1
|
Gomaa S, Nassef M, Tabl G, Gabry SE. Immunoenhancing of the anti-cancer therapy and anti-oxidative stress by co-administration of granulocyte-colony stimulating factor-mobilized stem cells or cells derived from bone marrow and/or spleen plus vaccination with chemotherapeutic cyclophosphamide. Immunol Res 2025; 73:62. [PMID: 40091102 DOI: 10.1007/s12026-025-09610-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 02/19/2025] [Indexed: 03/19/2025]
Abstract
The combination of immunotherapy and chemotherapy, referred to as chemo-immunotherapy, represents a promising regimen for developing new cancer treatments that target the local tumor microenvironment and target tumors in their early stages. However, this approach carries potential risks, including myelo- and immunosuppression, as well as the emergence of chemo-resistant tumor cells. The purpose of this study was to investigate how well mobilizing hematopoietic stem cells (HSCs) work when used alongside chemotherapy and immunotherapy to enhance and modulate the immune response, thereby overcoming immunosuppression and eliminating distant cancer cells. Ehrlich ascetic carcinoma (EAC) tumor-bearing mice were intraperitoneal (i.p.) preconditioned with CTX (4 mg/mouse). EAC-bearing mice that were preconditioned with CTX were intravenous (i.v.) administered with adoptive transferred naive mice-derived bone marrow cells (nBMCs) at 5 × 106 through lateral tail vein (nBMCs group), adoptive transferred tumor-bearing mice-derived bone marrow cells (tBMCs) at 5 × 106 cell/mouse (tBMCs group), a combination of adoptive transferred naïve mice-derived bone marrow cells (nBMCs) and naïve mice-derived splenocytes (nSPs) at 5 × 106 (nBMCs/nSPs group), a combination of adoptive transferred tumor-bearing mice-derived bone marrow cells (tBMCs) and tumor-bearing mice derived-splenocytes (tSPs) at 5 × 106 cell/mouse (tBMCs/tSPs group), or G-CSF administrated subcutaneously (s.c.) at 5 µg/mouse (G-CSF group). Subsequently, all mice groups were vaccinated with tumor lysate at a dosage of 100 µg/mouse. Treating EAC tumor-bearing mice with G-CSF, adoptive transferred nBMCs, adoptive transferred tBMCs, adoptive transferred nBMCs/nSPs, adoptive transferred tBMCs/tSPs, resulted in a significantly enhanced anti-tumor effect that was evidenced by increased anti-proliferative activity and growth inhibition against EAC tumor cells, increased necrosis and apoptosis rates among EAC tumor cells, restricted tumor growth in EAC tumor-bearing mice, and reduced levels of carcinoembryonic antigen (CEA) tumor marker. Furthermore, there was an improvement in serum levels of antioxidant enzyme superoxide dismutase (SOD) and malondialdehyde (MDA) in EAC tumor-bearing mice receiving G-CSF, adoptive transferred tBMCs, adoptive transferred nBMCs/nSPs, and adoptive transferred tBMCs/tSPs. Notably, this treatment regimen ameliorates liver and kidney damage associated with CTX administration in EA tumor-bearing mice. The integration of G-CSF-mobilized HSCs, adoptive transferred nBMCs, adoptive transferred tBMCs, adoptive transferred nBMCs/nSPs combination, and adoptive transferred tBMCs/tSPs combination may yield powerful anti-cancer therapy, thereby facilitating more effective anti-tumor immunotherapy strategies when align with anti-tumor responses. This research may propose a novel therapeutic approach that combines chemotherapy and immunotherapy for addressing early-stage cancer. Further research is necessary to connect the biomedical application and heterogeneity of human tumors and immune systems of this regimen to both diagnostic and therapeutic methodologies.
Collapse
Affiliation(s)
- Soha Gomaa
- Department of Zoology, Science Faculty, University of Tanta, 31527, Tanta, Egypt.
| | - Mohamed Nassef
- Department of Zoology, Science Faculty, University of Tanta, 31527, Tanta, Egypt
| | - Ghada Tabl
- Department of Zoology, Science Faculty, University of Tanta, 31527, Tanta, Egypt
| | - Shaimaa El Gabry
- Department of Zoology, Science Faculty, University of Tanta, 31527, Tanta, Egypt
| |
Collapse
|
2
|
Qian F, Du X, He Y. Causal association of inflammation with ischemic stroke and its subtypes: a bidirectional Mendelian randomization study. J Stroke Cerebrovasc Dis 2025; 34:108190. [PMID: 39675594 DOI: 10.1016/j.jstrokecerebrovasdis.2024.108190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 11/28/2024] [Accepted: 12/09/2024] [Indexed: 12/17/2024] Open
Abstract
BACKGROUND Emerging evidence underscores a bidirectional relationship between ischemic stroke (IS) and inflammation, yet the causality of this association remains uncertain. We conducted a two-sample bidirectional Mendelian randomization (MR) study aimed at investigating the causal links between inflammation and IS. METHODS Single nucleotide polymorphism from genome-wide association studies of 112 inflammatory cytokines and IS were chosen as instrumental variables. We evaluated the causal effects of inflammatory factors on IS outcomes and examined the mediating effects of risk factors for IS. Additionally, reverse MR analysis was conducted to determine whether the occurrence of IS influenced levels of inflammatory cytokines. Causal associations were assessed using inverse variance weighting, complemented by sensitivity analyses incorporating weighted median and MR-Egger methods. RESULTS We found associations between genetically predicted plasma levels of 25 inflammatory factors and IS along with its subtypes. MR supports smoking, body mass index, atrial fibrillation, coronary artery disease, heart failure, systolic blood pressure, diastolic blood pressure and type 2 diabetes as risk factors for IS. Notably, coronary artery disease and heart failure seemed to mediate the RANTES, HGF, IL-5 associations with IS. In addition, reverse MR analysis suggested a causal relationship between IS and its subtypes and 19 inflammatory factors. CONCLUSION In summary, inflammation was suggestively causally associated with the risk of IS, and inflammatory cytokines had downstream effect on IS. Future studies should explore whether inflammatory factors found to have significant associations with IS risk could be manipulated to reduce IS risk, and the neuroinflammatory mechanisms after IS.
Collapse
Affiliation(s)
- FangFang Qian
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, PR China.
| | - XiaoHui Du
- Department of Rehabilitation Medicine, Community Health Service Center, Lvxiang Town, Jinshan District, Shanghai, PR China.
| | - YouHua He
- Department of Comprehensive Medical Treatment Ward, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China.
| |
Collapse
|
3
|
Fujimaki Y, Kondo K, Nishijima H, Kikuta S, Yamasoba T. Granulocyte colony-stimulating factor promotes regeneration of severed facial nerve in rats. Front Neurosci 2024; 18:1442614. [PMID: 39712221 PMCID: PMC11662712 DOI: 10.3389/fnins.2024.1442614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 11/04/2024] [Indexed: 12/24/2024] Open
Abstract
Background and aim The administration of growth and neurotrophic factors has been attempted experimentally as a new therapeutic strategy for severe facial paralysis. Granulocyte colony-stimulating factor (G-CSF) has an effect on the treatment of central nervous system injuries, such as cerebral infarction and spinal cord injury. This study aimed at examining the effects of G-CSF on facial nerve regeneration in rats. Methods The left facial nerve of rats was either partially resected (resection group) or severed and sutured (suture group) at the main trunk outside the temporal bone. In each surgical group, saline or G-CSF was administered via the gelatin hydrogel drug delivery system. The suture group was further divided into two subgroups for the late administration of G-CSF (2 weeks after surgical treatment) or immediate administration of G-CSF after surgical treatment. Recovery of the facial nerve was assessed by the evaluation of facial movements (after 12 weeks), complex muscle action potential amplitude measurements (after 2, 4, 8, and 12 weeks), electroneurography values (after 12 weeks), and histological evaluation (comparison of myelinated axon diameters among the groups). Results Recovery of the function and morphology of damaged nerves was faster in the suture groups than in the resection group. In the suture groups, recovery was faster for G-CSF-treated rats than for saline-treated rats. Furthermore, recovery was faster in the group that received G-CSF immediately after surgical treatment than in the group that received G-CSF 2 weeks later. However, the group that received G-CSF 2 weeks later also showed faster recovery than did the control group. Conclusion G-CSF effectively promoted nerve regeneration during facial nerve paralysis. Thus, G-CSF may be a potential treatment strategy for injured facial nerves as it has been safely administered in clinical treatments for hematological diseases.
Collapse
Affiliation(s)
- Yoko Fujimaki
- Department of Otorhinolaryngology and Head and Neck Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kenji Kondo
- Department of Otorhinolaryngology and Head and Neck Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hironobu Nishijima
- Department of Otorhinolaryngology and Head and Neck Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shu Kikuta
- Department of Otolaryngology-Head and Neck Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Tatsuya Yamasoba
- Department of Otorhinolaryngology and Head and Neck Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
4
|
Weisbrod LJ, Nilles-Melchert TT, Bergjord JR, Surdell DL. Granulocyte-Colony Stimulating Factor Improves Neurological and Functional Outcomes in Patients With Traumatic Incomplete Spinal Cord Injuries: A Systematic Review With Meta-Analyses. Neurotrauma Rep 2024; 5:467-482. [PMID: 39582880 PMCID: PMC11579545 DOI: 10.1089/neur.2023.0099] [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: 11/26/2024] Open
Abstract
Spinal cord injury (SCI) is a cause for significant morbidity, often resulting in long-term disability. We compared outcomes after administration of granulocyte-colony stimulating factor (G-CSF) versus controls. MEDLINE, Embase, and Cochrane Library database searches yielded 222 records; six met study inclusion criteria. Fixed-effect and random-effects models were used to establish odds ratios (ORs) and mean difference (MD) with 95% confidence intervals (CIs) for each outcome. The results of the pooled analysis showed that in patients with incomplete SCI, G-CSF resulted in increased American Spinal Cord Injury Association (ASIA) motor scores at 3 months (MD = 0.57 [95% CI = 0.04, 1.10], I 2 = 63.84%, p = 0.036), 6 months (MD = 4.18 [95% CI = 0.55, 7.80], I 2 = 98.75%, p = 0.024), change in ASIA pinprick scores at 6 months (MD = 3.38 [95% CI = 1.48, 5.28], I 2 = 89.78%, p < 0.001), and increased Spinal Cord Independence Measure (SCIM) III score at 6 months (MD = 3.27 [95% CI = 1.13, 5.41], I 2 = 91.86%, p = 0.003). G-CSF resulted in more adverse events than the non-MP control groups (OR = 1.44 [95% CI = 0.38, 2.50], I 2 = 0%, p = 0.008), but fewer than the MP control groups (OR = -4.2 [95% CI = -5.72, -2.68], I 2 = 0%, p < 0.001). Systemic white blood cell count increased after administration of G-CSF in comparison to baseline (MD = 3.57 [95% CI = 2.79, 4.35], I 2 = 55.06%, p < 0.001). G-CSF did not statistically increase ASIA Impairment Scale at 3 months (MD = 0.48 [95% CI = -0.33, 1.28], I 2 = 0%, p = 0.246) or at 6 months (MD = 1.84 [95% CI = -0.10, 3.79], I 2 = 50.09%, p = 0.063). These meta-analyses of six studies suggest that G-CSF for the treatment of incomplete SCI may result in improved neurological outcomes when compared to the controls. The results are limited by a small sample size with heterogeneity between studies. More robust prospective, randomized studies are necessary to help inform the safety and efficacy of G-CSF.
Collapse
Affiliation(s)
- Luke J. Weisbrod
- Department of Neurosurgery, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | | | | | - Daniel L. Surdell
- Department of Neurosurgery, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
5
|
Chen MS, Hu CL, Jiang SK, Chong ZY, Chen JC. Modulation of secretory factors by lipofundin contributes to its anti‑neuroinflammatory effects. Exp Ther Med 2024; 27:169. [PMID: 38476917 PMCID: PMC10929000 DOI: 10.3892/etm.2024.12456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 01/16/2024] [Indexed: 03/14/2024] Open
Abstract
As the global population ages, the prevalence of neuroinflammatory diseases such as Alzheimer's disease, Parkinson's disease and stroke continues to increase. Therefore, it is necessary to develop preventive and therapeutic methods against neuroinflammatory diseases. Lipofundin is a lipid emulsion commonly used in clinical anesthetic solvents and nutritional supplements. Lipid emulsions have been shown to possess anti-inflammatory properties. However, the potential beneficial effect of lipofundin against neuroinflammation requires elucidation. In the present study, two cell models were used to investigate the efficacy of lipofundin against neuroinflammation. In the first model, BV2 mouse microglial cells were treated with lipopolysaccharide (LPS) to induce nitric oxide (NO) production as a model of neuroinflammation. In the second model, HMC3 human microglial were activated by LPS, and changes in the secretion of factors associated with inflammation were analyzed using Luminex xMAP® technology. Griess assay results revealed that lipofundin significantly prevented and treated LPS-induced NO production. An anti-neuroinflammatory effect was also observed in HMC3 cells, where lipofundin exhibited excellent preventive and therapeutic properties by reducing the LPS-induced expression and secretion of interleukin-1β. Notably, lipofundin also promoted the secretion of certain growth factors, suggesting a potential neuroprotective effect. These results demonstrate that, in addition to its role as a solvent for drugs and nutritional support, lipofundin may also have beneficial effects in alleviating the progression of neuroinflammation. These findings may serve as an important reference for future translational medicine applications.
Collapse
Affiliation(s)
- Ming-Shan Chen
- Department of Anesthesiology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 60002, Taiwan, R.O.C
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung 41354, Taiwan, R.O.C
| | - Chia-Lin Hu
- Department of Anesthesiology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 60002, Taiwan, R.O.C
| | - Shin-Kuang Jiang
- Department of Neurology, China Medical University Hospital, Taichung 404332, Taiwan, R.O.C
| | - Zhi-Yong Chong
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi 600355, Taiwan, R.O.C
| | - Jui-Chieh Chen
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi 600355, Taiwan, R.O.C
| |
Collapse
|
6
|
Gržeta Krpan N, Harej Hrkać A, Janković T, Dolenec P, Bekyarova E, Parpura V, Pilipović K. Chemically Functionalized Single-Walled Carbon Nanotubes Prevent the Reduction in Plasmalemmal Glutamate Transporter EAAT1 Expression in, and Increase the Release of Selected Cytokines from, Stretch-Injured Astrocytes in Vitro. Cells 2024; 13:225. [PMID: 38334617 PMCID: PMC10854924 DOI: 10.3390/cells13030225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 01/15/2024] [Accepted: 01/17/2024] [Indexed: 02/10/2024] Open
Abstract
We tested the effects of water-soluble single-walled carbon nanotubes, chemically functionalized with polyethylene glycol (SWCNT-PEG), on primary mouse astrocytes exposed to a severe in vitro simulated traumatic brain injury (TBI). The application of SWCNT-PEG in the culture media of injured astrocytes did not affect cell damage levels, when compared to those obtained from injured, functionalization agent (PEG)-treated cells. Furthermore, SWCNT-PEG did not change the levels of oxidatively damaged proteins in astrocytes. However, this nanomaterial prevented the reduction in plasmalemmal glutamate transporter EAAT1 expression caused by the injury, rendering the level of EAAT1 on par with that of control, uninjured PEG-treated astrocytes; in parallel, there was no significant change in the levels of GFAP. Additionally, SWCNT-PEG increased the release of selected cytokines that are generally considered to be involved in recovery processes following injuries. As a loss of EAATs has been implicated as a culprit in the suffering of human patients from TBI, the application of SWCNT-PEG could have valuable effects at the injury site, by preventing the loss of astrocytic EAAT1 and consequently allowing for a much-needed uptake of glutamate from the extracellular space, the accumulation of which leads to unwanted excitotoxicity. Additional potential therapeutic benefits could be reaped from the fact that SWCNT-PEG stimulated the release of selected cytokines from injured astrocytes, which would promote recovery after injury and thus counteract the excess of proinflammatory cytokines present in TBI.
