1
|
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
|
2
|
Luo S, Li P, Zhang A, Meng L, Huang L, Wu X, Cheng H, Tu H, Gong X. G-CSF improving combined whole brain radiotherapy and immunotherapy prognosis of non-small cell lung cancer brain metastases. Int Immunopharmacol 2024; 130:111705. [PMID: 38412673 DOI: 10.1016/j.intimp.2024.111705] [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: 12/05/2023] [Revised: 02/04/2024] [Accepted: 02/13/2024] [Indexed: 02/29/2024]
Abstract
OBJECTIVE To evaluate the therapeutic advantage of G-CSF to whole brain radiotherapy (WBRT) in combination with immunotherapy as a first-line treatment for non-small cell lung cancer (NSCLC) brain metastases (BMs). METHODS In this retrospective study, 117 patients (37 in G-CSF group and 80 in no G-CSF group) who underwent first-line WBRT combined with immunotherapy were enrolled. Their survival, intracranial response, BM-related symptoms and toxicity were evaluated. RESULTS The overall survival (OS) of patients in G-CSF group was significantly improved compared to patients no G-CSF group (median time: 14.8 vs 10.2 months; HR: 0.61, 95 % CI: 0.38-0.97, p = 0.035). However, there were no significant differences in intracranial responses between the two groups (p > 0.05). The G-CSF group exhibited a significantly higher rate of relief from BM-related symptoms compared to the no G-CSF group (91.7 % vs 59.5 %, p = 0.037). Cox proportional hazards regression analyses indicated that after-treatment ALC > 0.9 × 10^9/L (HR 0.57, 95 % CI 0.32-0.99, p = 0.046) and Hb > 110 g/dL (HR 0.41, 95 % CI 0.24-0.71, p = 0.001) were significant potential factors associated with extended OS. The addition of G-CSF was well tolerated and effectively reduced the incidence of neutropenia (0 % vs 5.0 %, p = 0.17). CONCLUSION Integrating G-CSF with WBRT and immunotherapy as a first-line treatment for NSCLC-BMs has exhibited significant efficacy and favorable tolerability.
Collapse
Affiliation(s)
- Shilan Luo
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Peng Li
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Anqi Zhang
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lu Meng
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Litang Huang
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaoting Wu
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hongxia Cheng
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hongbin Tu
- Department of Integrated TCM & Western Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaomei Gong
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
3
|
Aronowski J, Sansing LH, Xi G, Zhang JH. Mechanisms of Damage After Cerebral Hemorrhage. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00008-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
4
|
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
|
5
|
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
|
6
|
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]
|
7
|
Xavier-Elsas P, Ferreira RN, Gaspar-Elsas MIC. Surgical and immune reconstitution murine models in bone marrow research: Potential for exploring mechanisms in sepsis, trauma and allergy. World J Exp Med 2017; 7:58-77. [PMID: 28890868 PMCID: PMC5571450 DOI: 10.5493/wjem.v7.i3.58] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 06/11/2017] [Accepted: 06/30/2017] [Indexed: 02/06/2023] Open
Abstract
Bone marrow, the vital organ which maintains lifelong hemopoiesis, currently receives considerable attention, as a source of multiple cell types which may play important roles in repair at distant sites. This emerging function, distinct from, but closely related to, bone marrow roles in innate immunity and inflammation, has been characterized through a number of strategies. However, the use of surgical models in this endeavour has hitherto been limited. Surgical strategies allow the experimenter to predetermine the site, timing, severity and invasiveness of injury; to add or remove aggravating factors (such as infection and defects in immunity) in controlled ways; and to manipulate the context of repair, including reconstitution with selected immune cell subpopulations. This endows surgical models overall with great potential for exploring bone marrow responses to injury, inflammation and infection, and its roles in repair and regeneration. We review three different murine surgical models, which variously combine trauma with infection, antigenic stimulation, or immune reconstitution, thereby illuminating different aspects of the bone marrow response to systemic injury in sepsis, trauma and allergy. They are: (1) cecal ligation and puncture, a versatile model of polymicrobial sepsis; (2) egg white implant, an intriguing model of eosinophilia induced by a combination of trauma and sensitization to insoluble allergen; and (3) ectopic lung tissue transplantation, which allows us to dissect afferent and efferent mechanisms leading to accumulation of hemopoietic cells in the lungs. These models highlight the gain in analytical power provided by the association of surgical and immunological strategies.
Collapse
|
8
|
Redondo-Castro E, Cunningham C, Miller J, Martuscelli L, Aoulad-Ali S, Rothwell NJ, Kielty CM, Allan SM, Pinteaux E. Interleukin-1 primes human mesenchymal stem cells towards an anti-inflammatory and pro-trophic phenotype in vitro. Stem Cell Res Ther 2017; 8:79. [PMID: 28412968 PMCID: PMC5393041 DOI: 10.1186/s13287-017-0531-4] [Citation(s) in RCA: 169] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 03/01/2017] [Accepted: 03/08/2017] [Indexed: 12/25/2022] Open
Abstract
Background Inflammation is a key contributor to central nervous system (CNS) injury such as stroke, and is a major target for therapeutic intervention. Effective treatments for CNS injuries are limited and applicable to only a minority of patients. Stem cell-based therapies are increasingly considered for the treatment of CNS disease, because they can be used as in-situ regulators of inflammation, and improve tissue repair and recovery. One promising option is the use of bone marrow-derived mesenchymal stem cells (MSCs), which can secrete anti-inflammatory and trophic factors, can migrate towards inflamed and injured sites or can be implanted locally. Here we tested the hypothesis that pre-treatment with inflammatory cytokines can prime MSCs towards an anti-inflammatory and pro-trophic phenotype in vitro. Methods Human MSCs from three different donors were cultured in vitro and treated with inflammatory mediators as follows: interleukin (IL)-1α, IL-1β, tumour necrosis factor alpha (TNF-α) or interferon-γ. After 24 h of treatment, cell supernatants were analysed by ELISA for expression of granulocyte-colony stimulating factor (G-CSF), IL-10, brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), IL-1 receptor antagonist (IL-1Ra) and vascular endothelial growth factor (VEGF). To confirm the anti-inflammatory potential of MSCs, immortalised mouse microglial BV2 cells were treated with bacterial lipopolysaccharide (LPS) and exposed to conditioned media (CM) of naïve or IL-1-primed MSCs, and levels of secreted microglial-derived inflammatory mediators including TNF-α, IL-10, G-CSF and IL-6 were measured by ELISA. Results Unstimulated MSCs constitutively expressed anti-inflammatory cytokines and trophic factors (IL-10, VEGF, BDNF, G-CSF, NGF and IL-1Ra). MSCs primed with IL-1α or IL-1β showed increased secretion of G-CSF, which was blocked by IL-1Ra. Furthermore, LPS-treated BV2 cells secreted less inflammatory and apoptotic markers, and showed increased secretion of the anti-inflammatory IL-10 in response to treatment with CM of IL-1-primed MSCs compared with CM of unprimed MSCs. Conclusions Our results demonstrate that priming MSCs with IL-1 increases expression of trophic factor G-CSF through an IL-1 receptor type 1 (IL-1R1) mechanism, and induces a reduction in the secretion of inflammatory mediators in LPS-activated microglial cells. The results therefore support the potential use of preconditioning treatments of stem cells in future therapies. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0531-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Elena Redondo-Castro
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Catriona Cunningham
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jonjo Miller
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Licia Martuscelli
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Sarah Aoulad-Ali
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Nancy J Rothwell
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Cay M Kielty
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Stuart M Allan
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Emmanuel Pinteaux
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
9
|
Wang D, Liu K, Wake H, Teshigawara K, Mori S, Nishibori M. Anti-high mobility group box-1 (HMGB1) antibody inhibits hemorrhage-induced brain injury and improved neurological deficits in rats. Sci Rep 2017; 7:46243. [PMID: 28393932 PMCID: PMC5385548 DOI: 10.1038/srep46243] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 03/09/2017] [Indexed: 02/06/2023] Open
Abstract
As one of the most lethal stroke subtypes, intracerebral hemorrhage (ICH) is acknowledged as a serious clinical problem lacking effective treatment. Available evidence from preclinical and clinical studies suggests that inflammatory mechanisms are involved in the progression of ICH-induced secondary brain injury. High mobility group box-1 (HMGB1) is a ubiquitous and abundant nonhistone DNA-binding protein, and is also an important proinflammatory molecule once released into the extracellular space from the nuclei. Here, we show that treatment with neutralizing anti-HMGB1 mAb (1 mg/kg, i.v. twice) remarkably ameliorated ICH-injury induced by local injection of collagenase IV in the striatum of rats. Administration of anti-HMGB1 mAb inhibited the release of HMGB1 into the extracellular space in the peri-hematomal region, reduced serum HMGB1 levels and decreased brain edema by protecting blood-brain barrier integrity, in association with decreased activated microglia and the expression of inflammation-related factors at 24 h after ICH. Consequently, anti-HMGB1 mAb reduced the oxidative stress and improved the behavioral performance of rats. These results strongly indicate that HMGB1 plays a critical role in the development of ICH-induced secondary injury through the amplification of plural inflammatory responses. Intravenous injection of neutralizing anti-HMGB1 mAb has potential as a novel therapeutic strategy for ICH.
