine and Hospital, Dokuz Eylul University, Izmir, Turkey
| | - Ismail Kaya
- Department of Neurosurgery, Kilis State Hospital, Kilis, Turkey
| | - Hulya Yılmaz
- Department of Biostatistics and Medical Informatics, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Nuri M Arda
- Department of Neurosurgery, School of Medicine and Hospital, Dokuz Eylul University, Izmir, Turkey
| |
Collapse
|
80
|
Abstract
Erythropoiesis is tightly regulated by the growth factor erythropoietin (Epo). Signal activation begins when Epo engages its cognate receptor, Epo-R, triggering receptor homodimerization, and recruitment of signaling intermediates including Jak2 that phosphorylates both the receptor cytoplasmic tail and downstream effectors including the transcription factor, STAT5. Transcription factors subsequently activate transcription of prosurvival and prodifferentiation genes responsible for red blood cell production. The fidelity of Epo-R signaling is dependent upon residence within detergent insoluble membrane lipid raft fractions. Lipid rafts are membrane microdomains that serve as signaling scaffolds composed of densely packed sphingolipids and cholesterol where receptors and intermediate signaling proteins are recruited and interact to execute stimuli. Disruption of lipid rafts is detrimental to Epo signaling, a phenomenon that may be utilized to design novel therapeutics for conditions in which Epo signaling is deficient. Here, we review the Epo signaling cascade, particularly, as it relates to localization and dependence on lipid rafts, and discuss considerations for novel therapeutic design.
Collapse
Affiliation(s)
- Kathy McGraw
- Division of Clinical Sciences, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States.
| | - Alan List
- Division of Clinical Sciences, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| |
Collapse
|
81
|
Nematbakhsh M, Pezeshki Z, Eshraghi Jazi F, Mazaheri B, Moeini M, Safari T, Azarkish F, Moslemi F, Maleki M, Rezaei A, Saberi S, Dehghani A, Malek M, Mansouri A, Ghasemi M, Zeinali F, Zamani Z, Navidi M, Jilanchi S, Shirdavani S, Ashrafi F. Cisplatin-Induced Nephrotoxicity; Protective Supplements and Gender Differences. Asian Pac J Cancer Prev 2017; 18:295-314. [PMID: 28345324 PMCID: PMC5454720 DOI: 10.22034/apjcp.2017.18.2.295] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cisplatin (CDDP) has been widely used as a chemotherapeutic agent for solid tumors. The most common side effect of CDDP is nephrotoxicity, and many efforts have been made in the laboratory and the clinic to employ candidate adjuvants to CDDP to minimize this adverse influence. Many synthetic and herbal antioxidants as well as trace elements have been investigated for this purpose in recent years and a variety of positive and negative results have been yielded. However, no definitive supplement has so far been proposed to prevent CDDP-induced nephrotoxicity; however, this condition is gender related and the sex hormone estrogen may protect the kidney against CDDP damage. In this review, the results of research related to the effect of different synthetic and herbal antioxidants supplements are presented and discussed with suggestions included for future work.
Collapse
Affiliation(s)
- Mehdi Nematbakhsh
- Water and Electrolytes Research Center, Department of Physiology, Isfahan University of Medical Sciences, Isfahan, Iran. *
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Yu YB, Su KH, Kou YR, Guo BC, Lee KI, Wei J, Lee TS. Role of transient receptor potential vanilloid 1 in regulating erythropoietin-induced activation of endothelial nitric oxide synthase. Acta Physiol (Oxf) 2017; 219:465-477. [PMID: 27232578 DOI: 10.1111/apha.12723] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 05/25/2016] [Accepted: 05/25/2016] [Indexed: 01/02/2023]
Abstract
AIMS Erythropoietin (EPO), the key hormone involved in erythropoiesis, beneficially affects endothelial cells (ECs), but the detailed mechanisms are yet to be completely understood. In this study, we investigated the role of transient receptor potential vanilloid type 1 (TRPV1), a ligand-gated non-selective calcium (Ca2+ ) channel, in EPO-mediated endothelial nitric oxide synthase (eNOS) activation and angiogenesis. METHODS AND RESULTS In ECs, EPO time dependently increased intracellular levels of calcium; this increase was abrogated by the Ca2+ chelators and pharmacological inhibitors of TRPV1 in bovine aortic ECs (BAECs) and TRPV1-transfected HEK293 cells. In addition, EPO-induced nitrite oxide (NO) production, phosphorylation of eNOS, Akt and AMP-activated protein kinase (AMPK) and the formation of TRPV1-Akt-AMPK-eNOS complex as well as tube formation were diminished by the pharmacological inhibition of TRPV1 in BAECs. Moreover, EPO time dependently induced the phosphorylation of phospholipase C-γ1 (PLC-γ1). Inhibition of PLC-γ1 activity blunted the EPO-induced Ca2+ influx, eNOS phosphorylation, TRPV1-eNOS complex formation and NO production. The phosphorylated level of eNOS increased in the aortas of EPO-treated wild-type (WT) mice or EPO-transgenic (Tg) mice but not in those of EPO-treated TRPV1-deficient (TRPV1-/- ) mice or EPO-Tg/TRPV1-/- mice. Matrigel plug assay showed that EPO-induced angiogenesis was abrogated in TRPV1 antagonist capsazepine-treated WT mice and TRPV1-/- mice. CONCLUSION These findings indicate the EPO-induced Ca2+ influx via the activation of the PLC-γ1 signalling pathway, which leads to TRPV1 activation and consequently increases the association of the TRPV1-Akt-AMPK-eNOS complex, eNOS activation, NO production and angiogenesis.