Collapse
Affiliation(s)
- Nika Gržeta Krpan
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, HR-51000 Rijeka, Croatia; (N.G.K.); (A.H.H.); (T.J.); (P.D.)
| | - Anja Harej Hrkać
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, HR-51000 Rijeka, Croatia; (N.G.K.); (A.H.H.); (T.J.); (P.D.)
| | - Tamara Janković
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, HR-51000 Rijeka, Croatia; (N.G.K.); (A.H.H.); (T.J.); (P.D.)
| | - Petra Dolenec
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, HR-51000 Rijeka, Croatia; (N.G.K.); (A.H.H.); (T.J.); (P.D.)
| | - Elena Bekyarova
- Department of Chemistry, University of California, Riverside, CA 92521, USA;
| | - Vladimir Parpura
- International Translational Neuroscience Research Institute, Zhejiang Chinese Medical University, Hangzhou 310053, China;
| | - Kristina Pilipović
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, HR-51000 Rijeka, Croatia; (N.G.K.); (A.H.H.); (T.J.); (P.D.)
| |
Collapse
|
7
|
Dharra R, Kumar Sharma A, Datta S. Emerging aspects of cytokine storm in COVID-19: The role of proinflammatory cytokines and therapeutic prospects. Cytokine 2023; 169:156287. [PMID: 37402337 PMCID: PMC10291296 DOI: 10.1016/j.cyto.2023.156287] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 06/24/2023] [Indexed: 07/06/2023]
Abstract
COVID-19 has claimed millions of lives during the last 3 years since initial cases were reported in Wuhan, China, in 2019. Patients with COVID-19 suffer from severe pneumonia, high fever, acute respiratory distress syndrome (ARDS), and multiple-organ dysfunction, which may also result in fatality in extreme cases. Cytokine storm (CS) is hyperactivation of the immune system, wherein the dysregulated production of proinflammatory cytokines could result in excessive immune cell infiltrations in the pulmonary tissues, resulting in tissue damage. The immune cell infiltration could also occur in other tissues and organs and result in multiple organs' dysfunction. The key cytokines implicated in the onset of disease severity include TNF-α, IFN-γ, IL-6, IL-1β, GM-CSF, and G-CSF. Controlling the CS is critical in treating COVID-19 disease. Therefore, different strategies are employed to mitigate the effects of CS. These include using monoclonal antibodies directed against soluble cytokines or the cytokine receptors, combination therapies, mesenchymal stem cell therapy, therapeutic plasma exchange, and some non-conventional treatment methods to improve patient immunity. The current review describes the role/s of critical cytokines in COVID-19-mediated CS and the respective treatment modalities.
Collapse
Affiliation(s)
- Renu Dharra
- CSIR-Institute of Microbial Technology, Sector 39 A, Chandigarh 160036, India
| | - Anil Kumar Sharma
- Department of Bio-Science and Technology, M. M. Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala 133207, India
| | - Sonal Datta
- Department of Bio-Science and Technology, M. M. Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala 133207, India.
| |
Collapse
|
8
|
Mousavi SR, Mohammadpour AH, Moshiri M, Feizy J, Pourtaji A, Samadi S. A pilot study of neuroprotective effect of granulocyte colony-stimulating factor (G-CSF) in patients with carbon monoxide poisoning: a double-blind, randomized, placebo-controlled trial. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:1257-1267. [PMID: 36715735 DOI: 10.1007/s00210-023-02395-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/11/2023] [Indexed: 01/31/2023]
Abstract
Although neuroprotective effects of granulocyte colony-stimulating factor (G-CSF) have been shown in rats exposed to carbon monoxide (CO), this pilot clinical trial was performed to assess the feasibility of treatment with G-CSF in patients with acute CO poisoning. A double-blind, randomized, placebo-controlled pilot clinical trial was conducted on twenty-six patients with acute CO poisoning. G-CSF (90 μg/kg) was administered intravenously for 72 h. Demographic data, routine laboratory tests, differential blood counts, venous blood gas, and adverse reactions were recorded. The primary endpoint was brain ischemia improvement based on CT findings and the secondary endpoints examined improvements in the modified Rankin Scale (mRS), National Institutes of Health Stroke Scale (NIHSS), and Barthel Index as well as S-100β concentrations. Fourteen patients received G-CSF, and 12 received a placebo. Twenty-six were followed for 30 days and no one in both groups died during follow-up. Neurological complications, brain ischemic changes, Barthel, and mRS were compared between the two groups on determined days after the onset of therapeutic intervention, and no significant differences were observed between the two groups. Favorable results were achieved for treated patients by different measures; NIHSS was decreased 72 h after treatment (p = 0.046), and S-100β levels were significantly higher in the G-CSF group than in the control group, 12 h and 72 h after the treatment. G-CSF appears to have potential effects on several clinical parameters in patients with acute CO poisoning. The trial was registered at the Iranian Registry of Clinical Trials with the ID: (IRCT201607232083N7).
Collapse
Affiliation(s)
- Seyed Reza Mousavi
- Medical Toxicology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Hooshang Mohammadpour
- Department of Clinical Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Moshiri
- Medical Toxicology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Javad Feizy
- Department of Food Safety and Quality Control, Research Institute of Food Science and Technology (RIFST), Mashhad, Iran
| | - Atena Pourtaji
- Pharmaceutical Research Center, Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Sara Samadi
- Department of Clinical Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
9
|
Zhang DD, Zhang CY, Zhang YX, Cui HP, Jiao Chen, Wen-Zhi Ma, Jia H. G-CSF reduces loss of dopaminergic neurons by inhibiting TNF-α and IL-1β in mouse model of Parkinson's disease. Int J Neurosci 2023; 133:278-289. [PMID: 33781148 DOI: 10.1080/00207454.2021.1910259] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
PURPOSE OF THE STUDY granulocyte-colony stimulating factor (G-CSF) is a hematopoietic growth factor existing in neutrophils, glial cells and neurons. Increasing researches discovered that G-CSF improved cell survival in neurodegenerative diseases by its anti-inflammatory effect. However, the effect of G-CSF in suppressing inflammation in Parkinson's disease (PD) remains unclear. Thus, the purpose of this study is to explored the anti-inflammatory effect of G-CSF in mouse model of PD. MATERIALS AND METHODS G-CSF was administrated in the PD model induced by MPTP. Subsequently, the protein of tyrosine hydroxylase (TH), ionized calcium-binding adaptor molecule 1 (Iba-1) and the inflammatory cytokines including tumor necrosis factor-alpha (TNF-α) and interleukin-1 beta (IL-1β) in the midbrain were examined. In addition, the phosphorylated mitogen-activated protein kinases (MAPK) including c-Jun NH2-terminal kinase (JNK), extracellular signal-regulated kinase (ERK) and p38 MAPK in the midbrain were investigated. RESULTS Compared with the MPTP group, the protein of TH in the midbrain was increased, while the Iba-1 and the inflammatory factors were decreased. In addition, the expression of phosphorylated JNK (p-JNK) in the midbrain of the MPTP + G-CSF group was decreased, while the phosphorylated ERK (p-ERK) levels were elevated. CONCLUSIONS These findings emphasize that G-CSF inhibited the degradation of DA neurons. The protective effect is associated with the reduction of the inflammatory factors caused by the inhibition of the microglial activation. Moreover, G-CSF may decrease the inflammatory factors through the decrease of P-JNK and the increase of P-ERK.
Collapse
Affiliation(s)
- Dan-Dan Zhang
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China.,Department of physiology, School of Basic Medical Sciences, Chengde Medical University, Chengde, China
| | - Cheng-Yun Zhang
- Department of Human Anatomy, School of Basic Medical Sciences, Chengde Medical University, Chengde, China
| | - Yu-Xin Zhang
- Department of Human Anatomy, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, China
| | - Hai-Peng Cui
- Department of Pathophysiology, School of Basic Medical Sciences, Chengde Medical University, Chengde, China
| | - Jiao Chen
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Wen-Zhi Ma
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China.,Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Key Laboratory of Reproduction and Genetics of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China.,Center for Reproductive Biology and Health, School of Agricultural Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Hua Jia
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China.,Center for Reproductive Biology and Health, School of Agricultural Sciences, The Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
10
|
Maharaj D, Kaur K, Saltese A, Gouvea J. Personalized Precision Immunotherapy for Amyotrophic Lateral Sclerosis (ALS). Crit Rev Immunol 2023; 43:1-11. [PMID: 37938192 DOI: 10.1615/critrevimmunol.2023048372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2023]
Abstract
Neurological syndrome amyotrophic lateral sclerosis (ALS) affects motor neurons and is characterized by progressive motor neuron loss in the brain and spinal cord. ALS starts with mainly focal onset but when the disease progresses, it spreads to different parts of the body, with survival limits of 2-5 years after disease initiation. To date, only supportive care is provided for ALS patients, and no effective treatment or cure has been discovered. This review is focused on clinical and immunological aspects of ALS patients, based on our case studies, and we discuss the treatment we have provided to those patients based on a detailed evaluation of their peripheral blood immune cells and blood-derived serum secreted factors, cytokines, chemokines and growth factors. We show that using a personalized approach of low dose immunotherapy there is an improvement in the effects on inflammation and immunological dysfunction.
Collapse
Affiliation(s)
- Dipnarine Maharaj
- South Florida Bone Marrow Stem Cell Transplant Institute, DBA Maharaj Institute of Immune Regenerative Medicine, Boynton Beach, FL 33437
| | - Kawaljit Kaur
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, University of California School of Dentistry, 10833 Le Conte Ave, 90095 Los Angeles, CA, USA
| | - Adrian Saltese
- South Florida Bone Marrow Stem Cell Transplant Institute, DBA Maharaj Institute of Immune Regenerative Medicine, Boynton Beach, FL 33437, USA
| | - Jacqueline Gouvea
- South Florida Bone Marrow Stem Cell Transplant Institute, DBA Maharaj Institute of Immune Regenerative Medicine, Boynton Beach, FL 33437, USA
| |
Collapse
|
11
|
Knorr DY, Rodriguez Polo I, Pies HS, Schwedhelm-Domeyer N, Pauls S, Behr R, Heinrich R. The cytokine receptor CRLF3 is a human neuroprotective EV-3 (Epo) receptor. Front Mol Neurosci 2023; 16:1154509. [PMID: 37168680 PMCID: PMC10165946 DOI: 10.3389/fnmol.2023.1154509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 03/15/2023] [Indexed: 05/13/2023] Open
Abstract
The evolutionary conserved orphan cytokine receptor-like factor 3 (CRLF3) has been implicated in human disease, vertebrate hematopoiesis and insect neuroprotection. While its specific functions are elusive, experimental evidence points toward a general role in cell homeostasis. Erythropoietin (Epo) is a major regulator of vertebrate hematopoiesis and a general cytoprotective cytokine. Erythropoietic functions mediated by classical Epo receptor are understood in great detail whereas Epo-mediated cytoprotective mechanisms are more complex due to involvement of additional Epo receptors and a non-erythropoietic splice variant with selectivity for certain receptors. In the present study, we show that the human CRLF3 mediates neuroprotection upon activation with the natural Epo splice variant EV-3. We generated CRLF3 knock-out iPSC lines and differentiated them toward the neuronal lineage. While apoptotic death of rotenone-challenged wild type iPSC-derived neurons was prevented by EV-3, EV-3-mediated neuroprotection was absent in CRLF3 knock-out neurons. Rotenone-induced apoptosis and EV-3-mediated neuroprotection were associated with differential expression of pro-and anti-apoptotic genes. Our data characterize human CRLF3 as a receptor involved in Epo-mediated neuroprotection and identify CRLF3 as the first known receptor for EV-3.
Collapse
Affiliation(s)
- Debbra Y. Knorr
- Department of Cellular Neurobiology, Johann-Friedrich-Blumenbach Institute for Zoology and Anthropology, Georg-August University Göttingen, Göttingen, Germany
- *Correspondence: Debbra Y. Knorr,
| | - Ignacio Rodriguez Polo
- Department of Developmental Biology, Göttingen Center for Molecular Biosciences, Georg-August University Göttingen, Göttingen, Germany
- Research Platform Degenerative Diseases, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany
- Developmental Models Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Hanna S. Pies
- Department of Cellular Neurobiology, Johann-Friedrich-Blumenbach Institute for Zoology and Anthropology, Georg-August University Göttingen, Göttingen, Germany
| | - Nicola Schwedhelm-Domeyer
- Department of Cellular Neurobiology, Johann-Friedrich-Blumenbach Institute for Zoology and Anthropology, Georg-August University Göttingen, Göttingen, Germany
| | - Stephanie Pauls
- Department of Cellular Neurobiology, Johann-Friedrich-Blumenbach Institute for Zoology and Anthropology, Georg-August University Göttingen, Göttingen, Germany
| | - Rüdiger Behr
- Research Platform Degenerative Diseases, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany
| | - Ralf Heinrich
- Department of Cellular Neurobiology, Johann-Friedrich-Blumenbach Institute for Zoology and Anthropology, Georg-August University Göttingen, Göttingen, Germany
- Ralf Heinrich,
| |
Collapse
|
12
|
Pharmacological Strategies for Stroke Intervention: Assessment of Pathophysiological Relevance and Clinical Trials. Clin Neuropharmacol 2023; 46:17-30. [PMID: 36515293 DOI: 10.1097/wnf.0000000000000534] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVES The present review describes stroke pathophysiology in brief and discusses the spectrum of available treatments with different promising interventions that are in clinical settings or are in clinical trials. METHODS Relevant articles were searched using Google Scholar, Cochrane Library, and PubMed. Keywords for the search included ischemic stroke, mechanisms, stroke interventions, clinical trials, and stem cell therapy. RESULTS AND CONCLUSION Stroke accounts to a high burden of mortality and morbidity around the globe. Time is an important factor in treating stroke. Treatment options are limited; however, agents with considerable efficacy and tolerability are being continuously explored. With the advances in stroke interventions, new therapies are being formulated with a hope that these may aid the ongoing protective and reparative processes. Such therapies may have an extended therapeutic time window in hours, days, weeks, or longer and may have the advantage to be accessible by a majority of the patients.