Collapse
Affiliation(s)
- Dengli Wang
- Department of Pharmacology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Keyue Liu
- Department of Pharmacology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hidenori Wake
- Department of Pharmacology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kiyoshi Teshigawara
- Department of Pharmacology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shuji Mori
- School of Pharmacy, Shujitsu University, Okayama, Japan
| | - Masahiro Nishibori
- Department of Pharmacology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
10
|
Ghorbani M, Mohammadpour AH, Abnous K, Movassaghi AR, Sarshoori JR, Shahsavand S, Hashemzaei M, Moallem SA. G-CSF administration attenuates brain injury in rats following carbon monoxide poisoning via different mechanisms. ENVIRONMENTAL TOXICOLOGY 2017; 32:37-47. [PMID: 26502830 DOI: 10.1002/tox.22210] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 10/04/2015] [Accepted: 10/04/2015] [Indexed: 06/05/2023]
Abstract
Acute severe carbon monoxide (CO) poisoning induces hypoxia that leads to cardiovascular and nervous systems disturbances. Different complex mechanisms lead to CO neurotoxicity including lipid peroxidation, inflammatory and immune-mediated reactions, myelin degeneration and finally neuronal apoptosis and necrosis. Granulocyte colony-stimulating factor (G-CSF) is considered to be a novel neuroprotective agent. In this study, we evaluated the efficacy of G-CSF therapy on CO neurotoxicity in rats with acute CO poisoning. Rats were exposed to 3000 ppm CO in air (0.3%) for 1 h, and then different doses (50,100, and 150 µg/kg) of G-CSF or normal saline were administrated intraperitoneally. Water content of brain as an indicator for total edema and blood brain barrier integrity (Evans blue extravasation) were evaluated. Malondialydehyde was determined in order to evaluate the effect of G-CSF on CO-induced lipid peroxidation in brain tissues. Also, the effect of G-CSF on myeloperoxidase activity in the brain tissue was evaluated. The effect of G-CSF administration on induced apoptosis in the brain was measured using TUNEL method. To evaluate the level of MBP, STAT3 and pSTAT3 and HO-1 proteins and the effect of G-CSF on these proteins Western blotting was carried out. G-CSF reduced water content of the edematous poisoned brains (100 µg/kg) and BBB permeability (100 and 150 µg/kg) (P < 0.05). G-CSF (150 µg/kg) reduced the MDA level in the brain tissues (P < 0.05 as compared to CO poisoned animals). G-CSF did not decrease the MPO activity after CO poisoning in any doses. G-CSF significantly reduced the number of apoptotic neurons and Caspase 3 protein levels in the brain. Western blotting results showed that G-CSF treatment enhanced expression of HO-1 and MBP, STAT3 and pSTAT3 proteins in the brain tissues. Based on our results, a single dose of G-CSF immediately after CO poisoning significantly attenuates CO neurotoxicity via different mechanisms. © 2015 Wiley Periodicals, Inc. Environ Toxicol 32: 37-47, 2017.
Collapse
Affiliation(s)
- Maryam Ghorbani
- Department of Pharmacology and Toxicology, School of Pharmacy, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Amir Hooshang Mohammadpour
- Pharmaceutical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ahmad Reza Movassaghi
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Javad Raouf Sarshoori
- Department of Anatomy, School of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Shabnam Shahsavand
- Department of Physiology and Pharmacology, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Mahmoud Hashemzaei
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Zabol University of Medical Sciences, Zabol, Iran
| | - Seyed Adel Moallem
- Pharmaceutical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
11
|
Edwardson MA, Wang X, Liu B, Ding L, Lane CJ, Park C, Nelsen MA, Jones TA, Wolf SL, Winstein CJ, Dromerick AW. Stroke Lesions in a Large Upper Limb Rehabilitation Trial Cohort Rarely Match Lesions in Common Preclinical Models. Neurorehabil Neural Repair 2017; 31:509-520. [PMID: 28337932 DOI: 10.1177/1545968316688799] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Stroke patients with mild-moderate upper extremity motor impairments and minimal sensory and cognitive deficits provide a useful model to study recovery and improve rehabilitation. Laboratory-based investigators use lesioning techniques for similar goals. OBJECTIVE To determine whether stroke lesions in an upper extremity rehabilitation trial cohort match lesions from the preclinical stroke recovery models used to drive translational research. METHODS Clinical neuroimages from 297 participants enrolled in the Interdisciplinary Comprehensive Arm Rehabilitation Evaluation (ICARE) study were reviewed. Images were characterized based on lesion type (ischemic or hemorrhagic), volume, vascular territory, depth (cortical gray matter, cortical white matter, subcortical), old strokes, and leukoaraiosis. Lesions were compared with those of preclinical stroke models commonly used to study upper limb recovery. RESULTS Among the ischemic stroke participants, median infarct volume was 1.8 mL, with most lesions confined to subcortical structures (61%) including the anterior choroidal artery territory (30%) and the pons (23%). Of ICARE participants, <1% had lesions resembling proximal middle cerebral artery or surface vessel occlusion models. Preclinical models of subcortical white matter injury best resembled the ICARE population (33%). Intracranial hemorrhage participants had small (median 12.5 mL) lesions that best matched the capsular hematoma preclinical model. CONCLUSIONS ICARE subjects are not representative of all stroke patients, but they represent a clinically and scientifically important subgroup. Compared with lesions in general stroke populations and widely studied animal models of recovery, ICARE participants had smaller, more subcortically based strokes. Improved preclinical-clinical translational efforts may require better alignment of lesions between preclinical and human stroke recovery models.