Collapse
Affiliation(s)
- Y.-B. Yu
- Institute of Physiology; National Yang-Ming University; Taipei Taiwan
- Division of Hematology; Department of Medicine; Taipei Veterans General Hospital; Taipei Taiwan
| | - K.-H. Su
- Institute of Physiology; National Yang-Ming University; Taipei Taiwan
- The Jackson Laboratory; Bar Harbor ME USA
| | - Y. R. Kou
- Institute of Physiology; National Yang-Ming University; Taipei Taiwan
| | - B.-C. Guo
- Institute of Physiology; National Yang-Ming University; Taipei Taiwan
| | - K.-I. Lee
- Institute of Physiology; National Yang-Ming University; Taipei Taiwan
| | - J. Wei
- Heart Center; Cheng-Hsin General Hospital; Taipei Taiwan
| | - T.-S. Lee
- Institute of Physiology; National Yang-Ming University; Taipei Taiwan
- Genome Research Center; National Yang-Ming University; Taipei Taiwan
- Aging and Health Research Center; National Yang-Ming University; Taipei Taiwan
| |
Collapse
|
83
|
Iturri P, Bairam A, Soliz J. Efficient breathing at neonatal ages: A sex and Epo-dependent issue. Respir Physiol Neurobiol 2016; 245:89-97. [PMID: 28041993 DOI: 10.1016/j.resp.2016.12.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Revised: 12/02/2016] [Accepted: 12/05/2016] [Indexed: 11/29/2022]
Abstract
During postnatal life, the respiratory control system undergoes intense development and is highly responsive to stimuli emerging from the environment. In fact, interruption of breathing prevents gas exchange and results in systemic hypoxia that, if prolonged, can lead to cardio-respiratory failure or sudden infant death. Moreover, in newborns and infants, respiratory disorders related to neural control dysfunction show significant sexual dimorphism with a higher prevalence in males. To this day, the therapeutic tools available to alleviate these respiratory disorders remain limited. Furthermore, the factors explaining the sexual dimorphism in newborns and during infancy remain unknown. Erythropoietin (Epo) was originally discovered as a cytokine able to increase the production of red blood cells upon conditions of reduced oxygen availability. We now know that Epo is a cytokine also secreted by neurons and astrocytes that protects the brain during trauma or hypoxic stress in a sex dependent manner. In this novel line of research, our previous studies demonstrated at adult ages that cerebral Epo acts as a respiratory stimulant in rodents and humans. These results provided a strong rationale for exploring the role of cerebral Epo in neuronal respiratory control during postnatal development. The objective of this review is to summarize our recent findings showing that cerebral Epo is a potent sex-specific respiratory stimulant at neonatal ages. Keeping in mind that Epo is routinely and safely administrated in newborn humans for anemia and neonatal asphyxia, we predict that our research provides the basis necessary to promote the clinical use of Epo against neonatal respiratory disorders related to neural control dysfunction.
Collapse
Affiliation(s)
- Pablo Iturri
- Centre de Recherche du CHU de Québec, Pavillon St François d'Assise, Département de Pédiatrie, Faculté de Médecine, Université Laval, Québec, QC, Canada; Molecular Biology and Biotechnology Institute, Universidad Mayor de San Andres, La Paz, Bolivia
| | - Aida Bairam
- Centre de Recherche du CHU de Québec, Pavillon St François d'Assise, Département de Pédiatrie, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Jorge Soliz
- Centre de Recherche du CHU de Québec, Pavillon St François d'Assise, Département de Pédiatrie, Faculté de Médecine, Université Laval, Québec, QC, Canada; Molecular Biology and Biotechnology Institute, Universidad Mayor de San Andres, La Paz, Bolivia.
| |
Collapse
|
84
|
Kaufner L, von Heymann C, Henkelmann A, Pace NL, Weibel S, Kranke P, Meerpohl JJ, Gill R. Erythropoietin with iron supplementation for preoperative anaemia in non‐cardiac surgery. Cochrane Database Syst Rev 2016; 2016:CD012451. [PMCID: PMC6463842 DOI: 10.1002/14651858.cd012451] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2024]
Abstract
This is a protocol for a Cochrane Review (Intervention). The objectives are as follows: We aim to evaluate the efficacy and safety of preoperative erythropoietin (rHuEPO) with supplemental iron (parenteral or oral) in reducing perioperative allogeneic red blood cell transfusions in preoperatively anaemic people undergoing non‐cardiac surgery.