Collapse
|
13
|
Song S, Kong X, Wang B, Sanchez-Ramos J. Recovery from Traumatic Brain Injury Following Treatment with Δ9-Tetrahydrocannabinol Is Associated with Increased Expression of Granulocyte-Colony Stimulating Factor and Other Neurotrophic Factors. Cannabis Cannabinoid Res 2022; 7:415-423. [PMID: 33998887 PMCID: PMC9418356 DOI: 10.1089/can.2020.0119] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Introduction: The hematopoietic cytokine granulocyte-colony stimulating factor (G-CSF) is well known to stimulate proliferation of blood stem/progenitor cells of the leukocyte lineage, but is also recognized as a neurotrophic factor involved in brain self-repair processes. G-CSF administration has been shown to promote recovery from experimental models of traumatic brain injury (TBI) and to modulate components of the endocannabinoid system (eCS). Conversely, Δ9-tetrahydrocannabinol (Δ9THC) treatment of normal mice has been shown to increase blood levels of G-CSF in the periphery. Hypothesis: Administration of the phytocannabinoid Δ9THC will enhance brain repair following controlled cortical impact (CCI) by upregulating G-CSF and other neurotrophic factors (brain-derived neurotrophic factor [BDNF] and glial-derived neurotrophic factor [GDNF]) in brain regions. Materials and Methods: C57BL/6J mice underwent CCI and were treated for 3 days with THC 3 mg/kg intraperitoneally. Motor function on a rotarod was recorded at baseline and 3, 7, and 14 days after CCI. Groups of mice were euthanized at 7 and 14 days. G-CSF, BDNF, and GDNF expression were measured at 7 and 14 days in cerebral cortex, striatum, and hippocampus on the side of the trauma. Results: Δ9THC-treated mice ran on the rotarod longer than vehicle-treated mice and recovered to normal rotarod performance levels at 2 weeks. These mice, compared to vehicle-treated animals, exhibited significant upregulation of G-CSF as well as BDNF and GDNF in cerebral cortex, striatum, and hippocampus. Conclusion: Administration of the phytocannabinoid Δ9THC promotes significant recovery from TBI and is associated with upregulation of brain G-CSF, BDNF, and GDNF, neurotrophic factors previously shown to mediate brain self-repair following TBI and stroke.
Collapse
Affiliation(s)
- Shijie Song
- James Haley VA Medical Center, Tampa, Florida, USA
- Department of Neurology, University of South Florida, Tampa, Florida, USA
| | | | - Bangmei Wang
- James Haley VA Medical Center, Tampa, Florida, USA
- Department of Neurology, University of South Florida, Tampa, Florida, USA
| | - Juan Sanchez-Ramos
- Department of Neurology, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
14
|
Chitu V, Biundo F, Stanley ER. Colony stimulating factors in the nervous system. Semin Immunol 2021; 54:101511. [PMID: 34743926 DOI: 10.1016/j.smim.2021.101511] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/23/2021] [Indexed: 01/02/2023]
Abstract
Although traditionally seen as regulators of hematopoiesis, colony-stimulating factors (CSFs) have emerged as important players in the nervous system, both in health and disease. This review summarizes the cellular sources, patterns of expression and physiological roles of the macrophage (CSF-1, IL-34), granulocyte-macrophage (GM-CSF) and granulocyte (G-CSF) colony stimulating factors within the nervous system, with a particular focus on their actions on microglia. CSF-1 and IL-34, via the CSF-1R, are required for the development, proliferation and maintenance of essentially all CNS microglia in a temporal and regional specific manner. In contrast, in steady state, GM-CSF and G-CSF are mainly involved in regulation of microglial function. The alterations in expression of these growth factors and their receptors, that have been reported in several neurological diseases, are described and the outcomes of their therapeutic targeting in mouse models and humans are discussed.
Collapse
Affiliation(s)
- Violeta Chitu
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Fabrizio Biundo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - E Richard Stanley
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
15
|
Chen X, Sun W, Zhong P, Wu D. Colony-Stimulating Factors on Mobilizing CD34 + Cells and Improving Neurological Functions in Patients With Stroke: A Meta-Analysis and a Systematic Review. Front Pharmacol 2021; 12:704509. [PMID: 34366857 PMCID: PMC8339259 DOI: 10.3389/fphar.2021.704509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 06/07/2021] [Indexed: 11/13/2022] Open
Abstract
Background and Purpose: CSF therapy is considered a promising therapeutic approach for stroke. We performed a meta-analysis to explore the safety and efficacy of CSF in published clinical stroke studies. Methods: We searched articles online and manually. Two reviewers selected studies independently, selecting data based on study quality, characteristics of intervention (administration time, observation time, type, dose, and injection approach of CSF), and the baseline characteristics of patients (age, sex, hypertension, diabetes, smoker, and lipids) were extracted. Main prognosis outcomes were measured as all-cause death in severe adverse events (SAE) and recurrent stroke in SAE. Secondary outcomes were measured as CD34+ cell counts in periphery blood at day 5, National Institutes of Health Stroke Scale (NIHSS), and Barthel index (BI), Side effects of CSF were taken as the indicator of safety. STATA13 software was used to perform the meta-analysis.Keywords: Stroke, Colony-stimulating factor, Meta-analysis, therapy, Neurological Diseases Results: This meta-analysis involved 485 patients from eight studies. Among them, 475 patients from seven studies were gauged SAE (all-cause death), 393 patients from six studies were checked SAE (recurrent stroke); 137 patients from three studies underwent CD34+ measurement, 389 patients from six studies were tested NIHSS and 307 patients from five studies accessed BI. Compared with the control group, both all-causes death (RR= 1.73, 95%CI= (0.61, 4.92), P=0.735, I2=0.0%) and recurrent stroke (RR= 0.43, 95%CI= (0.14, 1.32), P=0.214, I2=33.1%) present no statistical differences, indicating that the application of CSF does not statistically alter the prognosis of patients with stroke. The application of CSF effectively enhanced CD34+ cell counts in periphery blood at day 5 (SMD= 1.23, 95%CI= (0.54, 1.92), P=0.04, I2=69.0%) but did not statistically impact NIHSS (SMD= -0.40, 95%CI= (-0.93, 0.13), P ≤ 0.001, I2=79.7%) or BI (SMD= 0.04, 95%CI= (-0.38, 0.46), P=0.068, I2=54.3%). Conclusion: Our study consolidates the security of CSF administration for its exerting no effect on detrimental outcomes. It has proven to be effective in elevating CD34+ cell counts in periphery blood at day 5, indicating CSF may participate in stroke recovery, but its efficacy in stroke recovery remains detected.
Collapse
Affiliation(s)
- Xiuqi Chen
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Wenbo Sun
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Ping Zhong
- Department of Neurology, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| | - Danhong Wu
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| |
Collapse
|
16
|
Yeh CC, Yang CP, Ma KH, Shih JH, Tseng CS, Huang YS. Endogenous Expression of G-CSF in Rat Dorsal Root Ganglion Neurons after Nerve Injury. Brain Sci 2021; 11:brainsci11070956. [PMID: 34356190 PMCID: PMC8303554 DOI: 10.3390/brainsci11070956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/16/2021] [Accepted: 07/17/2021] [Indexed: 11/25/2022] Open
Abstract
Granulocyte colony-stimulating factor (G-CSF) has been reported to modulate pain function following nerve injury. However, the expression of endogenous G-CSF in the dorsal root ganglion (DRG) and the response to nerve injury remain unclear. In the present study, we demonstrated that G-CSF and G-CSFR are mainly expressed in both small- and medium-diameter DRG neurons in rats and are responsible for transmitting pain responses. G-CSF and G-CSFR were co-expressed in certain nociceptive DRG neurons. In addition, G-CSF was expressed in satellite glial cells around large-diameter DRG neurons. After sciatic nerve injury, the number of G-CSF-positive DRG neurons was increased in both the ipsilateral and contralateral lesion sites in rats. However, G-CSF expression in satellite glial cells was not affected by nerve injury. To clarify the role of G-CSF in pain, exogenous G-CSF was administered to a rat model of neuropathic pain induced by partial sciatic nerve transaction (PST). Our results indicate that treatment with G-CSF did not attenuate but exacerbated neuropathic pain. In summary, G-CSF may directly activate sensory neurons and contribute to nociceptive signaling.
Collapse
Affiliation(s)
- Chun-Chang Yeh
- Department of Anesthesiology, National Defense Medical Center, Tri-Service General Hospital, Taipei 11490, Taiwan; (C.-C.Y.); (C.-P.Y.)
| | - Chih-Ping Yang
- Department of Anesthesiology, National Defense Medical Center, Tri-Service General Hospital, Taipei 11490, Taiwan; (C.-C.Y.); (C.-P.Y.)
- Department of Anesthesiology, Chi-Mei Medical Center, Tainan 71004, Taiwan
| | - Kuo-Hsing Ma
- Department of Biology and Anatomy, National Defense Medical Center, Taipei 11490, Taiwan; (K.-H.M.); (C.-S.T.)
| | - Jui-Hu Shih
- Department of Pharmacy Practice, Tri-Service General Hospital, Taipei 11490, Taiwan;
- School of Pharmacy, National Defense Medical Center, Taipei 11490, Taiwan
| | - Ching-San Tseng
- Department of Biology and Anatomy, National Defense Medical Center, Taipei 11490, Taiwan; (K.-H.M.); (C.-S.T.)
| | - Yuahn-Sieh Huang
- Department of Biology and Anatomy, National Defense Medical Center, Taipei 11490, Taiwan; (K.-H.M.); (C.-S.T.)
- Correspondence: ; Tel.: +886-87923100 (ext. 18735)
| |
Collapse
|
17
|
Guo YS, Yuan M, Han Y, Shen XY, Gao ZK, Bi X. Therapeutic Potential of Cytokines in Demyelinating Lesions After Stroke. J Mol Neurosci 2021; 71:2035-2052. [PMID: 33970426 DOI: 10.1007/s12031-021-01851-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/26/2021] [Indexed: 12/15/2022]
Abstract
White matter damage is a component of most human stroke and usually accounts for at least half of the lesion volume. Subcortical white matter stroke (WMS) accounts for 25% of all strokes and causes severe motor and cognitive dysfunction. The adult brain has a very limited ability to repair white matter damage. Pathological analysis shows that demyelination or myelin loss is the main feature of white matter injury and plays an important role in long-term sensorimotor and cognitive dysfunction. This suggests that demyelination is a major therapeutic target for ischemic stroke injury. An acute inflammatory reaction is triggered by brain ischemia, which is accompanied by cytokine production. The production of cytokines is an important factor affecting demyelination and myelin regeneration. Different cytokines have different effects on myelin damage and myelin regeneration. Exploring the role of cytokines in demyelination and remyelination after stroke and the underlying molecular mechanisms of demyelination and myelin regeneration after ischemic injury is very important for the development of rehabilitation treatment strategies. This review focuses on recent findings on the effects of cytokines on myelin damage and remyelination as well as the progress of research on the role of cytokines in ischemic stroke prognosis to provide a new treatment approach for amelioration of white matter damage after stroke.
Collapse
Affiliation(s)
- Yi-Sha Guo
- Shanghai University of Sport, Shanghai, 200438, China
| | - Mei Yuan
- Shanghai University of Sport, Shanghai, 200438, China
| | - Yu Han
- Shanghai University of Sport, Shanghai, 200438, China
| | - Xin-Ya Shen
- Shanghai University of Traditional Chinese Medicine, Shanghai, 200438, China
| | - Zhen-Kun Gao
- Shanghai University of Traditional Chinese Medicine, Shanghai, 200438, China
| | - Xia Bi
- Department of Rehabilitation Medicine, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China.
| |
Collapse
|
18
|
Aschauer-Wallner S, Leis S, Bogdahn U, Johannesen S, Couillard-Despres S, Aigner L. Granulocyte colony-stimulating factor in traumatic spinal cord injury. Drug Discov Today 2021; 26:1642-1655. [PMID: 33781952 DOI: 10.1016/j.drudis.2021.03.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 02/23/2021] [Accepted: 03/17/2021] [Indexed: 12/12/2022]
Abstract
Granulocyte colony-stimulating factor (G-CSF) is a cytokine used in pharmaceutical preparations for the treatment of chemotherapy-induced neutropenia. Evidence from experimental studies indicates that G-CSF exerts relevant activities in the central nervous system (CNS) in particular after lesions. In acute, subacute, and chronic CNS lesions, G-CSF appears to have strong anti-inflammatory, antiapoptotic, antioxidative, myelin-protective, and axon-regenerative activities. Additional effects result in the stimulation of angiogenesis and neurogenesis as well as in bone marrow stem cell mobilization to the CNS. There are emerging preclinical and clinical data indicating that G-CSF is a safe and effective drug for the treatment of acute and chronic traumatic spinal cord injury (tSCI), which we summarize in this review.
Collapse
Affiliation(s)
- Stephanie Aschauer-Wallner
- Department of Orthopedics and Traumatology, Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria; Institute of Molecular Regenerative Medicine, Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria.
| | - Stefan Leis
- Department of Neurology, Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Ulrich Bogdahn
- Velvio GmbH, Regensburg, Germany; Department of Neurology, University Hospital Regensburg, Regensburg, Germany
| | - Siw Johannesen
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany; Department of Neurology, BG Trauma Center Murnau, Murnau, Germany
| | - Sebastien Couillard-Despres
- Institute of Experimental Neuroregeneration, Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
19
|
Abri Aghdam K, Aghajani A, Ashraf Khorasani M, Soltan Sanjari M, Chaibakhsh S, Habibi A, Falavarjani KG. Intravitreal Injection Of The Granulocyte-Colony Stimulating Factor For The Treatment Of Non-Arteritic Anterior Ischemic Optic Neuropathy: A Pilot Study. Semin Ophthalmol 2021; 36:649-657. [PMID: 33689586 DOI: 10.1080/08820538.2021.1896749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Purpose To investigate the efficacy of intravitreal injection of granulocyte colony-stimulating factor (G-CSF) for the treatment of non-arteritic anterior ischemic optic neuropathy (NAION).Methods: Patients with acute NAION were enrolled in this prospective interventional case series. They received an intravitreal injection of 60 micrograms in 0.1 ml of G-CSF within 2 weeks of the onset of the disease. Visual acuity, visual field, intraocular pressure (IOP), corneal endothelial cell density, and peripapillary retinal nerve fiber layer (RNFL) thickness were recorded before injections and 1 week, 1 month, 3 months, 6 months, and one year after the injections. Full-field electroretinography (ERG) was obtained at the baseline, 1 month, and 12 months post- injections.Results: Fourteen eyes of 14 patients entered the study. Best-corrected visual acuity (BCVA) significantly improved in the first month following injections (p = .007), decreased subsequently, and the final BCVA showed no significant improvement (p = .278) compared to the baseline measurements. A significant decrease in RNFL thickness was observed in all quadrants compared to the baseline measurements. Also, no improvement in the visual field parameters was observed. From the toxicity aspect, no significant changes in the corneal endothelial cell density, IOP, and ERG recordings were observed.Conclusion: Intravitreal injection of G-CSF seems to be safe. The effect may last for one month and then decline.