Collapse
Affiliation(s)
- Matthew A Edwardson
- 1 Georgetown University, Washington, DC, USA.,2 MedStar National Rehabilitation Hospital, Washington, DC, USA
| | - Ximing Wang
- 3 University of Southern California, Los Angeles, CA, USA
| | - Brent Liu
- 3 University of Southern California, Los Angeles, CA, USA
| | - Li Ding
- 3 University of Southern California, Los Angeles, CA, USA
| | | | - Caron Park
- 3 University of Southern California, Los Angeles, CA, USA
| | | | | | - Steven L Wolf
- 5 Emory University, Atlanta, GA, USA.,6 VA Center on Visual and Neurocognitive Rehabilitation, Decatur, GA, USA
| | | | - Alexander W Dromerick
- 1 Georgetown University, Washington, DC, USA.,2 MedStar National Rehabilitation Hospital, Washington, DC, USA.,7 VA Medical Center, Washington, DC, USA
| |
Collapse
|
12
|
Liu XC, Jing LY, Yang MF, Wang K, Wang Y, Fu XY, Fang J, Hou YJ, Sun JY, Li DW, Zhang ZY, Mao LL, Tang YM, Fu XT, Fan CD, Yang XY, Sun BL. Enhanced Neuroprotection of Minimally Invasive Surgery Joint Local Cooling Lavage against ICH-induced Inflammation Injury and Apoptosis in Rats. Cell Mol Neurobiol 2016; 36:647-55. [PMID: 26224360 PMCID: PMC11482297 DOI: 10.1007/s10571-015-0245-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 07/23/2015] [Indexed: 11/30/2022]
Abstract
Hypothermia treatment is one of the neuroprotective strategies that improve neurological outcomes effectively after brain damage. Minimally invasive surgery (MIS) has been an important treatment of intracerebral hemorrhage (ICH). Herein, we evaluated the neuroprotective effect and mechanism of MIS joint local cooling lavage (LCL) treatment on ICH via detecting the inflammatory responses, oxidative injury, and neuronal apoptosis around the hematoma cavity in rats. ICH model was established by type IV collagenase caudatum infusion. The rats were treated with MIS 6 h after injection, and then were lavaged by normothermic (37 °C) and hypothermic (33 °C) normal saline in brain separately. The results indicated that MIS joint LCL treatment showed enhanced therapeutic effects against ICH-induced inflammation injury and apoptosis in rats, as convinced by the decline of TUNEL-positive cells, followed by the decrease of IL-1β and LDH and increase of IL-10 and SOD. This study demonstrated that the strategy of using MIS joint LCL may achieve enhanced neuroprotection against ICH-induced inflammation injury and apoptosis in rats with potential clinic application.
Collapse
Affiliation(s)
- Xi-Chang Liu
- Department of Neurology, Shandong University School of Medicine, Jinan, 250012, Shandong, China
- Key Lab of Cerebral Microcirculation in Universities of Shandong, Taishan Medical University, Taian, 271000, Shandong, China
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264100, Shandong, China
| | - Li-Yan Jing
- Taian Chinese Medicine Hospital, Taian, 271000, Shandong, China
| | - Ming-Feng Yang
- Key Lab of Cerebral Microcirculation in Universities of Shandong, Taishan Medical University, Taian, 271000, Shandong, China
| | - Kun Wang
- Key Lab of Cerebral Microcirculation in Universities of Shandong, Taishan Medical University, Taian, 271000, Shandong, China
- Taishan Vocational College of Nursing, Taian, 271000, Shandong, China
| | - Yuan Wang
- Shandong Provincial Hospital Affiliated To Shandong University, Jinan, 250012, Shandong, China
| | - Xiao-Yan Fu
- School of Basic Medicine, Taishan Medical University, Taian, 271000, Shandong, China
| | - Jie Fang
- Key Lab of Cerebral Microcirculation in Universities of Shandong, Taishan Medical University, Taian, 271000, Shandong, China
| | - Ya-Jun Hou
- Key Lab of Cerebral Microcirculation in Universities of Shandong, Taishan Medical University, Taian, 271000, Shandong, China
| | - Jing-Yi Sun
- School of Basic Medicine, Taishan Medical University, Taian, 271000, Shandong, China
| | - Da-Wei Li
- Key Lab of Cerebral Microcirculation in Universities of Shandong, Taishan Medical University, Taian, 271000, Shandong, China
| | - Zong-Yong Zhang
- Key Lab of Cerebral Microcirculation in Universities of Shandong, Taishan Medical University, Taian, 271000, Shandong, China
| | - Lei-Lei Mao
- Key Lab of Cerebral Microcirculation in Universities of Shandong, Taishan Medical University, Taian, 271000, Shandong, China
| | - You-Mei Tang
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264100, Shandong, China
| | - Xiao-Ting Fu
- Key Lab of Cerebral Microcirculation in Universities of Shandong, Taishan Medical University, Taian, 271000, Shandong, China
| | - Cun-Dong Fan
- Key Lab of Cerebral Microcirculation in Universities of Shandong, Taishan Medical University, Taian, 271000, Shandong, China.
| | - Xiao-Yi Yang
- Key Lab of Cerebral Microcirculation in Universities of Shandong, Taishan Medical University, Taian, 271000, Shandong, China
| | - Bao-Liang Sun
- Department of Neurology, Shandong University School of Medicine, Jinan, 250012, Shandong, China.
- Key Lab of Cerebral Microcirculation in Universities of Shandong, Taishan Medical University, Taian, 271000, Shandong, China.
- Affiliated Hospital of Taishan Medical University, Taian, 271000, Shandong, China.
| |
Collapse
|
13
|
Neubauer V, Wegleiter K, Posod A, Urbanek M, Wechselberger K, Kiechl-Kohlendorfer U, Keller M, Griesmaier E. Delayed application of the haematopoietic growth factors G-CSF/SCF and FL reduces neonatal excitotoxic brain injury. Brain Res 2016; 1634:94-103. [PMID: 26772988 DOI: 10.1016/j.brainres.2015.12.058] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 12/03/2015] [Accepted: 12/29/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND Developmental brain injury results in cognitive and motor deficits in the preterm infant. Enhanced glutamate release and subsequent receptor activation are major pathogenetic factors. The effect of haematopoietic growth factors, such as granulocyte colony-stimulating factor (G-CSF), stem cell factor (SCF) and flt-3 ligand (FL) on neonatal brain injury is controversially discussed. Timing of treatment is known to be a crucial factor. Based on the hypothesis that an exacerbation of injury is caused by administration of substances in the acute phase, the objective of this study was to evaluate the effect of delayed administration of G-CSF/SCF and FL to protect against excitotoxic brain injury in vivo. METHODS In an established neonatal mouse model of excitotoxic brain injury, we evaluated the effect of daily intraperitoneal doses of G-CSF/SCF or FL, starting 60 h after the excitotoxic insult. RESULTS Intraperitoneal injections of G-CSF/SCF and FL, given 60 h after the excitotoxic insult, significantly reduced lesion size at postnatal days 10, 18 and 90. G-CSF/SCF treatment resulted in a decrease in apoptotic cell death indicated by reduced caspase-3 activation. G-CSF/SCF and FL treatment did not affect apoptosis-inducing factor-dependent apoptosis or cell proliferation. CONCLUSION We show that delayed systemic treatment with the haematopoietic growth factors G-CSF/SCF and FL protects against N-methyl-D-aspartate receptor-mediated developmental excitotoxic brain damage. Our results suggest that neuroprotective effects in this neonatal animal model of excitotoxic brain injury depend on the timing of drug administration after the insult.