Collapse
Affiliation(s)
- Lutz Kaufner
- Charité ‐ University Medicine BerlinDepartment of Anaesthesiology and Intensive Care MedicineAugustenburger Platz 1BerlinGermany13353
| | - Christian von Heymann
- Vivantes Klinikum im FriedrichshainDepartment of Anaesthesiology, Intensive Care Medicine, Emergency Care Medicine and Pain TherapyLandsberger Allee 49BerlinGermany10249
| | - Anne Henkelmann
- Charité ‐ University Medicine BerlinDepartment of Anaesthesiology and Intensive Care MedicineAugustenburger Platz 1BerlinGermany13353
| | - Nathan Leon Pace
- University of UtahDepartment of Anesthesiology3C444 SOM30 North 1900 EastSalt Lake CityUSA84132‐2304
| | - Stephanie Weibel
- University of WürzburgDepartment of Anaesthesia and Critical CareOberduerrbacher Str. 6WürzburgGermany
| | - Peter Kranke
- University of WürzburgDepartment of Anaesthesia and Critical CareOberduerrbacher Str. 6WürzburgGermany
| | - Joerg J Meerpohl
- Medical Center ‐ University of FreiburgCochrane GermanyBreisacher Straße 153FreiburgGermany79110
| | - Ravi Gill
- Southampton University Hospital NHS TrustDepartment of AnaestheticsTremona RoadSouthamptonUKSO16 6YD
| |
Collapse
|
85
|
Tu X, Muhammad P, Liu J, Ma Y, Wang S, Yin D, Liu Z. Molecularly Imprinted Polymer-Based Plasmonic Immunosandwich Assay for Fast and Ultrasensitive Determination of Trace Glycoproteins in Complex Samples. Anal Chem 2016; 88:12363-12370. [DOI: 10.1021/acs.analchem.6b03597] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Xueying Tu
- State Key Laboratory of Analytical
Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| | - Pir Muhammad
- State Key Laboratory of Analytical
Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| | - Jia Liu
- State Key Laboratory of Analytical
Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| | - Yanyan Ma
- State Key Laboratory of Analytical
Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| | - Shuangshou Wang
- State Key Laboratory of Analytical
Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| | - Danyang Yin
- State Key Laboratory of Analytical
Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| | - Zhen Liu
- State Key Laboratory of Analytical
Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| |
Collapse
|
86
|
Daimon S, Nuka H, Kitada K, Suzuki Y, Kim F, Kawano M. Influence of continuous erythropoietin receptor activator (CERA) administration intervals on erythropoietic effect in hemodialysis patients. RENAL REPLACEMENT THERAPY 2016. [DOI: 10.1186/s41100-016-0071-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
87
|
Erythropoietin and thrombopoietin mimetics: Natural alternatives to erythrocyte and platelet disorders. Crit Rev Oncol Hematol 2016; 108:175-186. [DOI: 10.1016/j.critrevonc.2016.11.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 10/27/2016] [Accepted: 11/02/2016] [Indexed: 12/21/2022] Open
|
88
|
Abstract
The anoxemia theory proposes that an imbalance between the demand for and supply of oxygen in the arterial wall is a key factor in the development of atherosclerosis. There is now substantial evidence that there are regions within the atherosclerotic plaque in which profound hypoxia exists; this may fundamentally change the function, metabolism, and responses of many of the cell types found within the developing plaque and whether the plaque will evolve into a stable or unstable phenotype. Hypoxia is characterized in molecular terms by the stabilization of hypoxia-inducible factor (HIF) 1α, a subunit of the heterodimeric nuclear transcriptional factor HIF-1 and a master regulator of oxygen homeostasis. The expression of HIF-1 is localized to perivascular tissues, inflammatory macrophages, and smooth muscle cells adjacent to the necrotic core of atherosclerotic lesions and regulates several genes that are important to vascular function including vascular endothelial growth factor, nitric oxide synthase, endothelin-1, and erythropoietin. This review summarizes the effects of hypoxia on the functions of cells involved in atherogenesis and the evidence for its potential importance from experimental models and clinical studies.