Collapse
Affiliation(s)
- Kaveh Abri Aghdam
- , Eye Research Center, the Five Senses Institute, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Aghajani
- , Eye Research Center, the Five Senses Institute, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Ashraf Khorasani
- , Eye Research Center, the Five Senses Institute, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Mostafa Soltan Sanjari
- , Eye Research Center, the Five Senses Institute, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Samira Chaibakhsh
- , Eye Research Center, the Five Senses Institute, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Abbas Habibi
- , Eye Research Center, the Five Senses Institute, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
20
|
Dumbuya JS, Chen L, Wu JY, Wang B. The role of G-CSF neuroprotective effects in neonatal hypoxic-ischemic encephalopathy (HIE): current status. J Neuroinflammation 2021; 18:55. [PMID: 33612099 PMCID: PMC7897393 DOI: 10.1186/s12974-021-02084-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/14/2021] [Indexed: 12/23/2022] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is an important cause of permanent damage to central nervous system (CNS) that may result in neonatal death or manifest later as mental retardation, epilepsy, cerebral palsy, or developmental delay. The primary cause of this condition is systemic hypoxemia and/or reduced cerebral blood flow with long-lasting neurological disabilities and neurodevelopmental impairment in neonates. About 20 to 25% of infants with HIE die in the neonatal period, and 25-30% of survivors are left with permanent neurodevelopmental abnormalities. The mechanisms of hypoxia-ischemia (HI) include activation and/or stimulation of myriad of cascades such as increased excitotoxicity, oxidative stress, N-methyl-D-aspartic acid (NMDA) receptor hyperexcitability, mitochondrial collapse, inflammation, cell swelling, impaired maturation, and loss of trophic support. Different therapeutic modalities have been implicated in managing neonatal HIE, though translation of most of these regimens into clinical practices is still limited. Therapeutic hypothermia, for instance, is the most widely used standard treatment in neonates with HIE as studies have shown that it can inhibit many steps in the excito-oxidative cascade including secondary energy failure, increases in brain lactic acid, glutamate, and nitric oxide concentration. Granulocyte-colony stimulating factor (G-CSF) is a glycoprotein that has been implicated in stimulation of cell survival, proliferation, and function of neutrophil precursors and mature neutrophils. Extensive studies both in vivo and ex vivo have shown the neuroprotective effect of G-CSF in neurodegenerative diseases and neonatal brain damage via inhibition of apoptosis and inflammation. Yet, there are still few experimentation models of neonatal HIE and G-CSF's effectiveness, and extrapolation of adult stroke models is challenging because of the evolving brain. Here, we review current studies and/or researches of G-CSF's crucial role in regulating these cytokines and apoptotic mediators triggered following neonatal brain injury, as well as driving neurogenesis and angiogenesis post-HI insults.
Collapse
Affiliation(s)
- John Sieh Dumbuya
- Department of Pediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Lu Chen
- Department of Pediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Jang-Yen Wu
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
| | - Bin Wang
- Department of Pediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282, People's Republic of China.
| |
Collapse
|
21
|
Song S, Kong X, Borlongan C, Sava V, Sanchez-Ramos J. Granulocyte Colony-Stimulating Factor Enhances Brain Repair Following Traumatic Brain Injury Without Requiring Activation of Cannabinoid Receptors. Cannabis Cannabinoid Res 2021; 6:48-57. [PMID: 33614952 PMCID: PMC7891202 DOI: 10.1089/can.2019.0090] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Introduction: Treatment of traumatic brain injury (TBI) with granulocyte colony-stimulating factor (G-CSF) has been shown to enhance brain repair by direct neurotrophic actions on neural cells and by modulating the inflammatory response. Administration of cannabinoids after TBI has also been reported to enhance brain repair by similar mechanisms. Objectives: The primary objective of this study was to test the hypothesis that G-CSF mediates brain repair by interacting with the endocannabinoid system. Methods and Results: (i) Mice that underwent controlled cortical impact (CCI) were treated with G-CSF for 3 days either alone or in the presence of selective cannabinoid receptor 1 (CB1-R) or cannabinoid receptor 2 (CB2-R) agonists and antagonists. The trauma resulted in decreased expression of CB1-R and increased expression of CB2-R in the cortex, striatum, and hippocampus. Cortical and striatal levels of the major endocannabinoid ligand, 2-arachidonoyl-glycerol, were also increased by the CCI. Administration of the hematopoietic cytokine, G-CSF, following TBI, resulted in mitigation or reversal of trauma-induced CB1-R downregulation and CB2-R upregulation in the three brain regions. Treatment with CB1-R agonist (WIN55) or CB2-R agonist (HU308) mimicked the effects of G-CSF. (ii) Pharmacological blockade of CB1-R or CB2-R was not effective in preventing G-CSF's mitigation or reversal of trauma-induced alterations in these receptors. Conclusions: These results suggest that cellular and molecular mechanisms that mediate subacute effects of G-CSF do not depend on activation of CB1 or CB2 receptors. Failure of selective CB receptor antagonists to prevent the effects of G-CSF in this model has to be accepted with caution. CB receptor antagonists can interact with other CB and non-CB receptors. Investigation of the role of CB receptors in this TBI model will require studies with CB1-R and in CB2-R knockout mice to avoid nonspecific interaction of CB receptor agents with other receptors.
Collapse
MESH Headings
- Animals
- Arachidonic Acids/metabolism
- Arachidonic Acids/physiology
- Brain/metabolism
- Brain Injuries, Traumatic/drug therapy
- Brain Injuries, Traumatic/etiology
- Brain Injuries, Traumatic/metabolism
- Cannabinoid Receptor Agonists/pharmacology
- Cannabinoid Receptor Agonists/therapeutic use
- Cannabinoid Receptor Antagonists/pharmacology
- Cannabinoid Receptor Antagonists/therapeutic use
- Disease Models, Animal
- Endocannabinoids/metabolism
- Endocannabinoids/physiology
- Glycerides/metabolism
- Glycerides/physiology
- Granulocyte Colony-Stimulating Factor/pharmacology
- Granulocyte Colony-Stimulating Factor/therapeutic use
- Male
- Mice, Inbred C57BL
- Mitogen-Activated Protein Kinases/metabolism
- Receptor, Cannabinoid, CB1/genetics
- Receptor, Cannabinoid, CB1/metabolism
- Receptor, Cannabinoid, CB2/genetics
- Receptor, Cannabinoid, CB2/metabolism
- Receptors, Cannabinoid/genetics
- Receptors, Cannabinoid/metabolism
- Signal Transduction/drug effects
- Mice
Collapse
Affiliation(s)
- Shijie Song
- James Haley VA Medical Center, Tampa, Florida, USA
- Department of Neurology and University of South Florida, Tampa, Florida, USA
| | | | - Cesar Borlongan
- James Haley VA Medical Center, Tampa, Florida, USA
- Department of Neurosurgery, University of South Florida, Tampa, Florida, USA
| | - Vasyl Sava
- James Haley VA Medical Center, Tampa, Florida, USA
- Department of Neurology and University of South Florida, Tampa, Florida, USA
| | - Juan Sanchez-Ramos
- Department of Neurology and University of South Florida, Tampa, Florida, USA
| |
Collapse
|
22
|
Neuroprotection through G-CSF: recent advances and future viewpoints. Pharmacol Rep 2021; 73:372-385. [PMID: 33389706 DOI: 10.1007/s43440-020-00201-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 11/18/2020] [Accepted: 11/21/2020] [Indexed: 12/14/2022]
Abstract
Granulocyte-colony stimulating factor (G-CSF), a member of the cytokine family of hematopoietic growth factors, is 19.6 kDa glycoprotein which is responsible for the proliferation, maturation, differentiation, and survival of neutrophilic granulocyte lineage. Apart from its proven clinical application to treat chemotherapy-associated neutropenia, recent pre-clinical studies have highlighted the neuroprotective roles of G-CSF i.e., mobilization of haemopoietic stem cells, anti-apoptotic, neuronal differentiation, angiogenesis and anti-inflammatory in animal models of neurological disorders. G-CSF is expressed by numerous cell types including neuronal, immune and endothelial cells. G-CSF is released in autocrine manner and binds to its receptor G-CSF-R which further activates numerous signaling transduction pathways including PI3K/AKT, JAK/STAT and MAP kinase, and thereby promote neuronal survival, proliferation, differentiation, mobilization of hematopoietic stem and progenitor cells. The expression of G-CSF receptors (G-CSF-R) in the different brain regions and their upregulation in response to neuronal insult indicates the autocrine protective signaling mechanism of G-CSF by inhibition of apoptosis, inflammation, and stimulation of neurogenesis. These observed neuroprotective effects of G-CSF makes it an attractive target to mitigate neurodegeneration associated with neurological disorders. The objective of the review is to highlight and summarize recent updates on G-CSF as a therapeutically versatile neuroprotective agent along with mechanisms of action as well as possible clinical applications in neurodegenerative disorders including AD, PD and HD.
Collapse
|
23
|
Granulocyte-Colony Stimulating Factor Reduces Cocaine-Seeking and Downregulates Glutamatergic Synaptic Proteins in Medial Prefrontal Cortex. J Neurosci 2020; 41:1553-1565. [PMID: 33361463 DOI: 10.1523/jneurosci.1452-20.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 12/08/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
Psychostimulant use disorder is a major public health issue, and despite the scope of the problem there are currently no Food and Drug Administration (FDA)-approved treatments. There would be tremendous utility in development of a treatment that could help patients both achieve and maintain abstinence. Previous work from our group has identified granulocyte-colony stimulating factor (G-CSF) as a neuroactive cytokine that alters behavioral response to cocaine, increases synaptic dopamine release, and enhances cognitive flexibility. Here, we investigate the role of G-CSF in affecting extinction and reinstatement of cocaine-seeking and perform detailed characterization of its proteomic effects in multiple limbic substructures. Male Sprague Dawley rats were injected with PBS or G-CSF during (1) extinction or (2) abstinence from cocaine self-administration, and drug seeking behavior was measured. Quantitative assessment of changes in the proteomic landscape in the nucleus accumbens (NAc) and medial prefrontal cortex (mPFC) were performed via data-independent acquisition (DIA) mass spectrometry analysis. Administration of G-CSF during extinction accelerated the rate of extinction, and administration during abstinence attenuated cue-induced cocaine-seeking. Analysis of global protein expression demonstrated that G-CSF regulated proteins primarily in mPFC that are critical to glutamate signaling and synapse maintenance. Taken together, these findings support G-CSF as a viable translational research target with the potential to reduce drug craving or seeking behaviors. Importantly, recombinant G-CSF exists as an FDA-approved medication which may facilitate rapid clinical translation. Additionally, using cutting-edge multiregion discovery proteomics analyses, these studies identify a novel mechanism underlying G-CSF effects on behavioral plasticity.SIGNIFICANCE STATEMENT Pharmacological treatments for psychostimulant use disorder are desperately needed, especially given the disease's chronic, relapsing nature. However, there are currently no Food and Drug Administration (FDA)-approved pharmacotherapies. Emerging evidence suggests that targeting the immune system may be a viable translational research strategy; preclinical studies have found that the neuroactive cytokine granulocyte-colony stimulating factor (G-CSF) alters cocaine reward and reinforcement and can enhance cognitive flexibility. Given this basis of evidence we studied the effects of G-CSF treatment on extinction and reinstatement of cocaine seeking. We find that administration of G-CSF accelerates extinction and reduces cue-induced drug seeking after cocaine self-administration. In addition, G-CSF leads to downregulation of synaptic glutamatergic proteins in medial prefrontal cortex (mPFC), suggesting that G-CSF influences drug seeking via glutamatergic mechanisms.
Collapse
|
24
|
Menzie-Suderam JM, Modi J, Xu H, Bent A, Trujillo P, Medley K, Jimenez E, Shen J, Marshall M, Tao R, Prentice H, Wu JY. Granulocyte-colony stimulating factor gene therapy as a novel therapeutics for stroke in a mouse model. J Biomed Sci 2020; 27:99. [PMID: 33126859 PMCID: PMC7596942 DOI: 10.1186/s12929-020-00692-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/15/2020] [Indexed: 12/11/2022] Open
Abstract
Background Global ischemia is the resulting effect of a cardiopulmonary arrest (CPA). Presently there is no effective treatment to address neurological deficits in patients who survived a CPA. Granulocyte-colony stimulating factor is a growth factor (G-CSF) with a plethora of beneficial effects, including neuroprotection. Clinical application of human G-CSF (hG-CSF) is limited due to its plasma half-life of 4 h. Therefore, novel approaches need to be investigated that would (1) enable prolonged manifestation of hG-CSF and (2) demonstrate G-CSF efficacy from studying the underlying protective mechanisms of hG-CSF. In our previous work, we used the self-complementary adeno-associated virus (stereotype2: scAAV2) as a vector to transfect the hG-CSF gene into the global ischemic brain of a mouse. As an extension of that work, we now seek to elucidate the protective mechanisms of hG-CSF gene therapy against endoplasmic reticulum induced stress, mitochondrial dynamics and autophagy in global ischemia. Method A single drop of either AAV-CMV-hG-CSF or AAV-CMV-GFP was dropped into the conjunctival sac of the Swiss Webster mouse’s left eye, 30–60 min after bilateral common artery occlusion (BCAO). The efficacy of the expressed hG-CSF gene product was analyzed by monitoring the expression levels of endoplasmic reticulum stress (ER), mitochondrial dynamics and autophagic proteins over 4- and 7-days post-BCAO in vulnerable brain regions including the striatum, overlying cortex (frontal brain regions) and the hippocampus (middle brain regions). Statistical analysis was performed using mostly One-Way Analysis of variance (ANOVA), except for behavioral analysis, which used Repeated Measures Two-Way ANOVA, post hoc analysis was performed using the Tukey test. Results Several biomarkers that facilitated cellular death, including CHOP and GRP78 (ER stress) DRP1 (mitochondrial dynamics) and Beclin 1, p62 and LC3-ll (autophagy) were significantly downregulated by hG-CSF gene transfer. hG-CSF gene therapy also significantly upregulated antiapoptotic Bcl2 while downregulating pro-apoptotic Bax. The beneficial effects of hG-CSF gene therapy resulted in an overall improvement in functional behavior. Conclusion Taken together, this study has substantiated the approach of sustaining the protein expression of hG-CSF by eye drop administration of the hG-CSF gene. In addition, the study has validated the efficacy of using hG-CSF gene therapy against endoplasmic reticulum induced stress, mitochondrial dynamics and autophagy in global ischemia.
Collapse
Affiliation(s)
- Janet M Menzie-Suderam
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, 33431, USA.,Program in Integrative Biology, Florida Atlantic University, Boca Raton, FL, 33431, USA
| | - Jigar Modi
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, 33431, USA.,Complex Systems and Brain Sciences, Florida Atlantic University, Boca Raton, FL, USA
| | - Hongyaun Xu
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, 33431, USA
| | - Andrew Bent
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, 33431, USA
| | - Paula Trujillo
- Program in Integrative Biology, Florida Atlantic University, Boca Raton, FL, 33431, USA
| | - Kristen Medley
- College of Medicine, New York University, New York, NY, 10003, USA
| | - Eugenia Jimenez
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, 33431, USA
| | - Jessica Shen
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, 33431, USA
| | | | - Rui Tao
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, 33431, USA.
| | - Howard Prentice
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, 33431, USA. .,Program in Integrative Biology, Florida Atlantic University, Boca Raton, FL, 33431, USA. .,Complex Systems and Brain Sciences, Florida Atlantic University, Boca Raton, FL, USA.
| | - Jang-Yen Wu
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, 33431, USA. .,Program in Integrative Biology, Florida Atlantic University, Boca Raton, FL, 33431, USA. .,Complex Systems and Brain Sciences, Florida Atlantic University, Boca Raton, FL, USA.
| |
Collapse
|
25
|
Matsuyama M, Søraas A, Yu S, Kim K, Stavrou EX, Caimi PF, Wald D, deLima M, Dahl JA, Horvath S, Matsuyama S. Analysis of epigenetic aging in vivo and in vitro: Factors controlling the speed and direction. Exp Biol Med (Maywood) 2020; 245:1543-1551. [PMID: 32762265 DOI: 10.1177/1535370220947015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
IMPACT STATEMENT Aging is associated with DNA methylation (DNAm) changes. Recent advancement of the whole-genome DNAm analysis technology allowed scientists to develop DNAm-based age estimators. A majority of these estimators use DNAm data from a single tissue type such as blood. In 2013, a multi-tissue age estimator using DNAm pattern of 353 CpGs was developed by Steve Horvath. This estimator was named "epigenetic clock", and the improved version using DNAm pattern of 391 CpGs was developed in 2018. The estimated age by epigenetic clock is named DNAmAge. DNAmAge can be used as a biomarker of aging predicting the risk of age-associated diseases and mortality. Although the DNAm-based age estimators were developed, the mechanism of epigenetic aging is still enigmatic. The biological significance of epigenetic aging is not well understood, either. This minireview discusses the current understanding of the mechanism of epigenetic aging and the future direction of aging research.