Collapse
Affiliation(s)
- Vera Neubauer
- Department of Paediatrics II (Neonatology), Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Karina Wegleiter
- Department of Paediatrics II (Neonatology), Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Anna Posod
- Department of Paediatrics II (Neonatology), Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Martina Urbanek
- Department of Paediatrics II (Neonatology), Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Karina Wechselberger
- Department of Paediatrics II (Neonatology), Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Ursula Kiechl-Kohlendorfer
- Department of Paediatrics II (Neonatology), Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria
| | - Matthias Keller
- Kinderklinik Dritter Orden, Munich Technical University, Bischof Altmann-Strasse 9, 94032 Passau, Germany
| | - Elke Griesmaier
- Department of Paediatrics II (Neonatology), Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria.
| |
Collapse
|
14
|
Liew HK, Cheng HY, Huang LC, Li KW, Peng HF, Yang HI, Lin PBC, Kuo JS, Pang CY. Acute Alcohol Intoxication Aggravates Brain Injury Caused by Intracerebral Hemorrhage in Rats. J Stroke Cerebrovasc Dis 2016; 25:15-25. [DOI: 10.1016/j.jstrokecerebrovasdis.2015.08.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 08/06/2015] [Accepted: 08/19/2015] [Indexed: 11/29/2022] Open
|
15
|
Mechanisms of Cerebral Hemorrhage. Stroke 2016. [DOI: 10.1016/b978-0-323-29544-4.00008-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
16
|
Sang LYH, Liang YX, Li Y, Wong WM, Tay DKC, So KF, Ellis-Behnke RG, Wu W, Cheung RTF. A self-assembling nanomaterial reduces acute brain injury and enhances functional recovery in a rat model of intracerebral hemorrhage. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 11:611-20. [DOI: 10.1016/j.nano.2014.05.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 05/15/2014] [Accepted: 05/28/2014] [Indexed: 01/03/2023]
|
17
|
Krafft PR, McBride DW, Lekic T, Rolland WB, Mansell CE, Ma Q, Tang J, Zhang JH. Correlation between subacute sensorimotor deficits and brain edema in two mouse models of intracerebral hemorrhage. Behav Brain Res 2014; 264:151-60. [PMID: 24518201 DOI: 10.1016/j.bbr.2014.01.052] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 01/28/2014] [Accepted: 01/31/2014] [Indexed: 12/11/2022]
Abstract
Formation of brain edema after intracerebral hemorrhage (ICH) is highly associated with its poor outcome. However, the relationship between cerebral edema and behavioral deficits has not been thoroughly examined in the preclinical setting. Hence, this study aimed to evaluate the ability of common sensorimotor tests to predict the extent of brain edema in two mouse models of ICH. One hundred male CD-1 mice were subjected to sham surgery or ICH induction via intrastriatal injection of either autologous blood (30 μL) or bacterial collagenase (0.0375U or 0.075U). At 24 and 72 h after surgery, animals underwent a battery of behavioral tests, including the modified Garcia neuroscore (Neuroscore), corner turn test (CTT), forelimb placing test (FPT), wire hang task (WHT) and beam walking (BW). Brain edema was evaluated via the wet weight/dry weight method. Intrastriatal injection of autologous blood or bacterial collagenase resulted in a significant increase in brain water content and associated sensorimotor deficits (p<0.05). A significant correlation between brain edema and sensorimotor deficits was observed for all behavioral tests except for WHT and BW. Based on these findings, we recommend implementing the Neuroscore, CTT and/or FPT in preclinical studies of unilateral ICH in mice.
Collapse
Affiliation(s)
- Paul R Krafft
- Department of Physiology & Pharmacology Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Devin W McBride
- Department of Physiology & Pharmacology Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Tim Lekic
- Department of Physiology & Pharmacology Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - William B Rolland
- Department of Physiology & Pharmacology Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Charles E Mansell
- Department of Physiology & Pharmacology Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Qingyi Ma
- Department of Physiology & Pharmacology Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Jiping Tang
- Department of Physiology & Pharmacology Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - John H Zhang
- Department of Physiology & Pharmacology Loma Linda University School of Medicine, Loma Linda, CA, USA; Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA, USA.
| |
Collapse
|
18
|
Chen F, Liu Q, Zhang ZD, Zhu XH. Co-delivery of G-CSF and EPO released from fibrin gel for therapeutic neovascularization in rat hindlimb ischemia model. Microcirculation 2014; 20:416-24. [PMID: 23294128 DOI: 10.1111/micc.12037] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2012] [Accepted: 01/03/2013] [Indexed: 01/10/2023]
Abstract
OBJECTIVE G-CSF and EPO have shown a notable capability in neovascularization. However, their use is limited because of untoward leucocytosis, erythrogenesis, and short half-life in the plasma. Herein, we examined whether G-CSF and EPO released from fibrin gel injected into ischemic tissues would synergistically promote neovascularization with limited systematic effects in a rat hindlimb ischemic model. METHODS AND RESULTS In vivo study, group Gel received an intramuscular injection of fibrin gel; group Gel+G-CSF received fibrin gel containing human G-CSF; group Gel+EPO received fibrin gel containing human EPO; group Gel+G-CSF&EPO received fibrin gel containing G-CSF and EPO; group G-CSF&EPO received G-CSF and EPO. Through promoting the expression of SDF-1, local high concentration of EPO could traffic CXCR4+ cells mobilized by G-CSF to enhance neovascularization in ischemic muscle. The treatment with Gel+G-CSF&EPO was superior to the other treatments on blood flow reperfusion, capillary density, and α smooth muscle actin-positive vessel density. And this treatment induced a modest WBC count increase in peripheral blood. CONCLUSIONS G-CSF and EPO released from fibrin gel had a combined effect on postischemia neovascularization. This treatment may be a novel therapeutic modality for ischemic peripheral artery disease.
Collapse
Affiliation(s)
- Feng Chen
- Department of Vascular Surgery, the second affiliated Hospital, Nanchang University, Nanchang, China.
| | | | | | | |
Collapse
|
19
|
Protection of granulocyte-colony stimulating factor to hemorrhagic brain injuries and its involved mechanisms: Effects of vascular endothelial growth factor and aquaporin-4. Neuroscience 2014; 260:59-72. [DOI: 10.1016/j.neuroscience.2013.12.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Revised: 12/08/2013] [Accepted: 12/09/2013] [Indexed: 01/27/2023]
|
20
|
Kallmünzer B, Tauchi M, Schlachetzki JC, Machold K, Schmidt A, Winkler J, Schwab S, Kollmar R. Granulocyte colony-stimulating factor does not promote neurogenesis after experimental intracerebral haemorrhage. Int J Stroke 2013; 9:783-8. [PMID: 24920160 DOI: 10.1111/ijs.12217] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND Hematopoietic growth factors have been suggested to induce neuroprotective and regenerative effects in various animal models of cerebral injury. However, the pathways involved remain widely unexplored. AIMS This study aimed to investigate effects of local and systemic administration of granulocyte colony-stimulating factor on brain damage, functional recovery, and cerebral neurogenesis in an intracerebral haemorrhage whole blood injection model in rats. METHODS Eight-week-old male Wistar rats (n = 100) underwent induction of striatal intracerebral haemorrhage by autologous whole blood injection or sham procedure and were randomly assigned to either (a) systemic treatment with granulocyte colony-stimulating factor (60 μg/kg) for five-days; (b) single intracerebral injection of granulocyte colony-stimulating factor (60 μg/kg) into the cavity; or (c) application of vehicle for five-days. Bromodeoxyuridine-labelling and immunohistochemistry were used to analyze proliferation and survival of newly born cells in the sub-ventricular zone and the hippocampal dentate gyrus. Moreover, functional deficits and lesion volume were assessed until day 42 after intracerebral haemorrhage. RESULTS Differences in lesion size or hemispheric atrophy between granulocyte colony-stimulating factor-treated and control groups did not reach statistical significance. Neither systemic, nor local granulocyte colony-stimulating factor administration induced neurogenesis within the dentate gyrus or the sub-ventricular zone. The survival of newborn cells in these regions was prevented by intracerebral granulocyte colony-stimulating factor application. A subtle benefit in functional recovery at day 14 after intracerebral haemorrhage induction was observed after granulocyte colony-stimulating factor treatment. CONCLUSION There was a lack of neuroprotective or neuroregenerative effects of granulocyte colony-stimulating factor in the present rodent model of intracerebral haemorrhage. Conflicting results from functional outcome assessment require further research.