Collapse
Affiliation(s)
- Gordon A A Ferns
- 1 Department of Medical Education, Brighton & Sussex Medical School, Brighton, United Kingdom
| | - Lamia Heikal
- 1 Department of Medical Education, Brighton & Sussex Medical School, Brighton, United Kingdom
| |
Collapse
|
89
|
Understanding the biosimilar approval and extrapolation process—A case study of an epoetin biosimilar. Crit Rev Oncol Hematol 2016; 104:98-107. [DOI: 10.1016/j.critrevonc.2016.04.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 04/01/2016] [Accepted: 04/27/2016] [Indexed: 11/20/2022] Open
|
90
|
Lamon S, Zacharewicz E, Arentson-Lantz E, Gatta PAD, Ghobrial L, Gerlinger-Romero F, Garnham A, Paddon-Jones D, Russell AP. Erythropoietin Does Not Enhance Skeletal Muscle Protein Synthesis Following Exercise in Young and Older Adults. Front Physiol 2016; 7:292. [PMID: 27458387 PMCID: PMC4937030 DOI: 10.3389/fphys.2016.00292] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 06/27/2016] [Indexed: 01/07/2023] Open
Abstract
Purpose: Erythropoietin (EPO) is a renal cytokine that is primarily involved in hematopoiesis while also playing a role in non-hematopoietic tissues expressing the EPO-receptor (EPOR). The EPOR is present in human skeletal muscle. In mouse skeletal muscle, EPO stimulation can activate the AKT serine/threonine kinase 1 (AKT) signaling pathway, the main positive regulator of muscle protein synthesis. We hypothesized that a single intravenous EPO injection combined with acute resistance exercise would have a synergistic effect on skeletal muscle protein synthesis via activation of the AKT pathway. Methods: Ten young (24.2 ± 0.9 years) and 10 older (66.6 ± 1.1 years) healthy subjects received a primed, constant infusion of [ring-13C6] L-phenylalanine and a single injection of 10,000 IU epoetin-beta or placebo in a double-blind randomized, cross-over design. 2 h after the injection, the subjects completed an acute bout of leg extension resistance exercise to stimulate skeletal muscle protein synthesis. Results: Significant interaction effects in the phosphorylation levels of the members of the AKT signaling pathway indicated a differential activation of protein synthesis signaling in older subjects when compared to young subjects. However, EPO offered no synergistic effect on vastus lateralis mixed muscle protein synthesis rate in young or older subjects. Conclusions: Despite its ability to activate the AKT pathway in skeletal muscle, an acute EPO injection had no additive or synergistic effect on the exercise-induced activation of muscle protein synthesis or muscle protein synthesis signaling pathways.
Collapse
Affiliation(s)
- Séverine Lamon
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University Geelong, VIC, Australia
| | - Evelyn Zacharewicz
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University Geelong, VIC, Australia
| | - Emily Arentson-Lantz
- Department of Nutrition and Metabolism, University of Texas Medical Branch Galveston, TX, USA
| | - Paul A Della Gatta
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University Geelong, VIC, Australia
| | - Lobna Ghobrial
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University Geelong, VIC, Australia
| | - Frederico Gerlinger-Romero
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University Geelong, VIC, Australia
| | - Andrew Garnham
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University Geelong, VIC, Australia
| | - Douglas Paddon-Jones
- Department of Nutrition and Metabolism, University of Texas Medical Branch Galveston, TX, USA
| | - Aaron P Russell
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University Geelong, VIC, Australia
| |
Collapse
|
91
|
Kim SY, Shin DY, Kim SM, Lee M, Kim EJ. Aberrant DNA methylation-induced gene inactivation is associated with the diagnosis and/or therapy of T-cell leukemias. Leuk Res 2016; 47:116-22. [PMID: 27318093 DOI: 10.1016/j.leukres.2016.05.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 05/13/2016] [Accepted: 05/26/2016] [Indexed: 12/31/2022]
Abstract
Aberrant hypermethylation of tumor suppressor genes is known to play an important role in the development of many tumors, and aberrant DNA hypermethylation was recently identified in hematologic malignancies, where it is thought to hold relevance in leukemogenesis. Here, we report that there are differences in the DNA methylation patterns seen in normal peripheral blood and two T-cell leukemia cell lines. We identify nine genes (CLEC4E, CR1, DBC1, EPO, HAL-DOA, IGF2, IL12B, ITGA1, and LMX1B) that are significantly hypermethylated in T-cell leukemias cell lines, and suggest that aberrant hypermethylation of these normally unmethylated genes may induce their transcriptional and expressional silencing. Furthermore, we observed that the expression levels of DNMT1 and DNMT3a were significantly decreased by 5-aza-2'-deoxycytidine (5-Aza-dC), which is a demethylation agent known to deplete DNA methyltransferases (DNMTs) in leukemia cancer cells and restore the expression levels of their target genes in Jurkat cells. This result suggests that the overexpression of DNMTs could contribute to the development of T-cell leukemias by inducing hypermethylation of the target genes. Together, our results show that aberrant hypermethylation is an important molecular mechanism in the progression of T-cell leukemias, and thus could prove useful as a prognostic and/or diagnostic marker. Moreover, 5-Aza-dC might be a promising candidate for the treatment of T-cell leukemia.