Collapse
Affiliation(s)
- Mieko Matsuyama
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Case Western Reserve University and University Hospitals, Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| | - Arne Søraas
- Department of Microbiology, Oslo University Hospital, Case Comprehensive Cancer Center, Oslo 0372, Norway
| | - Sarah Yu
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Case Western Reserve University and University Hospitals, Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| | - Kyuhyeon Kim
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Case Western Reserve University and University Hospitals, Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| | - Evi X Stavrou
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Case Western Reserve University and University Hospitals, Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| | - Paolo F Caimi
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Case Western Reserve University and University Hospitals, Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| | - David Wald
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Case Western Reserve University and University Hospitals, Case Comprehensive Cancer Center, Cleveland, OH 44106, USA.,Department of Microbiology, Oslo University Hospital, Case Comprehensive Cancer Center, Oslo 0372, Norway
| | - Marcos deLima
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Case Western Reserve University and University Hospitals, Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| | - John A Dahl
- Department of Microbiology, Oslo University Hospital, Case Comprehensive Cancer Center, Oslo 0372, Norway
| | - Steve Horvath
- Department of Pathology, School of Medicine, Case Western Reserve University and University Hospitals, Case Comprehensive Cancer Center, Cleveland, OH 44106, USA.,Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Shigemi Matsuyama
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, Case Western Reserve University and University Hospitals, Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| |
Collapse
|
26
|
Rasouli B, Ghahari L, Safari M, Shahroozian E, Naeimi S. Combination therapy of the granulocyte colony stimulating factor and intravenous lipid emulsion protect the hippocampus after global ischemia in rat: focusing on CA1 region. Metab Brain Dis 2020; 35:991-997. [PMID: 32458336 DOI: 10.1007/s11011-020-00579-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 05/06/2020] [Indexed: 10/24/2022]
Abstract
Brain stroke is one of the causes of human death and disability worldwide. Global ischemia results in the accumulation of free radicals in the neurons. It leads to histologically brain damage. The CA1 region of the hippocampus is a sensitive area for free radicals. This study investigated the combined therapy of the Granulocyte colony stimulating factor (G-CSF) and the Intravenous lipid emulsion (ILE). These neuroprotective agents play a role in the regeneration of neurons. They improve the learning ability and memory in rats induced global ischemia. We divided 35 rats into five groups. The groups were sham group, ischemia group, G-CSF group, ILE group, and G-CSF plus ILE group. Ischemia was induced by occlusion of the bilateral common carotid about 10 min. The drugs applied on days 1, 3 and 7. The treated groups received subcutaneous injection of 20 μg/kg G-CSF and intravenous injection of 5 ml/kg ILE. After two weeks, the memory and learning ability of the rats was evaluated by the shuttle box. Hematoxylin and Eosin and Nissl and TUNEL stainings were used to determine the necrosis, normal and apoptotic cells. The combined therapy increased normal cells compared to the ischemia group. They decreased the number of necrotic and apoptosis cells in other groups. The combined group improved the passive avoidance test compared to the other groups. The combination therapy of G-CSF plus ILE is more effective than each alone.
Collapse
Affiliation(s)
- Babak Rasouli
- Department of Anatomy, Medical School, AJA University of Medical Sciences, Tehran, Iran
- Department of Basic Sciences, Faculty of Veterinary Medicine, Semnan University, Semnan, Iran
| | - Laya Ghahari
- Department of Anatomy, Medical School, AJA University of Medical Sciences, Tehran, Iran.
| | - Manouchehr Safari
- Research Center of Nervous System Stem Cells, Semnan University of Medical Science, Semnan, Iran
| | - Ebrahim Shahroozian
- Department of Basic Sciences, Faculty of Veterinary Medicine, Semnan University, Semnan, Iran
| | - Saeideh Naeimi
- Department of Basic Sciences, Faculty of Veterinary Medicine, Semnan University, Semnan, Iran
| |
Collapse
|
27
|
Hong Y, Yu Q, Kong Z, Wang M, Zhang R, Li Y, Liu Y. Exogenous endothelial progenitor cells reached the deficient region of acute cerebral ischemia rats to improve functional recovery via Bcl-2. Cardiovasc Diagn Ther 2020; 10:695-704. [PMID: 32968626 PMCID: PMC7487376 DOI: 10.21037/cdt-20-329] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 07/06/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND As discovered in our previous study, autologous endothelial progenitor cells (EPCs) protect against acute focal ischemia rat via the promotion of angiogenesis. However, it is unknown whether the EPCs that reached the deficient region were transplanted ones or the products of other auto-conversion cells they had promoted. This study aimed to gather direct evidence for determining if exogenous transplanted EPCs directly participate in angiogenesis in ischemic areas and attempted to clarify the related mechanism. METHODS First, EPCs were extracted in vitro from male rats, which were characterized by uptake of fluorescently labeled acetylated low-density lipoprotein (ac-LDL) intake and Ulex europaeus agglutinin (UEA-1) and subsequently introduced to middle cerebral artery occlusion (MCAO) female rats for 7 days after ischemia surgery. The EPC-treated animals received approximately 1×106 cells, while the control animals received phosphate buffered saline (PBS). The animals behavior function recovery were by a rotarod (TOR) test, while infarct volume was assessed by brain magnetic resonance imaging (MRI). CD31 antibody was used to determine the presence of EPCs in the ischemic zone, and sex-determining region Y (SRY) gene in-situ hybridization (ISH) traced the EPC process. In addition, immunohistochemistry and Western blot were used to assess B-cell lymphoma 2 (Bcl-2) expression in the ischemic brain. RESULTS Behavior tests and MRI of all ischemic stroke groups on postoperative day 14 indicated that EPCs were more effective in behavior function recovery and reducing infarct volume and gliosis status than the control group. Cluster of differentiation (CD31) immunofluorescent staining and SRY gene ISH demonstrated that EPCs yielded a better outcome in both angiogenesis and exogenous cell homing status. We also observed increased Bcl-2 distribution and higher plasma Bcl-2 levels in the EPC-treated group compared to the control group. CONCLUSIONS Our results provide direct evidence that exogenous EPCs can participate in angiogenesis to improve neurological outcome and revascularization directly after stroke, with Bcl-2 playing an important role in this process.
Collapse
Affiliation(s)
- Yan Hong
- Department of Pediatrics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Qing Yu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhaohong Kong
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Meiyao Wang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Renwei Zhang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yan Li
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yumin Liu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
28
|
Dumbuya JS, Chen L, Shu SY, Ma L, Luo W, Li F, Wu JY, Wang B. G-CSF attenuates neuroinflammation and neuronal apoptosis via the mTOR/p70SK6 signaling pathway in neonatal Hypoxia-Ischemia rat model. Brain Res 2020; 1739:146817. [PMID: 32246916 DOI: 10.1016/j.brainres.2020.146817] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 03/19/2020] [Accepted: 03/31/2020] [Indexed: 11/16/2022]
Abstract
BACKGROUND Hypoxic-ischemic encephalopathy (HIE) is an important cause of permanent damage to the central nervous system, associated with long-lasting neurological disabilities and neurodevelopmental impairment in neonates. Granulocyte-colony stimulating factor (G-CSF) has been shown to have neuroprotective activity in a variety of experimental brain injury models and G-CSF is a standard treatment in chemotherapeutic-induced neutropenia. The underlying mechanisms are still unclear. The mTOR (mammalian target of rapamycin) signaling pathway is a master regulator of cell growth and proliferation in the nervous system. However, the effects of G-CSF treatment on the mTOR signaling pathway have not been elucidated in neonates with hypoxic-ischemic (HI) brain injury. Our study investigated the neuroprotective effect of G-CSF on neonates with hypoxic-ischemic (HI) brain injury and the possible mechanism involving the mTOR/p70S6K pathway. METHODS Sprague-Dawley rat pups at postnatal day 7 (P7) were subjected to right unilateral carotid artery ligation followed by hypoxic (8% oxygen and balanced nitrogen) exposure for 2.5 h or sham surgery. Pups received normal saline, G-CSF, G-CSF combined with rapamycin or ethanol (vehicle for rapamycin) intraperitoneally. On postnatal day 9 (P9), TTC staining for infarct volume, and Nissl and TUNEL staining for neuronal cell injury were conducted. Activation of mTOR/p70S6K pathway, cleaved caspase-3 (CC3), Bax and Bcl-2 and cytokine expression levels were determined by western blotting. RESULTS The G-CSF treated group was associated with significantly reduced infarction volume and decreased TUNEL positive neuronal cells compared to the HI group treated with saline. The expression levels of TNF-α and IL-1ß were significantly decreased in the G-CSF treated group, while IL-10 expression level was increased. The relative immunoreactivity of p-mTOR and p-p70S6K was significantly reduced in the HI group compared to sham. The HI group treated with G-CSF showed significant upregulated protein expression for p-mTOR and p-p70S6K levels compared to the HI group treated with saline. Furthermore, G-CSF treatment increased Bcl-2 expression levels and decreased CC3 and Bax expression levels in the ipsilateral hemispheres of the HI brain. The effects induced by G-CSF were all reversed by rapamycin. CONCLUSION Treatment with G-CSF decreases inflammatory mediators and apoptotic factors, attenuating neuroinflammation and neuronal apoptosis via the mTOR/p70S6K signalling pathway, which represents a potential target for treating HI induced brain damage in neonatal HIE.
Collapse
Affiliation(s)
- John Sieh Dumbuya
- Department of Pediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou 510282 PR China
| | - Lu Chen
- Department of Pediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou 510282 PR China
| | - Si Yun Shu
- Department of Pediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou 510282 PR China
| | - Lin Ma
- Department of Radiotherapy, Chinese PLA General Hospital, Beijing 100853 PR China
| | - Wei Luo
- Department of Pediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou 510282 PR China
| | - Fei Li
- Department of Pediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou 510282 PR China
| | - Jang-Yen Wu
- Department of Biochemical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States.
| | - Bin Wang
- Department of Pediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou 510282 PR China.
| |
Collapse
|
29
|
Salamone P, Fuda G, Casale F, Marrali G, Lunetta C, Caponnetto C, Mazzini L, La Bella V, Mandrioli J, Simone IL, Moglia C, Calvo A, Tarella C, Chio A. G-CSF (filgrastim) treatment for amyotrophic lateral sclerosis: protocol for a phase II randomised, double-blind, placebo-controlled, parallel group, multicentre clinical study (STEMALS-II trial). BMJ Open 2020; 10:e034049. [PMID: 32209625 PMCID: PMC7202695 DOI: 10.1136/bmjopen-2019-034049] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION Amyotrophic lateral sclerosis (ALS) is a fatal progressive neurological disorder characterised by a selective degeneration of motor neurons (MNs). Stem cell transplantation is considered as a promising strategy in neurological disorders therapy and the possibility of inducing bone marrow cells (BMCs) to circulate in the peripheral blood is suggested to investigate stem cells migration in degenerated ALS nerve tissues where potentially repair MN damage. Granulocyte-colony stimulating factor (G-CSF) is a growth factor which stimulates haematopoietic progenitor cells, mobilises BMCs into injured brain and it is itself a neurotrophic factor for MN. G-CSF safety in humans has been demonstrated and many observations suggest that it may affect neural cells. Therefore, we decided to use G-CSF to mobilise BMCs into the peripheral circulation in patients with ALS, planning a clinical trial to evaluate the effect of G-CSF administration in ALS patients compared with placebo. METHODS AND ANALYSIS STEMALS-II is a phase II multicentre, randomised double-blind, placebo-controlled, parallel group clinical trial on G-CSF (filgrastim) and mannitol in ALS patients. Specifically, we investigate safety, tolerability and efficacy of four repeated courses of intravenous G-CSF and mannitol administered in 76 ALS patients in comparison with placebo (indistinguishable glucose solution 5%). We determine increase of G-CSF levels in serum and cerebrospinal fluid as CD34+ cells and leucocyte count after treatment; reduction in ALS Functional Rating Scale-Revised Score, forced vital capacity, Scale for Testing Muscle Strength Score and quality of life; the adverse events/reactions during the treatment; changes in neuroinflammation biomarkers before and after treatment. ETHICS AND DISSEMINATION The study protocol was approved by the Ethics Committee of Azienda Ospedaliera Universitaria 'Città della Salute e della Scienza', Torino, Italy. Results will be presented during scientific symposia or published in scientific journals. TRIAL REGISTRATION NUMBER Eudract 2014-002228-28.
Collapse
Affiliation(s)
- Paolina Salamone
- 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Torino, Piemonte, Italy
| | - Giuseppe Fuda
- 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Torino, Piemonte, Italy
| | - Federico Casale
- 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Torino, Piemonte, Italy
| | - Giuseppe Marrali
- 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Torino, Piemonte, Italy
| | - Christian Lunetta
- NEuroMuscular Omnicentre (NEMO), Fondazione Serena Onlus, Milan, Italy
| | - Claudia Caponnetto
- Neurological Clinic, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Letizia Mazzini
- Department of Neurology, Maggiore della Carità Hospital, University of Piemonte Orientale, Novara, Italy
| | - Vincenzo La Bella
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Sicilia, Italy
| | - Jessica Mandrioli
- Department of Neuroscience, Azienda Ospedaliera Universitaria Modena, St. Agostino-Estense Hospital, Modena, Italy
| | - Isabella Laura Simone
- Neurology Unit, Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari, Bari, Puglia, Italy
| | - Cristina Moglia
- 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Torino, Piemonte, Italy
- ALS Center, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, Torino, Piemonte, Italy
| | - Andrea Calvo
- 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Torino, Piemonte, Italy
- ALS Center, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, Torino, Piemonte, Italy
| | - Corrado Tarella
- Oncohematology Division, IEO European Institute of Oncology, IRCCS, University of Milan, Milano, Lombardia, Italy
| | - Adriano Chio
- 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Torino, Piemonte, Italy
- ALS Center, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, Torino, Piemonte, Italy
| |
Collapse
|
30
|
Jia J, Li C, Zhang T, Sun J, Peng S, Xie Q, Huang Y, Yi L. CeO 2@PAA-LXW7 Attenuates LPS-Induced Inflammation in BV2 Microglia. Cell Mol Neurobiol 2019; 39:1125-1137. [PMID: 31256326 PMCID: PMC11452219 DOI: 10.1007/s10571-019-00707-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 06/13/2019] [Indexed: 12/14/2022]
Abstract
Microglia are the inherent immune effector cells in the central nervous system (CNS), are activated rapidly when the CNS is stimulated by ischaemia, infection, injury, etc. and participate in and aggravate the development of inflammatory reactions in the CNS. During the process of microglial activation, inflammatory factors such as TNF-α and IL-1β and an abundance of reactive oxygen species (ROS)/reactive nitrogen species (RNS), are released by damaged nerve cells. LXW7 is a small molecule peptide and specifically binds with integrin αvβ3. Cerium oxide nanoparticles (nanoceria) are strong free radical scavengers and are widely used in many studies. In this research, a model of inflammation was established using lipopolysaccharide (LPS) to induce BV2 microglia activation, and the effects of CeO2@PAA (synthetic nanoscale cerium oxide particles), LXW7 and CeO2@PAA-LXW7 were evaluated. We detected the expression level of inflammatory factors, the release of NO in BV2 cells and the generation of intracellular ROS. The expression levels of focal adhesion kinase (FAK) and signal transducer and activator of transcription 3 (STAT3) and their phosphorylated proteins were detected in BV2 microglia. We found that CeO2@PAA, LXW7 and CeO2@PAA-LXW7 all effectively inhibited the activation of BV2 microglia, reduced the production of cytokines and the release of NO and reduced the production of intracellular ROS. The three treatments all inhibited the phosphorylation of FAK and STAT3 in BV2 microglia. Regarding these effects, CeO2@PAA-LXW7 was more effective than the other two monotherapies. Our data indicate that CeO2@PAA, LXW7 and CeO2@PAA-LXW7 can exert a neuroprotective function by inhibiting the inflammatory response of LPS-induced BV2 microglia. LXW7 may inhibit the activation of FAK and STAT3 signals in combination with integrin αvβ3 to restrain neuroinflammation and the antioxidative stress effect of cerium oxide; hence, CeO2@PAA-LXW7 can exert a more robust anti-inflammatory and neuroprotective effect via synergistically suppressing the ability of LXW7 to influence the integrin pathway and the free radical-scavenging ability of CeO2@PAA.