Collapse
Affiliation(s)
- Bernd Kallmünzer
- Department of Neurology, University Medical Center Erlangen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Belur PK, Chang JJ, He S, Emanuel BA, Mack WJ. Emerging experimental therapies for intracerebral hemorrhage: targeting mechanisms of secondary brain injury. Neurosurg Focus 2013; 34:E9. [PMID: 23634928 DOI: 10.3171/2013.2.focus1317] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Intracerebral hemorrhage (ICH) is associated with a higher degree of morbidity and mortality than other stroke subtypes. Despite this burden, currently approved treatments have demonstrated limited efficacy. To date, therapeutic strategies have principally targeted hematoma expansion and resultant mass effect. However, secondary mechanisms of brain injury are believed to be critical effectors of cell death and neurological outcome following ICH. This article reviews the pathophysiology of secondary brain injury relevant to ICH, examines pertinent experimental models, and highlights emerging therapeutic strategies. Treatment paradigms discussed include thrombin inhibitors, deferoxamine, minocycline, statins, granulocyte-colony stimulating factors, and therapeutic hypothermia. Despite promising experimental and preliminary human data, further studies are warranted prior to effective clinical translation.
Collapse
Affiliation(s)
- Praveen K Belur
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | | | | | | | | |
Collapse
|
22
|
Bath PMW, Sprigg N, England T. Colony stimulating factors (including erythropoietin, granulocyte colony stimulating factor and analogues) for stroke. Cochrane Database Syst Rev 2013; 2013:CD005207. [PMID: 23797623 PMCID: PMC11441151 DOI: 10.1002/14651858.cd005207.pub4] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Colony stimulating factors (CSFs), also called haematopoietic growth factors, regulate bone marrow production of circulating red and white cells, and platelets. Some CSFs also mobilise the release of bone marrow stem cells into the circulation. CSFs have been shown to be neuroprotective in experimental stroke. OBJECTIVES To assess (1) the safety and efficacy of CSFs in people with acute or subacute ischaemic or haemorrhagic stroke, and (2) the effect of CSFs on circulating stem and blood cell counts. SEARCH METHODS We searched the Cochrane Stroke Group Trials Register (last searched September 2012), the Cochrane Central Register of Controlled Trials (CENTRAL) (The Cochrane Library 2012, Issue 4), MEDLINE (1985 to September 2012), EMBASE (1985 to September 2012) and Science Citation Index (1985 to September 2012). In an attempt to identify further published, unpublished and ongoing trials we contacted manufacturers and principal investigators of trials (last contacted April 2012). We also searched reference lists of relevant articles and reviews. SELECTION CRITERIA We included randomised controlled trials recruiting people with acute or subacute ischaemic or haemorrhagic stroke. CSFs included stem cell factor (SCF), erythropoietin (EPO), granulocyte colony stimulating factor (G-CSF), granulocyte-macrophage colony stimulating factor (GM-CSF), macrophage-colony stimulating factor (M-CSF, CSF-1), thrombopoietin (TPO), or analogues of these. The primary outcome was functional outcome at the end of the trial. Secondary outcomes included safety at the end of treatment, death at the end of follow-up, infarct volume and haematology measures. DATA COLLECTION AND ANALYSIS Two review authors (TE and NS) independently extracted data and assessed trial quality. We contacted study authors for additional information. MAIN RESULTS We included a total of 11 studies involving 1275 participants. In three trials (n = 782), EPO therapy was associated with a significant increase in death by the end of the trial (odds ratio (OR) 1.98, 95% confidence interval (CI) 1.19 to 3.3, P = 0.009) and a non-significant increase in serious adverse events. EPO significantly increased the red cell count with no effect on platelet or white cell count, or infarct volume. Two small trials of carbamylated EPO have been completed but have yet to be reported. We included eight small trials (n = 548) of G-CSF. G-CSF was associated with a non-significant reduction in early impairment (mean difference (MD) -0.4, 95% CI -1.82 to 1.01, P = 0.58) but had no effect on functional outcome at the end of the trial. G-CSF significantly elevated the white cell count and the CD34+ cell count, but had no effect on infarct volume. Further trials of G-CSF are ongoing. AUTHORS' CONCLUSIONS There are significant safety concerns regarding EPO therapy for stroke. It is too early to know whether other CSFs improve functional outcome.
Collapse
Affiliation(s)
- Philip M W Bath
- Division of Stroke Medicine, University of Nottingham, Nottingham, UK.
| | | | | |
Collapse
|
23
|
Prakash A, Medhi B, Chopra K. Granulocyte colony stimulating factor (GCSF) improves memory and neurobehavior in an amyloid-β induced experimental model of Alzheimer's disease. Pharmacol Biochem Behav 2013; 110:46-57. [PMID: 23756182 DOI: 10.1016/j.pbb.2013.05.015] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2012] [Revised: 05/21/2013] [Accepted: 05/25/2013] [Indexed: 10/26/2022]
Abstract
GCSF is an endogenous neuronal hematopoietic factor that displays robust in vitro and in vivo neuroprotective activity. The present study aimed to evaluate the effect of GCSF on Aβ-induced memory loss in an Alzheimer's disease model of rats. A total of 42 male adult Wistar rats weighing 200-250 g were used in the study and were divided into 7 experimental groups. Animals were subjected to intracerebroventricular (ICV) injection stereotaxically at day 0 to instill amyloid-β(1-42) (Aβ(1-42)) or PBS (sham operated group) at 10 μl (5 μl bilaterally). GCSF treatment was given from day 7 to 12 of Aβ injection. On day 21, behavioral tests (short term memory, exploratory behavior and motor coordination) in all groups were evaluated. Biochemical parameters and RNA expression were measured to ensure the efficacy of GCSF. GCSF (35 and 70 μg/kg, s.c.) showed statistically significant improvement in memory as compared to control and sham operated groups (p<0.05). Mean time spent in the platform placed quadrant was found to be significantly increased in the GCSF (70 μg/kg, s.c.) as compared to GCSF (35 μg/kg, s.c.) and GCSF (10 μg/kg, s.c.) groups (p<0.001). GCSF (35 and 70 μg/kg, s.c.) also improved motor coordination and exploratory behavior significantly as compared to naïve sham operated and GCSF (10 μg/kg, s.c.) groups (p<0.05). Improvement in memory by GCSF (35 and 70 μg/kg, s.c.) was coupled with marked reduction of lipid peroxidation, acetylcholinesterase levels and a significant increase in antioxidant enzymes as well as total RNA expression in the brain. Additionally, GCSF (35 and 70 μg/kg, s.c.) significantly increased progenitor cells (iPSCs) and surface marker CD34+ in the brain and hence induced neurogenesis. The present findings demonstrate an improvement of memory and neurobehavioral function with GCSF in Aβ-induced Alzheimer's disease model in rats.
Collapse
Affiliation(s)
- Ajay Prakash
- Pharmacology Division, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | | | | |
Collapse
|
24
|
Peng BH, Borisevich V, Popov VL, Zacks MA, Estes DM, Campbell GA, Paessler S. Production of IL-8, IL-17, IFN-gamma and IP-10 in human astrocytes correlates with alphavirus attenuation. Vet Microbiol 2013; 163:223-34. [PMID: 23428380 PMCID: PMC7117234 DOI: 10.1016/j.vetmic.2012.11.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Revised: 11/06/2012] [Accepted: 11/22/2012] [Indexed: 01/30/2023]
Abstract
Venezuelan equine encephalitis virus (VEEV) is an important, naturally emerging zoonotic pathogen. Recent outbreaks in Venezuela and Colombia in 1995 indicate that VEEV still poses a serious public health threat. Astrocytes may be target cells in human and mouse infection and they play an important role in repair through gliosis. In this study, we report that virulent VEEV efficiently infects cultured normal human astrocytes, three different murine astrocyte cell lines and astrocytes in the mouse brain. The attenuation of virus replication positively correlates with the increased levels of production of IL-8, IL-17, IFN-gamma and IP-10. In addition, VEEV infection induces release of basic fibroblast growth factor and production of potent chemokines such as RANTES and MIP-1-beta from cultured human astrocytes. This growth factor and cytokine profile modeled by astrocytes in vitro may contribute to both neuroprotection and repair and may play a role in leukocyte recruitment in vivo.