Collapse
Affiliation(s)
- Sun Young Kim
- Division of Radiation Effect, Korea Institute of Radiological & Medical Sciences, Seoul 01812, Korea
| | - Dong-Yeop Shin
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Korea
| | - Sang-Man Kim
- Health Services Management, KH School of Management, Kyung Hee University, Seoul 02453, Korea
| | - Minyoung Lee
- Division of Radiation Effect, Korea Institute of Radiological & Medical Sciences, Seoul 01812, Korea.
| | - Eun Ju Kim
- Division of Radiation Effect, Korea Institute of Radiological & Medical Sciences, Seoul 01812, Korea; Health Services Management, KH School of Management, Kyung Hee University, Seoul 02453, Korea.
| |
Collapse
|
92
|
Bianchi S, Fusi J, Franzoni F, Giovannini L, Galetta F, Mannari C, Guidotti E, Tocchini L, Santoro G. "Effects of recombinant human erythropoietin high mimicking abuse doses on oxidative stress processes in rats". Biomed Pharmacother 2016; 82:355-63. [PMID: 27470373 DOI: 10.1016/j.biopha.2016.05.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 05/12/2016] [Accepted: 05/13/2016] [Indexed: 12/21/2022] Open
Abstract
Although many studies highlight how long-term moderate dose of Recombinant Human Erythropoietin (rHuEPO) treatments result in beneficial and antioxidants effects, few studies take into account the effects that short-term high doses of rHuEPO (mimicking abuse conditions) might have on the oxidative stress processes. Thus, the aim of this study was to investigate the in vivo antioxidant activity of rHuEPO, administered for a short time and at high doses to mimic its sports abuse as doping. Male Wistar healthy rats (n=36) were recruited for the study and were treated with three different concentrations of rHuEPO: 7.5, 15, 30μg/kg. Plasma concentrations of erythropoietin, 8-epi Prostaglandin F2α, plasma and urinary concentrations of NOx were evaluated with specific assay kit, while hematocrit levels were analyzed with an automated cell counter. Antioxidant activity of rHuEPO was assessed analyzing the possible variation of the plasma scavenger capacity against hydroxylic and peroxylic radicals by TOSC (Total Oxyradical Scavenging Capacity) assay. Statistical analyses showed higher hematocrit values, confirmed by a statistically significant increase of plasmatic EPO concentration. An increase in plasma scavenging capacity against peroxyl and hydroxyl radicals, in 8-isoprostane plasmatic concentrations and in plasmatic and urinary levels of NOX were also found in all the treated animals, though not always statistically significant. Our results confirm the literature data regarding the antioxidant action of erythropoietin administered at low doses and for short times, whereas they showed an opposite incremental oxidative stress action when erythropoietin is administered at high doses.
Collapse
Affiliation(s)
- Sara Bianchi
- Department of Translational Research and New Technologies in Medicine and Surgery, Pharmacology, Medical School, University of Pisa, Via Roma 55, 56126, Pisa, Italy.
| | - Jonathan Fusi
- Department of Clinical and Experimental Medicine, University of Pisa, Via Savi 10, 56126, Pisa, Italy
| | - Ferdinando Franzoni
- Department of Clinical and Experimental Medicine, University of Pisa, Via Savi 10, 56126, Pisa, Italy
| | - Luca Giovannini
- Department of Translational Research and New Technologies in Medicine and Surgery, Pharmacology, Medical School, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Fabio Galetta
- Department of Clinical and Experimental Medicine, University of Pisa, Via Savi 10, 56126, Pisa, Italy
| | - Claudio Mannari
- Department of Translational Research and New Technologies in Medicine and Surgery, Pharmacology, Medical School, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Emanuele Guidotti
- Department of Clinical and Experimental Medicine, University of Pisa, Via Savi 10, 56126, Pisa, Italy
| | - Leonardo Tocchini
- Department of Clinical and Experimental Medicine, University of Pisa, Via Savi 10, 56126, Pisa, Italy
| | - Gino Santoro
- Department of Clinical and Experimental Medicine, University of Pisa, Via Savi 10, 56126, Pisa, Italy
| |
Collapse
|
93
|
Manukhina EB, Downey HF, Shi X, Mallet RT. Intermittent hypoxia training protects cerebrovascular function in Alzheimer's disease. Exp Biol Med (Maywood) 2016; 241:1351-63. [PMID: 27190276 DOI: 10.1177/1535370216649060] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is a leading cause of death and disability among older adults. Modifiable vascular risk factors for AD (VRF) include obesity, hypertension, type 2 diabetes mellitus, sleep apnea, and metabolic syndrome. Here, interactions between cerebrovascular function and development of AD are reviewed, as are interventions to improve cerebral blood flow and reduce VRF. Atherosclerosis and small vessel cerebral disease impair metabolic regulation of cerebral blood flow and, along with microvascular rarefaction and altered trans-capillary exchange, create conditions favoring AD development. Although currently there are no definitive therapies for treatment or prevention of AD, reduction of VRFs lowers the risk for cognitive decline. There is increasing evidence that brief repeated exposures to moderate hypoxia, i.e. intermittent hypoxic training (IHT), improve cerebral vascular function and reduce VRFs including systemic hypertension, cardiac arrhythmias, and mental stress. In experimental AD, IHT nearly prevented endothelial dysfunction of both cerebral and extra-cerebral blood vessels, rarefaction of the brain vascular network, and the loss of neurons in the brain cortex. Associated with these vasoprotective effects, IHT improved memory and lessened AD pathology. IHT increases endothelial production of nitric oxide (NO), thereby increasing regional cerebral blood flow and augmenting the vaso- and neuroprotective effects of endothelial NO. On the other hand, in AD excessive production of NO in microglia, astrocytes, and cortical neurons generates neurotoxic peroxynitrite. IHT enhances storage of excessive NO in the form of S-nitrosothiols and dinitrosyl iron complexes. Oxidative stress plays a pivotal role in the pathogenesis of AD, and IHT reduces oxidative stress in a number of experimental pathologies. Beneficial effects of IHT in experimental neuropathologies other than AD, including dyscirculatory encephalopathy, ischemic stroke injury, audiogenic epilepsy, spinal cord injury, and alcohol withdrawal stress have also been reported. Further research on the potential benefits of IHT in AD and other brain pathologies is warranted.