Collapse
Affiliation(s)
- Jingjing Jia
- Department of Neurology, Peking University First Hospital, No.8 Xishiku Street, Xicheng District, Beijing, 100034, China
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, 518036, Guangdong Province, China
| | - Changyan Li
- College of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot, Inner Mongolia Province, China
| | - Ting Zhang
- Department of Phoenix International Medical Center, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong Province, China
| | - Jingjing Sun
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, 518036, Guangdong Province, China
| | - Sijia Peng
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, 518036, Guangdong Province, China
| | - Qizhi Xie
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, 518036, Guangdong Province, China
| | - Yining Huang
- Department of Neurology, Peking University First Hospital, No.8 Xishiku Street, Xicheng District, Beijing, 100034, China.
| | - Li Yi
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, 518036, Guangdong Province, China.
| |
Collapse
|
31
|
Tsang JKW, Liu J, Lo ACY. Vascular and Neuronal Protection in the Developing Retina: Potential Therapeutic Targets for Retinopathy of Prematurity. Int J Mol Sci 2019; 20:E4321. [PMID: 31484463 PMCID: PMC6747312 DOI: 10.3390/ijms20174321] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/21/2019] [Accepted: 08/29/2019] [Indexed: 12/13/2022] Open
Abstract
Retinopathy of prematurity (ROP) is a common retinal disease in preterm babies. To prolong the lives of preterm babies, high oxygen is provided to mimic the oxygen level in the intrauterine environment for postnatal organ development. However, hyperoxia-hypoxia induced pathological events occur when babies return to room air, leading to ROP with neuronal degeneration and vascular abnormality that affects retinal functions. With advances in neonatal intensive care, it is no longer uncommon for increased survival of very-low-birth-weight preterm infants, which, therefore, increased the incidence of ROP. ROP is now a major cause of preventable childhood blindness worldwide. Current proven treatment for ROP is limited to invasive retinal ablation, inherently destructive to the retina. The lack of pharmacological treatment for ROP creates a great need for effective and safe therapies in these developing infants. Therefore, it is essential to identify potential therapeutic agents that may have positive ROP outcomes, especially in preserving retinal functions. This review gives an overview of various agents in their efficacy in reducing retinal damages in cell culture tests, animal experiments and clinical studies. New perspectives along the neuroprotective pathways in the developing retina are also reviewed.
Collapse
Affiliation(s)
- Jessica K W Tsang
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jin Liu
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Amy C Y Lo
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
32
|
Antiapoptotic Effect of Granulocyte-Colony Stimulating Factor After Peripheral Nerve Trauma. World Neurosurg 2019; 129:e6-e15. [DOI: 10.1016/j.wneu.2019.04.059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 04/05/2019] [Accepted: 04/06/2019] [Indexed: 11/18/2022]
|
33
|
Lipták N, Bősze Z, Hiripi L. GFP transgenic animals in biomedical research: a review of potential disadvantages. Physiol Res 2019; 68:525-530. [PMID: 31342754 DOI: 10.33549/physiolres.934227] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Green Fluorescent protein (GFP) transgenic animals are accepted tools for studying various physiological processes, including organ development and cell migration. However, several in vivo studies claimed that GFP may impair transgenic animals' health. Glomerulosclerosis was observed in transgenic mice and rabbits with ubiquitous reporter protein expression. Heart-specific GFP expression evoked dilated cardiomyopathy and altered cardiac function in transgenic mouse and zebrafish lines, respectively. Moreover, growth retardation and increased axon swelling were observed in GFP and yellow fluorescent protein (YFP) transgenic mice, respectively. This review will focus on the potential drawbacks of the applications of GFP transgenic animals in biomedical research.
Collapse
Affiliation(s)
- N Lipták
- NARIC-Agricultural Biotechnology Institute, Animal Biotechnology Department, Gödöllő, Hungary.
| | | | | |
Collapse
|
34
|
Li X, Lin S, Chen X, Huang W, Li Q, Zhang H, Chen X, Yang S, Jin K, Shao B. The Prognostic Value of Serum Cytokines in Patients with Acute Ischemic Stroke. Aging Dis 2019; 10:544-556. [PMID: 31164999 PMCID: PMC6538221 DOI: 10.14336/ad.2018.0820] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 08/20/2018] [Indexed: 12/12/2022] Open
Abstract
The inflammatory response is an unavoidable process and contributes to the destruction of cerebral tissue during the acute ischemic stroke (AIS) phase and has not been addressed fully to date. Insightful understanding of correlation of inflammatory mediators and stroke outcome may provide new biomarkers or therapeutic approaches for ischemic stroke. Here, we prospectively recruited 180 first-ever AIS patients within 72 hrs after stroke onset. We used the National Institutes of Health Stroke Scale (NIHSS) to quantify stroke severity and modified Rankin scale (mRS) to assess the 3-month outcome for AIS patients. Initially, we screened 35 cytokines, chemokines, and growth factors in sera from 75 AIS patients and control subjects. Cytokines that were of interest were further investigated in the 180 AIS patients and 14 heathy controls. We found that IL-1RA, IL-1β, IL-4, IL-5, IL-6, IL-7, IL-9, IL-10, IL-13, IL-15, EGF, G-CSF, Flt-3L, GM-CSF and Fractalkine levels were significantly decreased in severe stroke patients. In particular, IL-1β, IL-4, IL-5, IL-7, IL-9, IL-10, IL-15, G-CSF and GM-CSF were significantly reduced in AIS patients with poor outcome, compared to those with good prognosis. IL-6 was notably higher in the poor outcome group. Only IL-9 level decreased in the large infarct volume group. After adjusting for confounders, we found that IL-5 was an independent protective factor for prognosis in AIS patients with an adjusted OR of 0.042 (P = 0.007), whereas IL-6 was an independent risk predictor for AIS patients with an adjusted OR of 1.293 (P = 0.003). Our study suggests the levels of serum cytokines are related to stroke severity, short-term prognosis and cerebral infarct volume in AIS patients.
Collapse
Affiliation(s)
- Xianmei Li
- 1Department of Rehabilitation, Wenzhou People's Hospital, Wenzhou, China
| | - Siyang Lin
- 2Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaoli Chen
- 1Department of Rehabilitation, Wenzhou People's Hospital, Wenzhou, China
| | - Wensi Huang
- 3Department of Neurology, The People's Hospital of Pingyang, Wenzhou, China
| | - Qian Li
- 4Department of Neurology, Jinhua Municipal Central Hospital, Wenzhou, China
| | - Hongxia Zhang
- 5Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Xudong Chen
- 2Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shaohua Yang
- 5Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Kunlin Jin
- 5Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Bei Shao
- 2Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
35
|
Pang CY, Yang KL, Fu CH, Sun LY, Chen SY, Liao CH. G-CSF enhances the therapeutic potency of stem cells transplantation in spinal cord-injured rats. Regen Med 2019; 14:571-583. [DOI: 10.2217/rme-2018-0104] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Aim: The therapeutic effects of human wisdom teeth-derived neuronal stem cell (tNSC) cotreatment with granulocyte-colony-stimulating factor (G-CSF) were evaluated for contusion-induced spinal cord injury in rats. Materials & methods: 7 days after contusion, tNSCs were transplanted to the injury site and followed by G-CSF cotreatment for 5 days. Behavioral deficits were evaluated by the Basso, Beattie and Bresnahan test. The injury site was collected for immunohistochemistry analysis. Results: The Basso, Beattie and Bresnahan test significantly improved in the cotreated group compared with the tNSCs or G-CSF single treatment groups. However, inflammation indices did not differ among the three groups. In vitro experiment demonstrated that tNSCs express both G-CSF and its relevant receptor. G-CSF enhanced tNSC proliferation and neurotrophins secretion in vitro. Conclusion: This study demonstrated that G-CSF enhances neurotrophins secretion of tNSCs, and might help improving functional recovery from spinal cord injury in rats if they were given together.
Collapse
Affiliation(s)
- Cheng-Yoong Pang
- Department of Medical Research, Hualien Tzu Chi Hospital, Hualien, Taiwan 970
- Cardiovascular & Metabolomics Research Center, Hualien Tzu Chi Hospital, Hualien, Taiwan 970
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan 970
| | - Kuo-Liang Yang
- Buddhist Tzu Chi Stem Cells Center, Hualien Tzu Chi Hospital, Hualien, Taiwan 970
- Tzu Chi Cord Blood Bank, Hualien Tzu Chi Hospital, Hualien, Taiwan 970
| | - Chin-Hua Fu
- Department of Neurology, Taichung Tzu Chi Hospital, Taichung, Taiwan 427
| | - Li-Yi Sun
- Department of Medical Research, Hualien Tzu Chi Hospital, Hualien, Taiwan 970
- Gene & Stem Cell Production Center, Hualien Tzu Chi Hospital, Hualien, Taiwan 970
| | - Shin-Yuan Chen
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Hualien, Taiwan 970
| | - Chia-Hsin Liao
- Department of Medical Research, Hualien Tzu Chi Hospital, Hualien, Taiwan 970
- Department of Nature Science, Holistic Education Center, Tzu Chi University of Science & Technology, Hualien, Taiwan 970
| |
Collapse
|
36
|
Derakhshanrad N, Saberi H, Yekaninejad MS, Joghataei MT. Subcutaneous granulocyte colony-stimulating factor administration for subacute traumatic spinal cord injuries, report of neurological and functional outcomes: a double-blind randomized controlled clinical trial. J Neurosurg Spine 2019; 30:19-30. [PMID: 30497202 DOI: 10.3171/2018.6.spine18209] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 06/07/2018] [Indexed: 11/06/2022]
Abstract
In BriefSpinal cord injury is among the most devastating neurological conditions affecting humans. The authors assessed the therapeutic efficacy of subcutaneous recombinant granulocyte colony-stimulating factor as an adjunct to classic surgical and rehabilitative treatments for subacute traumatic spinal cord injuries. This safe and noninvasive treatment may be helpful for better care and satisfaction of patients with this devastating condition throughout the world.
Collapse
Affiliation(s)
- Nazi Derakhshanrad
- 1Brain and Spinal cord Injury Research Center, Neuroscience Institute, and
| | - Hooshang Saberi
- 1Brain and Spinal cord Injury Research Center, Neuroscience Institute, and
- 2Department of Neurosurgery, Imam Khomeini Hospital, Tehran University of Medical Sciences
| | - Mir Saeed Yekaninejad
- 3Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences; and
| | - Mohammad Taghi Joghataei
- 4Neuroscience Department, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
37
|
Bobinger T, Burkardt P, B Huttner H, Manaenko A. Programmed Cell Death after Intracerebral Hemorrhage. Curr Neuropharmacol 2018; 16:1267-1281. [PMID: 28571544 PMCID: PMC6251052 DOI: 10.2174/1570159x15666170602112851] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 04/26/2017] [Accepted: 06/01/2017] [Indexed: 01/01/2023] Open
Abstract
Background: Intracerebral hemorrhage (ICH) accounts for up to 15% of all strokes and is characterized by high rates of mortality and morbidity. The post-ICH brain injury can be distinguished in 1) primary, which are caused by disrup-tion and mechanical deformation of brain tissue due to hematoma growth and 2) secondary, which are induced by microglia activation, mitochondrial dysfunction, neurotransmitter and inflammatory mediator release. Although these events typically lead to necrosis, the occurrence of programmed cell death has also been reported after ICH. Methods: We reviewed recent publications describing advance in pre- and clinic ICH research. Results: At present, treatment of ICH patients is based on oral anticoagulant reversal, management of blood pressure and other medical complications. Several pre-clinical studies showed promising results and demonstrated that anti-oxidative and anti-inflammatory treatments reduced neuronal cell death, however, to date, all of these attempts have failed in randomized controlled clinical trials. Yet, the time frame of administration may be crucial in translation from animal to clinical studies. Furthermore, the latest pre-clinical research points toward the existence of other, apoptosis-unrelated forms kinds of pro-grammed cell death. Conclusion: Our review summarizes current knowledge of pathways leading to programmed cell death after ICH in addition to data from clinical trials. Some of the pre-clinical results have not yet demonstrated clinical confirmation, however they sig-nificantly contribute to our understanding of post-ICH pathology and can contribute to development of new therapeutic ap-proaches, decreasing mortality and improving ICH patients’ quality of life.
Collapse
Affiliation(s)
- Tobias Bobinger
- Department of Neurology, University of Erlangen-Nuremberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Petra Burkardt
- Department of Neurology, University of Erlangen-Nuremberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Hagen B Huttner
- Department of Neurology, University of Erlangen-Nuremberg, Schwabachanlage 6, Erlangen 91054, Germany
| | - Anatol Manaenko
- Department of Neurology, University of Erlangen-Nuremberg, Schwabachanlage 6, Erlangen 91054, Germany
| |
Collapse
|
38
|
Jiao Y, Li XY, Liu J. A New Approach to Cerebral Palsy Treatment: Discussion of the Effective Components of Umbilical Cord Blood and its Mechanisms of Action. Cell Transplant 2018; 28:497-509. [PMID: 30384766 PMCID: PMC7103597 DOI: 10.1177/0963689718809658] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cerebral palsy (CP) includes a group of persistent non-progressive disorders
affecting movement, muscle tone, and/or posture. The total economic loss during
the life-span of an individual with CP places a heavy financial burden on such
patients and their families worldwide; however, a complete cure is still
lacking. Umbilical cord blood (UCB)-based interventions are emerging as a
scientifically plausible treatment and possible cure for CP. Stem cells have
been used in many experimental CP animal models and achieved good results.