Collapse
Affiliation(s)
- Bi-Hung Peng
- Department of Pathology/Institute for Human Infections and Immunity, Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555-0609, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
SANG YH, LIANG YX, LIU LG, ELLIS-BEHNKE RG, WU WT, SO KF, CHEUNG RTF. A Rat Model of Intracerebral Hemorrhage Permitting Hematoma Aspiration plus Intralesional Injection. Exp Anim 2013; 62:63-9. [DOI: 10.1538/expanim.62.63] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Affiliation(s)
- Yan-Hua SANG
- Department of Anatomy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yu-Xiang LIANG
- State Key Laboratory of Brain and Cognitive Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Department of Anatomy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ling-Guang LIU
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Rutledge G. ELLIS-BEHNKE
- Department of Ophthalmology, Medical Faculty Mannheim, Ruprecht-Karls-University, Heidelberg, Germany
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Anatomy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Brain and Cognitive Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Wu-Tian WU
- Research Centre of Reproduction, Development and Growth, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Brain and Cognitive Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Department of Anatomy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Kwok-Fai SO
- State Key Laboratory of Brain and Cognitive Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Research Centre of Heart, Brain, Hormone and Healthy Aging, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Department of Anatomy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Raymond Tak-Fai CHEUNG
- Research Centre of Heart, Brain, Hormone and Healthy Aging, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
26
|
Pereira Lopes FR, Martin PKM, Frattini F, Biancalana A, Almeida FM, Tomaz MA, Melo PA, Borojevic R, Han SW, Martinez AMB. Double gene therapy with granulocyte colony-stimulating factor and vascular endothelial growth factor acts synergistically to improve nerve regeneration and functional outcome after sciatic nerve injury in mice. Neuroscience 2012; 230:184-97. [PMID: 23103791 DOI: 10.1016/j.neuroscience.2012.10.025] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Revised: 10/09/2012] [Accepted: 10/10/2012] [Indexed: 12/17/2022]
Abstract
Peripheral-nerve injuries are a common clinical problem and often result in long-term functional deficits. Reconstruction of peripheral-nerve defects is currently undertaken with nerve autografts. However, there is a limited availability of nerves that can be sacrificed and the functional recovery is never 100% satisfactory. We have previously shown that gene therapy with vascular endothelial growth factor (VEGF) significantly improved nerve regeneration, neuronal survival, and muscle activity. Our hypothesis is that granulocyte colony-stimulating factor (G-CSF) synergizes with VEGF to improve the functional outcome after sciatic nerve transection. The left sciatic nerves and the adjacent muscle groups of adult mice were exposed, and 50 or 100 μg (in 50 μl PBS) of VEGF and/or G-CSF genes was injected locally, just below the sciatic nerve, and transferred by electroporation. The sciatic nerves were transected and placed in an empty polycaprolactone (PCL) nerve guide, leaving a 3-mm gap to challenge nerve regeneration. After 6 weeks, the mice were perfused and the sciatic nerve, the dorsal root ganglion (DRG), the spinal cord and the gastrocnemius muscle were processed for light and transmission electron microscopy. Treated animals showed significant improvement in functional and histological analyses compared with the control group. However, the best results were obtained with the G-CSF+VEGF-treated animals: quantitative analysis of regenerated nerves showed a significant increase in the number of myelinated fibers and blood vessels, and the number of neurons in the DRG and motoneurons in the spinal cord was significantly higher. Motor function also showed that functional recovery occurred earlier in animals receiving G-CSF+VEGF-treatment. The gastrocnemius muscle showed an increase in weight and in the levels of creatine phosphokinase, suggesting an improvement of reinnervation and muscle activity. These results suggest that these two factors acted synergistically and optimized the nerve repair potential, improving regeneration after a transection lesion.
Collapse
Affiliation(s)
- F R Pereira Lopes
- Programa de Neurociência Básica e Clínica, Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, RJ, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Brunswick AS, Hwang BY, Appelboom G, Hwang RY, Piazza MA, Connolly ES. Serum biomarkers of spontaneous intracerebral hemorrhage induced secondary brain injury. J Neurol Sci 2012; 321:1-10. [PMID: 22857988 DOI: 10.1016/j.jns.2012.06.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2011] [Revised: 06/13/2012] [Accepted: 06/23/2012] [Indexed: 01/01/2023]
Abstract
Intracerebral hemorrhage (ICH) is a devastating form of stroke associated with a high rate of morbidity and mortality. It is now believed that much of this damage occurs in the subacute period following the initial insult via a cascade of complex pathophysiologic pathways that continues to be investigated. Increased levels of certain serum proteins have been identified as biomarkers that may reflect or directly participate in the inflammation, blood brain barrier disruption, endothelial dysfunction, and neuronal and glial toxicity that occur during this secondary period of cerebral injury. Some of these biomarkers have the potential to serve as therapeutic targets or surrogate endpoints for future research or clinical trials. Others may someday augment current clinical techniques in diagnosis, risk-stratification, prognostication, treatment decision and measurement of therapeutic efficacy. While much work remains to be done, biomarkers show significant potential to expand clinical options and improve clinical management, thereby reducing mortality and improving functional outcomes in ICH patients.
Collapse
Affiliation(s)
- Andrew S Brunswick
- Department of Neurological Surgery, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | | | | | | | | | | |
Collapse
|
28
|
Liew HK, Pang CY, Hsu CW, Wang MJ, Li TY, Peng HF, Kuo JS, Wang JY. Systemic administration of urocortin after intracerebral hemorrhage reduces neurological deficits and neuroinflammation in rats. J Neuroinflammation 2012; 9:13. [PMID: 22257737 PMCID: PMC3271957 DOI: 10.1186/1742-2094-9-13] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Accepted: 01/19/2012] [Indexed: 12/18/2022] Open
Abstract
Background Intracerebral hemorrhage (ICH) remains a serious clinical problem lacking effective treatment. Urocortin (UCN), a novel anti-inflammatory neuropeptide, protects injured cardiomyocytes and dopaminergic neurons. Our preliminary studies indicate UCN alleviates ICH-induced brain injury when administered intracerebroventricularly (ICV). The present study examines the therapeutic effect of UCN on ICH-induced neurological deficits and neuroinflammation when administered by the more convenient intraperitoneal (i.p.) route. Methods ICH was induced in male Sprague-Dawley rats by intrastriatal infusion of bacterial collagenase VII-S or autologous blood. UCN (2.5 or 25 μg/kg) was administered i.p. at 60 minutes post-ICH. Penetration of i.p. administered fluorescently labeled UCN into the striatum was examined by fluorescence microscopy. Neurological deficits were evaluated by modified neurological severity score (mNSS). Brain edema was assessed using the dry/wet method. Blood-brain barrier (BBB) disruption was assessed using the Evans blue assay. Hemorrhagic volume and lesion volume were assessed by Drabkin's method and morphometric assay, respectively. Pro-inflammatory cytokine (TNF-α, IL-1β, and IL-6) expression was evaluated by enzyme-linked immunosorbent assay (ELISA). Microglial activation and neuronal loss were evaluated by immunohistochemistry. Results Administration of UCN reduced neurological deficits from 1 to 7 days post-ICH. Surprisingly, although a higher dose (25 μg/kg, i.p.) also reduced the functional deficits associated with ICH, it is significantly less effective than the lower dose (2.5 μg/kg, i.p.). Beneficial results with the low dose of UCN included a reduction in neurological deficits from 1 to 7 days post-ICH, as well as a reduction in brain edema, BBB disruption, lesion volume, microglial activation and neuronal loss 3 days post-ICH, and suppression of TNF-α, IL-1β, and IL-6 production 1, 3 and 7 days post-ICH. Conclusion Systemic post-ICH treatment with UCN reduces striatal injury and neurological deficits, likely via suppression of microglial activation and inflammatory cytokine production. The low dose of UCN necessary and the clinically amenable peripheral route make UCN a potential candidate for development into a clinical treatment regimen.