Collapse
Affiliation(s)
- Eugenia B Manukhina
- University of North Texas Health Science Center, Fort Worth, TX 76107-2699, USA Institute of General Pathology and Pathophysiology, Moscow 125315, Russian Federation
| | - H Fred Downey
- University of North Texas Health Science Center, Fort Worth, TX 76107-2699, USA
| | - Xiangrong Shi
- University of North Texas Health Science Center, Fort Worth, TX 76107-2699, USA
| | - Robert T Mallet
- University of North Texas Health Science Center, Fort Worth, TX 76107-2699, USA
| |
Collapse
|
94
|
Jukić S, Bubenik D, Pavlović N, Tušek AJ, Srček VG. Adaptation of CHO cells in serum-free conditions for erythropoietin production: Application of EVOP technique for process optimization. Biotechnol Appl Biochem 2016; 63:633-641. [DOI: 10.1002/bab.1468] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 12/07/2015] [Indexed: 01/17/2023]
Affiliation(s)
| | | | | | - Ana Jurinjak Tušek
- Faculty of Food Technology and Biotechnology; University of Zagreb; Zagreb Croatia
| | - Višnja Gaurina Srček
- Faculty of Food Technology and Biotechnology; University of Zagreb; Zagreb Croatia
| |
Collapse
|
95
|
Pichon A, Jeton F, El Hasnaoui-Saadani R, Hagström L, Launay T, Beaudry M, Marchant D, Quidu P, Macarlupu JL, Favret F, Richalet JP, Voituron N. Erythropoietin and the use of a transgenic model of erythropoietin-deficient mice. HYPOXIA 2016; 4:29-39. [PMID: 27800506 PMCID: PMC5085313 DOI: 10.2147/hp.s83540] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Despite its well-known role in red blood cell production, it is now accepted that erythropoietin (Epo) has other physiological functions. Epo and its receptors are expressed in many tissues, such as the brain and heart. The presence of Epo/Epo receptors in these organs suggests other roles than those usually assigned to this protein. Thus, the aim of this review is to describe the effects of Epo deficiency on adaptation to normoxic and hypoxic environments and to suggest a key role of Epo on main physiological adaptive functions. Our original model of Epo-deficient (Epo-TAgh) mice allowed us to improve our knowledge of the possible role of Epo in O2 homeostasis. The use of anemic transgenic mice revealed Epo as a crucial component of adaptation to hypoxia. Epo-TAgh mice survive well in hypoxic conditions despite low hematocrit. Furthermore, Epo plays a key role in neural control of ventilatory acclimatization and response to hypoxia, in deformability of red blood cells, in cerebral and cardiac angiogenesis, and in neuro- and cardioprotection.