Compared with other types of stem cells, those from UCB have advantages in terms
of treatment safety and efficacy, ethics, non-neoplastic proliferation,
accessibility, ease of preservation, and regulation of immune responses, based
on findings in animal models and clinical trials. Currently, the use of
UCB-based interventions for CP is limited as the components of UCB are complex
and possess different therapeutic mechanisms. These can be categorized by three
aspects: homing and neuroregeneration, trophic factor secretion, and
neuroprotective effects. Our review summarizes the features of active components
of UCB and their therapeutic mechanism of action. This review highlights current
research findings and clinical evidence regarding UCB that contribute to
treatment suggestions, inform decision-making for therapeutic interventions, and
help to direct future research.
Collapse
Affiliation(s)
- Yang Jiao
- 1 Stem Cell Clinical Research Center, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, P.R. China
| | - Xiao-Yan Li
- 1 Stem Cell Clinical Research Center, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, P.R. China
| | - Jing Liu
- 1 Stem Cell Clinical Research Center, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, P.R. China
| |
Collapse
|
39
|
Kutlu MG, Brady LJ, Peck EG, Hofford RS, Yorgason JT, Siciliano CA, Kiraly DD, Calipari ES. Granulocyte Colony Stimulating Factor Enhances Reward Learning through Potentiation of Mesolimbic Dopamine System Function. J Neurosci 2018; 38:8845-8859. [PMID: 30150359 PMCID: PMC6181308 DOI: 10.1523/jneurosci.1116-18.2018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 08/14/2018] [Accepted: 08/21/2018] [Indexed: 12/15/2022] Open
Abstract
Deficits in motivation and cognition are hallmark symptoms of multiple psychiatric diseases. These symptoms are disruptive to quality of life and often do not improve with available medications. In recent years there has been increased interest in the role of the immune system in neuropsychiatric illness, but to date no immune-related treatment strategies have come to fruition. The cytokine granulocyte-colony stimulating factor (G-CSF) is known to have trophic and neuroprotective properties in the brain, and we recently identified it as a modulator of neuronal and behavioral plasticity. By combining operant tasks that assess discrete aspects of motivated behavior and decision-making in male mice and rats with subsecond dopamine monitoring via fast-scan cyclic voltammetry, we defined the role of G-CSF in these processes as well as the neural mechanism by which it modulates dopamine function to exert these effects. G-CSF enhanced motivation for sucrose as well as cognitive flexibility as measured by reversal learning. These behavioral outcomes were driven by mesolimbic dopamine system plasticity, as systemically administered G-CSF increased evoked dopamine release in the nucleus accumbens independent of clearance mechanisms. Importantly, sustained increases in G-CSF were required for these effects as acute exposure did not enhance behavioral outcomes and decreased dopamine release. These effects seem to be a result of the ability of G-CSF to alter local inflammatory signaling cascades, particularly tumor necrosis factor α. Together, these data show G-CSF as a potent modulator of the mesolimbic dopamine circuit and its ability to appropriately attend to salient stimuli.SIGNIFICANCE STATEMENT Emerging evidence has highlighted the importance of the immune system in psychiatric diseases states. However, the effects of peripheral cytokines on motivation and cognitive function are largely unknown. Here, we report that granulocyte-colony stimulating factor (G-CSF), a pleiotropic cytokine with known trophic and neuroprotective properties in the brain, acts directly on dopaminergic circuits to enhance their function. These changes in dopaminergic dynamics enhance reward learning and motivation for natural stimuli. Together, these results suggest that targeting immune factors may provide a new avenue for therapeutic intervention in the multiple psychiatric disorders that are characterized by motivational and cognitive deficits.
Collapse
Affiliation(s)
| | | | - Emily G Peck
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina 07141
| | | | - Jordan T Yorgason
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah 84602
| | - Cody A Siciliano
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina 07141
| | - Drew D Kiraly
- Department of Psychiatry,
- Friedman Brain Institute
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, New York 10029, and
| | - Erin S Calipari
- Department of Pharmacology,
- Vanderbilt Center for Addiction Research
- Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| |
Collapse
|
40
|
Prentice H, Pan C, Gharibani PM, Ma Z, Price AL, Giraldo GS, Retz HM, Gupta A, Chen PC, Chiu H, Modi J, Menzie J, Tao R, Wu JY. Analysis of Neuroprotection by Taurine and Taurine Combinations in Primary Neuronal Cultures and in Neuronal Cell Lines Exposed to Glutamate Excitotoxicity and to Hypoxia/Re-oxygenation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 975 Pt 1:207-216. [PMID: 28849456 DOI: 10.1007/978-94-024-1079-2_18] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ischemic stroke is one of the greatest contributors to death and long term disability in developed countries. Ischemia induced brain injury arises due to excessive release of glutamate and involves cell death due to apoptosis and endoplasmic reticulum (ER) stress responses. Despite major research efforts there are currently no effective treatments for stroke. Taurine, a free amino acid found in high concentrations in many invertebrate and vertebrate systems can provide protection against a range of neurological disorders. Here we demonstrate that taurine can combat ER stress responses induced by glutamate or by hypoxia/re-oxygenation in neuronal cell lines and primary neuronal cultures. Taurine decreased expression of ER stress markers GRP78, CHOP, Bim and caspase 12 in primary neuronal cultures exposed to hypoxia/re-oxygenation. In analyzing individual ER stress pathways we demonstrated that taurine treatment can result in reduced levels of cleaved ATF6 and decreased p-IRE1 levels. We hypothesized that because of the complex nature of stroke a combination therapy approach may be optimal. For this reason we proceeded to test combination therapies using taurine plus low dose administration of an additional drug: either granulocyte colony stimulating factor (G-CSF) or sulindac a non-steroidal anti-inflammatory drug with potent protective functions through signaling via ischemic preconditioning pathways. When primary neurons were pretreated with 25 mM taurine and 25 ng/mL G-CSF for I hour and then exposed to high levels of glutamate, the taurine/G-CSF combination increased the protective effect against glutamate toxicity to 88% cell survival compared to 75% cell survival from an individual treatment with taurine or G-CSF alone. Pre-exposure of PC12 cells to 5 mM taurine or 25 μM sulindac did not protect the cells from hypoxia/re-oxygenation stress whereas at these concentrations the combination of taurine plus sulindac provided significant protection. In summary we have demonstrated the protective effect of taurine in primary neuronal cultures against hypoxia with re-oxygenation through inhibition of ATF6 or p-IRE-1 pathway but not the PERK pathway of ER stress. Furthermore the combinations of taurine plus an additional drug (either G-CSF or sulindac) can show enhanced potency for protecting PC 12 cells from glutamate toxicity or hypoxia/re-oxygenation through inhibition of ER stress responses.
Collapse
Affiliation(s)
- Howard Prentice
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, 33431, USA.
| | - Chunliu Pan
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, 33431, USA
| | - Payam M Gharibani
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, 33431, USA
| | - Zhiyuan Ma
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, 33431, USA
| | - Allison L Price
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, 33431, USA
| | - Grace S Giraldo
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, 33431, USA
| | - Howard M Retz
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, 33431, USA
| | - Amit Gupta
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, 33431, USA
| | - Po-Chih Chen
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, 33431, USA
| | - Hongyuan Chiu
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, 33431, USA
| | - Jigar Modi
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, 33431, USA
| | - Janet Menzie
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, 33431, USA
| | - Rui Tao
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, 33431, USA
| | - Jang-Yen Wu
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, 33431, USA.
| |
Collapse
|
41
|
Mervosh NL, Wilson R, Rauniyar N, Hofford RS, Kutlu MG, Calipari ES, Lam TT, Kiraly DD. Granulocyte-Colony-Stimulating Factor Alters the Proteomic Landscape of the Ventral Tegmental Area. Proteomes 2018; 6:proteomes6040035. [PMID: 30249060 PMCID: PMC6313867 DOI: 10.3390/proteomes6040035] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 09/17/2018] [Accepted: 09/20/2018] [Indexed: 12/12/2022] Open
Abstract
Cocaine addiction is characterized by aberrant plasticity of the mesolimbic dopamine circuit, leading to dysregulation of motivation to seek and take drug. Despite the significant toll that cocaine use disorder exacts on society, there are currently no available pharmacotherapies. We have recently identified granulocyte-colony stimulating factor (G-CSF) as a soluble cytokine that alters the behavioral response to cocaine and which increases dopamine release from the ventral tegmental area (VTA). Despite these known effects on behavior and neurophysiology, the molecular mechanisms by which G-CSF affects brain function are unclear. In this study mice were treated with repeated injections of G-CSF, cocaine or a combination and changes in protein expression in the VTA were examined using an unbiased proteomics approach. Repeated G-CSF treatment resulted in alterations in multiple signaling pathways related to synaptic plasticity and neuronal morphology. While the treatment groups had marked overlap in their effect, injections of cocaine and the combination of cocaine and G-CSF lead to distinct patterns of significantly regulated proteins. These experiments provide valuable information as to the molecular pathways that G-CSF activates in an important limbic brain region and will help to guide further characterization of G-CSF function and evaluation as a possible translational target.
Collapse
Affiliation(s)
- Nicholas L Mervosh
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Rashaun Wilson
- Yale/NIDA Neuroproteomics Center, New Haven, CT 06511, USA.
| | - Navin Rauniyar
- Yale/NIDA Neuroproteomics Center, New Haven, CT 06511, USA.
| | - Rebecca S Hofford
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Munir Gunes Kutlu
- Department of Pharmacology, Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| | - Erin S Calipari
- Department of Pharmacology, Vanderbilt Center for Addiction Research, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| | - TuKiet T Lam
- Yale/NIDA Neuroproteomics Center, New Haven, CT 06511, USA.
- Department of Molecular Biophysics & Biochemistry, New Haven, CT 06510, USA.
- Yale MS & Proteomics Resource, New Haven, CT 06510, USA.
| | - Drew D Kiraly
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
42
|
Park CH, Joa KL, Lee MO, Yoon SH, Kim MO. The combined effect of granulocyte-colony stimulating factor (G-CSF) treatment and exercise in rats with spinal cord injury. J Spinal Cord Med 2018; 43:339-346. [PMID: 30230978 PMCID: PMC7241473 DOI: 10.1080/10790268.2018.1521567] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Objective: To identify that the combined G-CSF and treadmill exercise is more effective in functional recovery after spinal cord injury (SCI).Design: Rats were divided into 4 groups: a SCI group treated with G-CSF (G-CSF group, n = 6), a SCI group treated with treadmill exercise plus G-CSF (G-CSF/exercise group, n = 6), a SCI group with treadmill exercise (exercise group, n = 6), and a SCI group without treatments (control group, n = 6). We performed laminectomy at the T8-10 spinal levels with compression injury of the spinal cord in all rats. G-CSF (20 μg/ml) was administered intraperitoneally for 5 consecutive days after SCI in G-CSF and G-CSF/exercise groups. From one week after surgery, animals in G-CSF/exercise and exercise groups received 30 min of exercise 5 days per week for 4 weeks. Functional recoveries were assessed using the Basso, Beattie, and Bresnahan (BBB) scale and the inclined plane test. Five weeks after SCI, hematoxylin and eosin staining for cavity size and immunohistochemistry for glial scar formation and neuro-regeneration factor expression were conducted.Setting: Inha University School of medicine, Incheon, KoreaResults: Rats in G-CSF/exercise group showed the most effective functional recovery in the BBB scale and the inclined plane test, and spinal cord cavity size by injury were the smallest, and immunohistochemistry revealed expression of higher BDNF (brain-derived neurotrophic factor) and VEGF (vascular endothelial growth factor) and lower GFAP (glial fibrillary acidic protein) than others.Conclusion: Combined treatment provided more effective neuroplasty and functional recovery than individual treatments.
Collapse
Affiliation(s)
- Chan-Hyuk Park
- Department of Physical & Rehabilitation Medicine, School of Medicine, Inha University, Incheon, Republic of Korea
| | - Kyung-Lim Joa
- Department of Physical & Rehabilitation Medicine, School of Medicine, Inha University, Incheon, Republic of Korea
| | - Mi-Ok Lee
- School of Medicine, Inha University, Incheon, Republic of Korea
| | - Seung-Hwan Yoon
- Department of Neurosurgery, School of Medicine, Inha University, Incheon, Republic of Korea
| | - Myeong-Ok Kim
- Department of Physical & Rehabilitation Medicine, School of Medicine, Inha University, Incheon, Republic of Korea,Correspondence to: Myeong-Ok Kim, Department of Physical & Rehabilitation Medicine, School of Medicine, Inha University, Inha University Hospital, 27, Inhang-ro, Jung-Gu, Incheon, 222–332, Korea; Ph: +82-32-890-2480.
| |
Collapse
|
43
|
Kong Z, Hong Y, Zhu J, Cheng X, Liu Y. Endothelial progenitor cells improve functional recovery in focal cerebral ischemia of rat by promoting angiogenesis via VEGF. J Clin Neurosci 2018; 55:116-121. [PMID: 30041898 DOI: 10.1016/j.jocn.2018.07.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 05/13/2018] [Accepted: 07/08/2018] [Indexed: 02/05/2023]
Abstract
To investigate the role of venous infusion of endothelial progenitor cells (EPCs) in the reendothelialization of acute focal cerebral ischemia model in rats. And explore the mechanism of VEGF to promote angiogenesis of functional recovery in focal cerebral ischemia of rat. A model of middle cerebral artery occlusion (MCAo) was used to mimic ischemia following EPCs extraction from the same donor rats. EPCs were characterized by CD34, CD45 and CD133 expressions, and confirmed by uptake of fluorescently labeled Dil-ac-LDL and FITC-UEA-1 and flow cytometry analysis. EPCs were expanded in vitro and injected into the jugular vein of the same donor animals daily for 5 days after ischemia surgery. EPC-treated animals received approximately 1 × 106 cells, while control animals received PBS. Animals were evaluated the functional recovery, endothelial cell proliferation, vascular distribution, and VEGF levels. The EPC-treated group showed lower infarct volume and a significant recovery of neurological function. We also observed increased vascular distribution through bromodeoxyuridine (BrdU) staining and high plasma VEGF levels in the EPC-treated group compared to control groups. Our results provided direct evidence that auto-graft EPCs can improve neurological outcome and revascularization after ischemic stroke and indicated an important role of VEGF in this process. Our study suggested that EPCs may have potential therapeutic applications for the ischemic cerebrovascular disease.
Collapse
Affiliation(s)
- Zhaohong Kong
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yan Hong
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Jiang Zhu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Xin Cheng
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yumin Liu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| |
Collapse
|
44
|
Azmy MS, Menze ET, El-Naga RN, Tadros MG. Neuroprotective Effects of Filgrastim in Rotenone-Induced Parkinson's Disease in Rats: Insights into its Anti-Inflammatory, Neurotrophic, and Antiapoptotic Effects. Mol Neurobiol 2018; 55:6572-6588. [PMID: 29327204 DOI: 10.1007/s12035-017-0855-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 12/20/2017] [Indexed: 12/13/2022]
Abstract
All current treatments of Parkinson's disease (PD) focus on enhancing the dopaminergic effects and providing symptomatic relief; however, they cannot delay the disease progression. Filgrastim, a recombinant methionyl granulocyte colony-stimulating factor, demonstrated neuroprotection in many neurodegenerative and neurological diseases. This study aimed to assess the neuroprotective effects of filgrastim in rotenone-induced rat model of PD and investigate the potential underlying mechanisms of filgrastim actions. The effects of two doses of filgrastim (20 and 40 μg/kg) on spontaneous locomotion, catalepsy, body weight, histology, and striatal dopamine (DA) content, as well as tyrosine hydroxylase (TH) and α-synuclein expression, were evaluated. Then, the effective dose was further tested for its potential anti-inflammatory, neurotrophic, and antiapoptotic effects. Filgrastim (40 μg/kg) prevented rotenone-induced motor deficits, weight reduction, striatal DA depletion, and histological damage. Besides, it significantly inhibited rotenone-induced decrease in TH expression and increase in α-synuclein immunoreactivity in the midbrains and striata of the rats. These effects were associated with reduction of rotenone-induced neuroinflammation, apoptosis, and brain-derived neurotrophic factor depletion. Collectively, these results suggest that filgrastim might be a good candidate for management of PD.