Collapse
Affiliation(s)
- Hock-Kean Liew
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Liew HK, Hsu CW, Wang MJ, Kuo JS, Li TY, Peng HF, Wang JY, Pang CY. Therapeutic benefit of urocortin in rats with intracerebral hemorrhage. J Neurosurg 2012; 116:193-200. [DOI: 10.3171/2011.8.jns101637] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Object
Intracerebral hemorrhage (ICH) accounts for about 15% of all deaths due to stroke. It frequently causes brain edema, leading to an expansion of brain volume that exerts a negative impact on ICH outcomes. The ICH-induced brain edema involves inflammatory mechanisms. The authors' in vitro study shows that urocortin (UCN) exhibits antiinflammatory and neuroprotective effects. Therefore, the neuroprotective effect of UCN on ICH in rats was investigated.
Methods
Intracerebral hemorrhage was induced by an infusion of bacteria collagenase type VII-S or autologous blood into the unilateral striatum of anesthetized rats. At 1 hour after the induction of ICH, UCN (0.05, 0.5, and 5 μg) was infused into the lateral ventricle on the ipsilateral side. The authors examined the injury area, brain water content, blood-brain barrier permeability, and neurological function.
Results
The UCN, administered in the ipsilateral lateral ventricle, was able to penetrate into the injured striatum. Posttreatment with UCN reduced the injury area, brain edema, and blood-brain barrier permeability and improved neurological deficits of rats with ICH.
Conclusions
Posttreatment with UCN through improving neurological deficits of rats with ICH dose dependently provided a potential therapeutic agent for patients with ICH or other brain injuries.
Collapse
Affiliation(s)
- Hock-Kean Liew
- 1Departments of Medical Research and
- 3Graduate Institute of Life Sciences, National Defense Medical Center; and
| | - Chih-Wei Hsu
- 2Emergency Medicine, Tzu Chi General Hospital
- 6School of Medicine, Tzu Chi University, Hualien
| | - Mei-Jen Wang
- 1Departments of Medical Research and
- 4Graduate Institute of Medical Sciences, and
| | - Jon-Son Kuo
- 4Graduate Institute of Medical Sciences, and
| | | | | | - Jia-Yi Wang
- 3Graduate Institute of Life Sciences, National Defense Medical Center; and
- 5Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan
| | - Cheng-Yoong Pang
- 1Departments of Medical Research and
- 4Graduate Institute of Medical Sciences, and
| |
Collapse
|
30
|
Hematopoietic Growth Factor Family for Stroke Drug Development. Transl Stroke Res 2012. [DOI: 10.1007/978-1-4419-9530-8_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
31
|
Delayed granulocyte colony-stimulating factor treatment promotes functional recovery in rats with severe contusive spinal cord injury. Spine (Phila Pa 1976) 2012; 37:10-7. [PMID: 22024901 DOI: 10.1097/brs.0b013e31823b0440] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN We used a severe contusive spinal cord injury (SCI) model and electrophysiologic, motor functional, immunohistochemical, and electron microscopic examinations to analyze the neuroprotective effects of delayed granulocyte colony-stimulating factor (G-CSF) treatment. OBJECTIVE To determine the neuroprotective effects of delayed G-CSF treatment using multimodality evaluations after severe contusive SCI in rats. SUMMARY OF BACKGROUND DATA Despite some reports that G-CSF treatment in the acute stage of different central nervous system injury models was neuroprotective, it has not been determined whether delayed G-CSF treatment can promote neural recovery in severe contusive SCI. METHODS Rats with severe contusive SCI were divided into 2 groups: G-CSF group rats were given serial subcutaneous injections of G-CSF, and control group rats (controls) were given only saline injections on postcontusion days 9 to 13. Using the Basso-Beattie-Bresnahan scale and cortical somatosensory evoked potentials, we recorded functional evaluations weekly. The spinal cords were harvested for protein and immunohistochemical analysis, and for electron microscopy examination. RESULTS The preserved spinal cord area was larger in G-CSF group rats than in control group rats. Both sensory and motor functions improved after G-CSF treatment. Detachment and disruption of the myelin sheets in the myelinated axons were significantly decreased, and axons sprouted and regenerated. There were fewer microglia and macrophages in the G-CSF group than in the control group. The levels of brain-derived neurotrophic factor were comparable between the 2 groups. CONCLUSION Delayed G-CSF treatment at the subacute stage of severe contusive SCI promoted spinal cord preservation and improved functional outcomes. The mechanism of G-CSF's protection may be related in part to attenuating the infiltration of microglia and macrophages.
Collapse
|
32
|
Sang YH, Su HX, Wu WT, So KF, Cheung RTF. Elevated blood pressure aggravates intracerebral hemorrhage-induced brain injury. J Neurotrauma 2011; 28:2523-34. [PMID: 21988112 PMCID: PMC3235342 DOI: 10.1089/neu.2010.1680] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Elevated blood pressure (BP) is commonly seen in patients with intracerebral hemorrhage (ICH), and is independently associated with poor functional outcomes. Little is known about how elevated BP influences ICH-related brain injury. In the present study, we investigated the physiological and brain histological changes, as well as functional recovery following ICH in renovascular hypertensive rats. Renovascular hypertension (RVHT) was achieved by applying a silver clip onto the left renal artery of adult Sprague-Dawley rats. ICH was induced by an intrastriatal injection of bacterial collagenase IV about 5-6 weeks after left renal artery clipping or the sham operation. Following induction of ICH, both the normotensive and RVHT rats demonstrated an ultra-acute elevation in BP. Elevated BP increased hematoma volume, brain swelling, and apoptosis in the perihematomal areas. Brain degeneration, including local atrophy and lateral ventricle enlargement, was greater in the RVHT rats. In addition, many proliferating cells were seen over the ipsilateral striatum in the RVHT rats after ICH. The modified limb placing tests were done weekly for 3 weeks. In line with the histological damage, elevated BP worsened neurological deficits. These results suggest that ICH in the hypertensive rats mimics the clinical scenario of hypertensive ICH and may provide a platform to study the mechanisms of ICH-induced brain injury and potential therapies for ICH.