Collapse
Affiliation(s)
- Aurélien Pichon
- Laboratory "Hypoxia and Lung" EA 2363, University Paris 13, Sorbonne Paris Cité, Bobigny Cedex; Laboratory of Excellence GR-Ex, Paris; Laboratory MOVE EA 6314, FSS, Poitiers University, Poitiers, France
| | - Florine Jeton
- Laboratory "Hypoxia and Lung" EA 2363, University Paris 13, Sorbonne Paris Cité, Bobigny Cedex; Laboratory of Excellence GR-Ex, Paris
| | | | - Luciana Hagström
- Laboratório Interdisciplinar de Biociências, Universidade de Brasília, Brasília, Brazil
| | - Thierry Launay
- Unité de Biologie Intégrative des Adaptations à l'Exercice, University Paris Saclay and Genopole , University Sorbonne-Paris-Cité, Paris, France
| | - Michèle Beaudry
- Laboratory "Hypoxia and Lung" EA 2363, University Paris 13, Sorbonne Paris Cité, Bobigny Cedex
| | - Dominique Marchant
- Laboratory "Hypoxia and Lung" EA 2363, University Paris 13, Sorbonne Paris Cité, Bobigny Cedex
| | - Patricia Quidu
- Laboratory "Hypoxia and Lung" EA 2363, University Paris 13, Sorbonne Paris Cité, Bobigny Cedex
| | - Jose-Luis Macarlupu
- High Altitude Unit, Laboratories for Research and Development, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Fabrice Favret
- Laboratory "Mitochondrie, Stress Oxydant et Protection Musculaire" EA 3072, University of Strasbourg, Strasbourg, France
| | - Jean-Paul Richalet
- Laboratory "Hypoxia and Lung" EA 2363, University Paris 13, Sorbonne Paris Cité, Bobigny Cedex; Laboratory of Excellence GR-Ex, Paris
| | - Nicolas Voituron
- Laboratory "Hypoxia and Lung" EA 2363, University Paris 13, Sorbonne Paris Cité, Bobigny Cedex; Laboratory of Excellence GR-Ex, Paris
| |
Collapse
|
96
|
RADWAN ESAMM, ABDULLAH RASEDEE, AL-QUBAISI MOTHANNASADIQ, EL ZOWALATY MOHAMEDE, NAADJA SEÏFEDDINE, ALITHEEN NOORJAHANB, OMAR ABDULRAHMAN. Effect of recombinant human erythropoietin and doxorubicinin combination on the proliferation of MCF-7 and MDA-MB231 breast cancer cells. Mol Med Rep 2016; 13:3945-52. [DOI: 10.3892/mmr.2016.4989] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Accepted: 08/19/2015] [Indexed: 11/05/2022] Open
|
97
|
Erythropoietin reduces storage lesions and decreases apoptosis indices in blood bank red blood cells. Rev Bras Hematol Hemoter 2016; 38:15-20. [PMID: 26969770 PMCID: PMC4786759 DOI: 10.1016/j.bjhh.2015.10.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 09/14/2015] [Accepted: 10/19/2015] [Indexed: 11/20/2022] Open
Abstract
Background Recent evidence shows a selective destruction of the youngest circulating red blood cells (neocytolysis) trigged by a drop in erythropoietin levels. Objective The aim of this study was to evaluate the effect of recombinant human erythropoietin beta on the red blood cell storage lesion and apoptosis indices under blood bank conditions. Methods Each one of ten red blood cell units preserved in additive solution 5 was divided in two volumes of 100 mL and assigned to one of two groups: erythropoietin (addition of 665 IU of recombinant human erythropoietin) and control (isotonic buffer solution was added). The pharmacokinetic parameters of erythropoietin were estimated and the following parameters were measured weekly, for six weeks: Immunoreactive erythropoietin, hemolysis, percentage of non-discocytes, adenosine triphosphate, glucose, lactate, lactate dehydrogenase, and annexin-V/esterase activity. The t-test or Wilcoxon's test was used for statistical analysis with significance being set for a p-value <0.05. Results Erythropoietin, when added to red blood cell units, has a half-life >6 weeks under blood bank conditions, with persistent supernatant concentrations of erythropoietin during the entire storage period. Adenosine triphosphate was higher in the Erythropoietin Group in Week 6 (4.19 ± 0.05 μmol/L vs. 3.53 ± 0.02 μmol/L; p-value = 0.009). The number of viable cells in the Erythropoietin Group was higher than in the Control Group (77% ± 3.8% vs. 71% ± 2.3%; p-value <0.05), while the number of apoptotic cells was lower (9.4% ± 0.3% vs. 22% ± 0.8%; p-value <0.05). Conclusions Under standard blood bank conditions, an important proportion of red blood cells satisfy the criteria of apoptosis. Recombinant human erythropoietin beta seems to improve storage lesion parameters and mitigate apoptosis.