Collapse
Affiliation(s)
- Mariama S Azmy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Esther T Menze
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| | - Reem N El-Naga
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mariane G Tadros
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
45
|
Zhou DG, Shi YH, Cui YQ. Impact of G-CSF on expressions of Egr-1 and VEGF in acute ischemic cerebral injury. Exp Ther Med 2018; 16:2313-2318. [PMID: 30186473 PMCID: PMC6122443 DOI: 10.3892/etm.2018.6486] [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] [Received: 12/26/2017] [Accepted: 06/25/2018] [Indexed: 12/31/2022] Open
Abstract
The aim of the present study was to investigate the protective effect of granulocyte colony-stimulating factor (G-CSF) on acute ischemic cerebral injury, and its mechanism through the impact of G-CSF on early growth response-1 (Egr-1) and vascular endothelial growth factor (VEGF) expressions. Male Sprague-Dawley (SD) rats were divided them into three groups, i.e., the sham, model and G-CSF groups to measure the effect of G-CSF on the volume of cerebral infarction and level of lactate dehydrogenase (LDH) in rats. Hematoxylin and eosin (H&E) staining method was performed for histopathological examination. Reverse transcription-polymerase chain reaction (RT-PCR) and western blot analysis were used to detect the mRNA and protein expressions of Egr-1 and VEGF in different groups. Furthermore, Statistical Product and Service Solutions (SPSS) 17.0 software was applied to detect the differences in the expression of Egr-1 and VEGF between the two groups. Compared with the sham group, we found that the volume of cerebral infarction and LDH content in the model group were significantly elevated. By contrast, in the model group, those indicators in the G-CSF group were obviously decreased. H&E staining results also showed that G-CSF could decrease the necrotic area in cerebral infarction and the incidence of inflammation, and sustain the integrity of the molecular structure. Immunofluorescence staining results revealed that the protein expressions of Egr-1 and VEGF in the model group were all significantly decreased, while those in the G-CSF group were remarkably elevated. RT-PCR and western blot analysis revealed that the mRNA and protein expressions of Egr-1 and VEGF in the model group were decreased obviously, but those in the G-CSF group were elevated significantly, and the differences between the two groups showed statistical significance (P<0.05). G-CSF manifests a significant protective effect on the acute ischemic cerebral injury, which may be realized through its effect on the expressions of Egr-1 and VEGF.
Collapse
Affiliation(s)
- Dian-Gui Zhou
- Department of Neurology, Zhongshan Torch Development Zone Hospital, Zhongshan, Guangdong 528437, P.R. China
| | - Yang-Hong Shi
- Department II of Neurology, The First Hospital of Yulin, Yulin, Shaanxi 719000, P.R. China
| | - You-Qiang Cui
- Department of Neurosurgery, Qianfoshan Hospital Affiliated to Shandong University, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
46
|
Derakhshanrad N, Saberi H, Yekaninejad MS, Joghataei MT, Sheikhrezaei A. Granulocyte-colony stimulating factor administration for neurological improvement in patients with postrehabilitation chronic incomplete traumatic spinal cord injuries: a double-blind randomized controlled clinical trial. J Neurosurg Spine 2018; 29:97-107. [PMID: 29701561 DOI: 10.3171/2017.11.spine17769] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Granulocyte-colony stimulating factor (G-CSF) is a major growth factor for activation and differentiation of granulocyte colonies in the bone marrow. This cytokine has been widely and safely employed in different conditions over many years. The purpose of this study was to investigate the efficacy of G-CSF administration for traumatic spinal cord injury (TSCI). METHODS This double-blind parallel randomized, placebo-controlled, clinical trial, a phase III study, was performed from June 2013 to June 2016 in the Brain and Spinal Cord Injury Research (BASIR) center at Tehran University of Medical Sciences (TUMS). It included 120 patients with incomplete chronic TSCI, American Spinal Injury Association (ASIA) Impairment Scale (AIS) B, C, or D, of at least 6 months' duration. Sixty patients were allocated into the treatment group and 60 patients into the control group. All the patients had completed an outpatient rehabilitation program in the postacute period and were in a neurological and functional plateau. Patients were assessed with the ASIA grading system, the Spinal Cord Independence Measure (SCIM-III), and the International Association of Neurorestoratology-Spinal Cord Injury Functional Rating Scale (IANR-SCIFRS) just before intervention and at 1, 3, and 6 months after 7 subcutaneous administrations of 300 μg/day of G-CSF in the treatment group and placebo in the control group (administered once per day over the course of 1 week). Randomization was performed with randomized block design, and the patients and evaluators were blinded regarding the treatment groups. One patient did not receive the entire allocated intervention and 5 patients were lost to follow-up. Thus data from 114 patients were included in the analysis. RESULTS One hundred twenty patients were randomized and allocated into the study groups. Among them, 56 patients (93.3%) in the G-CSF group and 58 patients (96.6%) in the placebo group completed the study protocol. After 6 months of follow-up, AIS in the placebo group remained unchanged, whereas in the G-CSF group, 1 patient improved from AIS B to C, and 4 patients improved from AIS C to D. The mean (± SE) improvement in ASIA motor score in the G-CSF group was 5.5 ± 0.62, which was significantly more than in the placebo group (0.77 ± 0.20) (p < 0.001). The mean light touch and pinprick sensory scores, respectively, increased by 6.1 ± 1.1 and 8.7 ± 1.5 in the G-CSF group and by 1.3 ± 0.52 and 0.89 ± 0.44 scores in the placebo group (p < 0.001). Evaluation of functional improvement by the IANR-SCIFRS instrument revealed significantly more improvement in the G-CSF group (3.5 ± 0.37) than in the placebo group (0.41 ± 0.12) (p < 0.001). Also, a significant difference was observed in functional improvement between the 2 groups as measured by SCIM-III instrument (7.5 ± 0.95 vs 2.1 ± 0.51, p < 0.001). CONCLUSIONS Administration of G-CSF for incomplete chronic spinal cord injuries is associated with significant motor, sensory, and functional improvement. Clinical trial registration no.: IRCT201108297441N1 ( www.irct.ir ).
Collapse
Affiliation(s)
- Nazi Derakhshanrad
- 1Brain and Spinal Cord Injury Research Center (BASIR), Neuroscience Institute, Imam Khomeini Hospital, Tehran University of Medical Sciences
| | - Hooshang Saberi
- 1Brain and Spinal Cord Injury Research Center (BASIR), Neuroscience Institute, Imam Khomeini Hospital, Tehran University of Medical Sciences
- 2Department of Neurosurgery, Imam Khomeini Hospital, Tehran University of Medical Sciences
| | - Mir Saeed Yekaninejad
- 3Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences; and
| | - Mohammad Taghi Joghataei
- 4Cellular and Molecular Research Center and
- 5Neuroscience Department, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Abdolreza Sheikhrezaei
- 2Department of Neurosurgery, Imam Khomeini Hospital, Tehran University of Medical Sciences
| |
Collapse
|
47
|
Dela Peña IC, Yang S, Shen G, Fang Liang H, Solak S, Borlongan CV. Extension of Tissue Plasminogen Activator Treatment Window by Granulocyte-Colony Stimulating Factor in a Thromboembolic Rat Model of Stroke. Int J Mol Sci 2018; 19:ijms19061635. [PMID: 29857523 PMCID: PMC6032420 DOI: 10.3390/ijms19061635] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 05/27/2018] [Accepted: 05/29/2018] [Indexed: 12/26/2022] Open
Abstract
When given beyond 4.5 h of stroke onset, tissue plasminogen activator (tPA) induces deleterious side effects in the ischemic brain, notably, hemorrhagic transformation (HT). We examined the efficacy of granulocyte-colony stimulating factor (G-CSF) in reducing delayed tPA-induced HT, cerebral infarction, and neurological deficits in a thromboembolic (TE) stroke model, and whether the effects of G-CSF were sustained for longer periods of recovery. After stroke induction, rats were given intravenous saline (control), tPA (10 mg/kg), or G-CSF (300 μg/kg) + tPA 6 h after stroke. We found that G-CSF reduced delayed tPA-associated HT by 47%, decreased infarct volumes by 33%, and improved motor and neurological deficits by 15% and 25%, respectively. It also prevented delayed tPA treatment-induced mortality by 46%. Immunohistochemistry showed 1.5- and 1.8-fold enrichment of the endothelial progenitor cell (EPC) markers CD34+ and VEGFR2 in the ischemic cortex and striatum, respectively, and 1.7- and 2.8-fold increases in the expression of the vasculogenesis marker von Willebrand factor (vWF) in the ischemic cortex and striatum, respectively, in G-CSF-treated rats compared with tPA-treated animals. Flow cytometry revealed increased mobilization of CD34+ cells in the peripheral blood of rats given G-CSF. These results corroborate the efficacy of G-CSF in enhancing the therapeutic time window of tPA for stroke treatment via EPC mobilization and enhancement of vasculogenesis.
Collapse
Affiliation(s)
- Ike C Dela Peña
- Department of Pharmaceutical and Administrative Sciences, Loma Linda University, Loma Linda, CA 92350, USA.
| | - Samuel Yang
- Department of Pharmaceutical and Administrative Sciences, Loma Linda University, Loma Linda, CA 92350, USA.
| | - Guofang Shen
- Department of Pharmaceutical and Administrative Sciences, Loma Linda University, Loma Linda, CA 92350, USA.
| | - Hsiao Fang Liang
- Department of Pharmaceutical and Administrative Sciences, Loma Linda University, Loma Linda, CA 92350, USA.
| | - Sara Solak
- Department of Pharmaceutical and Administrative Sciences, Loma Linda University, Loma Linda, CA 92350, USA.
| | - Cesar V Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA.
| |
Collapse
|
48
|
Menzie-Suderam JM, Mohammad-Gharibani P, Modi J, Ma Z, Tao R, Prentice H, Wu JY. Granulocyte-colony stimulating factor protects against endoplasmic reticulum stress in an experimental model of stroke. Brain Res 2018; 1682:1-13. [DOI: 10.1016/j.brainres.2017.12.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 12/15/2017] [Accepted: 12/18/2017] [Indexed: 10/18/2022]
|
49
|
Teixeira WGJ, Cristante AF, Marcon RM, Bispo G, Ferreira R, de Barros-Filho TEP. Granulocyte Colony-Stimulating Factor Combined with Methylprednisolone Improves Functional Outcomes in Rats with Experimental Acute Spinal Cord Injury. Clinics (Sao Paulo) 2018; 73:e235. [PMID: 29466494 PMCID: PMC5808113 DOI: 10.6061/clinics/2018/e235] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 10/17/2017] [Indexed: 12/03/2022] Open
Abstract
OBJECTIVES To evaluate the effects of combined treatment with granulocyte colony-stimulating factor (G-CSF) and methylprednisolone in rats subjected to experimental spinal cord injury. METHODS Forty Wistar rats received a moderate spinal cord injury and were divided into four groups: control (no treatment); G-CSF (G-CSF at the time of injury and daily over the next five days); methylprednisolone (methylprednisolone for 24 h); and G-CSF/Methylprednisolone (methylprednisolone for 24 h and G-CSF at the time of injury and daily over the next five days). Functional evaluation was performed using the Basso, Beattie and Bresnahan score on days 2, 7, 14, 21, 28, 35 and 42 following injury. Motor-evoked potentials were evaluated. Histological examination of the spinal cord lesion was performed immediately after euthanasia on day 42. RESULTS Eight animals were excluded (2 from each group) due to infection, a normal Basso, Beattie and Bresnahan score at their first evaluation, or autophagy, and 32 were evaluated. The combination of methylprednisolone and G-CSF promoted greater functional improvement than methylprednisolone or G-CSF alone (p<0.001). This combination also exhibited a synergistic effect, with improvements in hyperemia and cellular infiltration at the injury site (p<0.001). The groups displayed no neurophysiological differences (latency p=0.85; amplitude p=0.75). CONCLUSION Methylprednisolone plus G-CSF promotes functional and histological improvements superior to those achieved by either of these drugs alone when treating spinal cord contusion injuries in rats. Combining the two drugs did have a synergistic effect.
Collapse
Affiliation(s)
- William Gemio Jacobsen Teixeira
- Divisao de Cirurgia da Coluna, Tumores da Coluna, Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
- *Corresponding author. E-mail:
| | - Alexandre Fogaça Cristante
- Divisao de Cirurgia da Coluna, Laboratorio de Investigacao Medica, Instituto de Ortopedia e Traumatologia (IOT), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Raphael Martus Marcon
- Divisao de Cirurgia da Coluna, Laboratorio de Investigacao Medica, Instituto de Ortopedia e Traumatologia (IOT), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Gustavo Bispo
- Laboratorio de Investigacao Medica – 41 (LIM-41), Instituto de Ortopedia e Traumatologia (IOT), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Ricardo Ferreira
- Divisao de Cirurgia da Coluna, Laboratorio de Investigacao Medica, Instituto de Ortopedia e Traumatologia (IOT), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Tarcísio Eloy Pessoa de Barros-Filho
- Divisao de Cirurgia da Coluna, Laboratorio de Investigacao Medica, Instituto de Ortopedia e Traumatologia (IOT), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| |
Collapse
|
50
|
Granulocyte Colony-Stimulating Factor Alleviates Bacterial-Induced Neuronal Apoptotic Damage in the Neonatal Rat Brain through Epigenetic Histone Modification. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:9797146. [PMID: 29484107 PMCID: PMC5816840 DOI: 10.1155/2018/9797146] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 11/07/2017] [Accepted: 11/23/2017] [Indexed: 12/17/2022]
Abstract
Bacterial meningitis during the perinatal period may cause long-term neurological deficits. The study investigated whether bacterial lipopolysaccharide (LPS) derived from E. coli. led to neuronal apoptosis with an impaired performance of long-term cognitive function involving the activation of histone modification in the TNF-α gene promoter. Further, we looked into the therapeutic efficacy of granulocyte colony-stimulating factor (G-CSF) in a neonatal brain suffering from perinatal bacterial meningitis. We applied the following research techniques: neurobehavioral tasks, confocal laser microscopy, chromatin immunoprecipitation, and Western blotting. At postnatal day 10, the animals were subjected to LPS and/or G-CSF. The target brain tissues were then collected at P17. Some animals (P45) were studied using neurobehavioral tasks. The LPS-injected group revealed significantly increased expression of NF-κB phosphorylation and trimethylated H3K4 in the TNFA gene promoter locus. Furthermore, the caspase-3, neuronal apoptosis expression, and an impaired performance in cognitive functions were also found in our study. Such deleterious outcomes described above were markedly alleviated by G-CSF therapy. This study suggests that selective therapeutic action sites of G-CSF through epigenetic regulation in the TNFA gene promoter locus may exert a potentially beneficial role for the neonatal brain suffering from perinatal bacterial-induced meningitis.
Collapse
|