Collapse
|
98
|
Leigh J, Saha MN, Mok A, Champsi O, Wang R, Lobb I, Sener A. Hydrogen Sulfide Induced Erythropoietin Synthesis is Regulated by HIF Proteins. J Urol 2016; 196:251-60. [PMID: 26880412 DOI: 10.1016/j.juro.2016.01.113] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2016] [Indexed: 12/27/2022]
Abstract
PURPOSE Anemia of end stage renal disease affects 90% of patients on hemodialysis and it is a tremendous concern of patients and health care providers. Renal disease creates a state of renal hypoxia, which may contribute to a lack of erythropoietin production from the kidney when low oxygen levels are sensed. This necessitates the use of exogenous erythropoietin preparations. MATERIALS AND METHODS Recent evidence suggests that endogenously derived hydrogen sulfide may mediate oxygen sensing in tissues. Given the known involvement of other small molecules such as nitric oxide in erythropoietin production and the observation of decreased urinary H2S levels in patients with renal failure, we postulated that H2S may be the primary mediator of erythropoietin production during hypoxia. PK1, 786-O and Hep3B cells were incubated in hypoxia (1% O2) for 24 hours. Hypoxic cells were treated with the H2S donor GYY 4137 and the H2S inhibitor hydroxylamine. Following hypoxia erythropoietin, HIF-1α, HIF-2α and CBS expression was measured by quantitative real-time polymerase chain reaction and Western blot. RESULTS Hydroxylamine administration led to a significant decrease in erythropoietin, HIF-1α, HIF-2α and CBS protein levels during hypoxia. This was rescued by administration of GYY 4137 for erythropoietin, CBS and HIF-2α. Additionally, CSE -/- mice placed in hypoxia for 72 hours showed decreased renal erythropoietin production compared to wild-type mice. CONCLUSIONS These data suggest previously undocumented interplay of the production and action of H2S during hypoxia with subsequent erythropoietin production. The use of novel hydrogen sulfide donors could represent an alternative to standard therapies of anemia of renal failure.
Collapse
Affiliation(s)
- Jennifer Leigh
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada; Matthew Mailing Centre for Translational Transplant Studies, London Health Sciences Centre, London, Ontario, Canada
| | - Manujendra N Saha
- Matthew Mailing Centre for Translational Transplant Studies, London Health Sciences Centre, London, Ontario, Canada
| | - Amy Mok
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada; Matthew Mailing Centre for Translational Transplant Studies, London Health Sciences Centre, London, Ontario, Canada
| | - Omar Champsi
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada; Matthew Mailing Centre for Translational Transplant Studies, London Health Sciences Centre, London, Ontario, Canada
| | - Rui Wang
- Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Ontario, Canada
| | - Ian Lobb
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada; Matthew Mailing Centre for Translational Transplant Studies, London Health Sciences Centre, London, Ontario, Canada
| | - Alp Sener
- Department of Surgery, Western University, London, Ontario, Canada; Multi-Organ Transplant Program, London Health Sciences Centre, London, Ontario, Canada.
| |
Collapse
|
99
|
Meininger M, Stepath M, Hennig R, Cajic S, Rapp E, Rotering H, Wolff M, Reichl U. Sialic acid-specific affinity chromatography for the separation of erythropoietin glycoforms using serotonin as a ligand. J Chromatogr B Analyt Technol Biomed Life Sci 2016; 1012-1013:193-203. [DOI: 10.1016/j.jchromb.2016.01.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 12/13/2015] [Accepted: 01/05/2016] [Indexed: 10/22/2022]
|
100
|
Nitric Oxide-cGMP Signaling Stimulates Erythropoiesis through Multiple Lineage-Specific Transcription Factors: Clinical Implications and a Novel Target for Erythropoiesis. PLoS One 2016; 11:e0144561. [PMID: 26727002 PMCID: PMC4699757 DOI: 10.1371/journal.pone.0144561] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 11/19/2015] [Indexed: 11/19/2022] Open
Abstract
Much attention has been directed to the physiological effects of nitric oxide (NO)-cGMP signaling, but virtually nothing is known about its hematologic effects. We reported for the first time that cGMP signaling induces human γ-globin gene expression. Aiming at developing novel therapeutics for anemia, we examined here the hematologic effects of NO-cGMP signaling in vivo and in vitro. We treated wild-type mice with NO to activate soluble guanylate cyclase (sGC), a key enzyme of cGMP signaling. Compared to untreated mice, NO-treated mice had higher red blood cell counts and total hemoglobin but reduced leukocyte counts, demonstrating that when activated, NO-cGMP signaling exerts hematopoietic effects on multiple types of blood cells in vivo. We next generated mice which overexpressed rat sGC in erythroid and myeloid cells. The forced expression of sGCs activated cGMP signaling in both lineage cells. Compared with non-transgenic littermates, sGC mice exhibited hematologic changes similar to those of NO-treated mice. Consistently, a membrane-permeable cGMP enhanced the differentiation of hematopoietic progenitors toward erythroid-lineage cells but inhibited them toward myeloid-lineage cells by controlling multiple lineage-specific transcription factors. Human γ-globin gene expression was induced at low but appreciable levels in sGC mice carrying the human β-globin locus. Together, these results demonstrate that NO-cGMP signaling is capable of stimulating erythropoiesis in both in vitro and vivo settings by controlling the expression of multiple lineage-specific transcription factors, suggesting that cGMP signaling upregulates erythropoiesis at the level of gene transcription. The NO-cGMP signaling axis may constitute a novel target to stimulate erythropoiesis in vivo.
Collapse
|