Collapse
Affiliation(s)
- Yan-Hua Sang
- Department of Medicine, The University of Hong Kong, China
- Department of Anatomy, The University of Hong Kong, China
| | - Huan-Xing Su
- Department of Anatomy, The University of Hong Kong, China
| | - Wu-Tian Wu
- Department of Anatomy, The University of Hong Kong, China
- Research Center of Reproduction, Development and Growth, The University of Hong Kong, China
| | - Kwok-Fai So
- Department of Anatomy, The University of Hong Kong, China
- Research Centre of Heart, Brain, Hormone and Healthy Aging, Li Ka Shing Faculty of Medicine, The University of Hong Kong, China
| | - Raymond Tak-Fai Cheung
- Department of Medicine, The University of Hong Kong, China
- Research Centre of Heart, Brain, Hormone and Healthy Aging, Li Ka Shing Faculty of Medicine, The University of Hong Kong, China
| |
Collapse
|
33
|
Sobrino T, Arias S, Pérez-Mato M, Agulla J, Brea D, Rodríguez-Yáñez M, Castillo J. Cd34+ progenitor cells likely are involved in the good functional recovery after intracerebral hemorrhage in humans. J Neurosci Res 2011; 89:979-85. [PMID: 21488087 DOI: 10.1002/jnr.22627] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Revised: 01/12/2011] [Accepted: 01/19/2011] [Indexed: 11/09/2022]
Abstract
Bone marrow-derived stem/progenitor cells (CD34(+) progenitor cells) were demonstrated to play an important role in the regeneration of damaged brain tissue. Our aim was to study the influence of CD34(+) progenitor cells in the outcome of intracerebral hemorrhage (ICH). Thirty-two patients with primary ICH (64.0% male, mean age 67.1 ± 10.8 years) were prospectively included in the study within 12 hr of symptom onset. The main outcome variable was good functional outcome at 3 months (modified Rankin scale ≤ 2). Circulating CD34(+) progenitor cell levels were measured by flow cytometry at admission and at 7 ± 1 days, and serum levels of growth factors (determined by ELISA) were measured at admission and at 24 and 72 hr. Circulating levels of CD34(+) progenitor cells at day 7 were independently associated with good functional outcome at 3 months (OR 1.17, CI95% 1.06-1.39, P = 0.012). On the other hand, CD34(+) progenitor cells at day 7 were negatively correlated with residual cavity volume at 3 months (r = -0.607, P = 0.001). Serum levels of vascular endothelial growth factor (r = 0.386), angiopoietin 1 (r = 0.518), brain-derived neurotrophic factor (r = 0.484), and stromal cell-derived factor-1α (r = 0.837) but not granulocyte-colony stimulating factor (r = -0.038) at 72 hr showed a strong correlation with CD34(+) progenitor cell levels at day 7. These findings suggest that CD34(+) progenitor cells may participate in the functional recovery of ICH patients.
Collapse
Affiliation(s)
- Tomás Sobrino
- Department of Neurology, Clinical Neurosciences Research Laboratory, Hospital Clínico Universitario, IDIS, University of Santiago de Compostela, Santiago de Compostela, Spain.
| | | | | | | | | | | | | |
Collapse
|
34
|
Frantzias J, Sena ES, Macleod MR, Al-Shahi Salman R. Treatment of intracerebral hemorrhage in animal models: meta-analysis. Ann Neurol 2011; 69:389-99. [PMID: 21387381 DOI: 10.1002/ana.22243] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Interventions that improve functional outcome after acute intracerebral hemorrhage (ICH) in animals might benefit humans. Therefore, we systematically reviewed the literature to find studies of nonsurgical treatments tested in animal models of ICH. METHODS In July 2009 we searched Ovid Medline (from 1950), Embase (from 1980), and ISI Web of Knowledge (from 1969) for controlled animal studies of nonsurgical interventions given after the induction of ICH that reported neurobehavioral outcome. We assessed study quality and performed meta-analysis using a weighted mean difference random effects model. RESULTS Of 13,343 publications, 88 controlled studies described the effects of 64 different medical interventions (given a median of 2 hours after ICH induction) on 38 different neurobehavioral scales in 2,616 treated or control animals (median 14 rodents per study). Twenty-seven (31%) studies randomized treatment allocation, and 7 (8%) reported allocation concealment; these studies had significantly smaller effect sizes than those without these attributes (p < 0.001). Of 64 interventions stem cells, calcium channel blockers, anti-inflammatory drugs, iron chelators, and estrogens improved both structural outcomes and neurobehavioral scores in >1 study. Meta-regression revealed that together, structural outcome and the intervention used accounted for 65% of the observed heterogeneity in neurobehavioral score (p < 0.001, adjusted r(2) = 0.65). INTERPRETATION Further animal studies of the interventions that we found to improve both functional and structural outcomes in animals, using better experimental designs, could target efforts to translate effective treatments for ICH in animals into randomized controlled trials in humans.
Collapse
Affiliation(s)
- Joseph Frantzias
- Division of Clinical Neurosciences, Centre for Clinical Brain Sciences, University of Edinburgh, United Kingdom
| | | | | | | |
Collapse
|
35
|
Khatibi NH, Jadhav V, Saidi M, Chen W, Martin R, Stier G, Tang J, Zhang JH. Granulocyte colony-stimulating factor treatment provides neuroprotection in surgically induced brain injured mice. ACTA NEUROCHIRURGICA. SUPPLEMENT 2011; 111:265-9. [PMID: 21725766 PMCID: PMC3569057 DOI: 10.1007/978-3-7091-0693-8_44] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Surgically induced brain injury (SBI) is a common concern after a neurosurgical procedure. Current treatments aimed at reducing the postoperative sequela are limited. Granulocyte-colony stimulating factor (G-CSF), a hematopoietic growth factor involved in the inflammatory process, has been shown in various animal models to be neuroprotective. Consequently, in this study, we investigated the use of G-CSF as a treatment modality to reduce cell death and brain edema, while improving neurobehavioral deficits following an SBI in mice. Eleven-week-old C57 black mice (n=76) were randomly placed into four groups: sham (n=19), SBI (n=21), SBI with G-CSF pre-treatment (n=15) and SBI with G-CSF pre/post-treatment (n=21). Treated groups received a single dose of G-CSF intraperitoneally at 24, 12 and 1 h pre-surgery and/or 6 and 12 h post-surgery. Postoperative assessment occurred at 24 h and included neurobehavioral testing and measurement for both cell death and brain edema. Results indicated that pre-treatment with G-CSF reduced both cell death and brain edema, while post-treatment reduced neurobehavioral deficits. This study implies that the morphological changes in the brain are effected by pre-treatment; however, in order to activate and/or amplify targets involved in the recovery process, more dosing regimens may be needed.
Collapse
Affiliation(s)
- Nikan H. Khatibi
- Department of Anesthesiology, Loma Linda Medical Center, Loma Linda, CA, USA
| | - Vikram Jadhav
- Department of Physiology, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - Mehdi Saidi
- Department of Physiology, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - Wanqiu Chen
- Department of Physiology, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - Robert Martin
- Department of Anesthesiology, Loma Linda Medical Center, Loma Linda, CA, USA
| | - Gary Stier
- Department of Anesthesiology, Loma Linda Medical Center, Loma Linda, CA, USA
| | - Jiping Tang
- Department of Physiology, Loma Linda University, School of Medicine, Loma Linda, CA, USA and Department of Physiology and Pharmacology, Loma Linda University, School of Medicine, Loma Linda, CA 92354, USA
| | - John H. Zhang
- Department of Anesthesiology, Loma Linda Medical Center, Loma Linda, CA, USA and Department of Physiology, Loma Linda University, School of Medicine, Loma Linda, CA, USA and Department of Neurosurgery, Loma Linda Medical Center, Loma Linda, CA, USA
| |
Collapse
|
36
|
Katsuki H. Exploring neuroprotective drug therapies for intracerebral hemorrhage. J Pharmacol Sci 2010; 114:366-78. [PMID: 21081835 DOI: 10.1254/jphs.10r05cr] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Intracerebral hemorrhage (ICH) is a devastating neurological disorder with high mortality and poor prognosis, for which virtually no effective drug therapies are available at present. Experimental animal models, based on intrastriatal injection of collagenase or autologous blood, have enabled great advances in elucidation of cellular/molecular events contributing to brain pathogenesis associated with ICH. Many lines of evidence indicate that blood constituents, including hemoglobin-derived products as well as proteases such as thrombin, play important roles in the pathogenic events. Inflammatory reactions involving neutrophils, activated microglia, and production of proinflammatory cytokines also constitute a critical aspect of pathology leading to neurodegeneration and tissue damage. Efforts are continuing to find drugs that potentially alleviate pathological and neurological outcomes of ICH. Various drugs that possess antioxidative, anti-inflammatory or neurotrophic/neuroprotective properties have been demonstrated to produce therapeutic effects on ICH animal models. Drugs already in clinical use such as minocycline, statins, and several nuclear receptor ligands are among the list of effective drugs, but whether they also show therapeutic efficacy in human ICH patients remains unproven. Here, current knowledge of ICH pathogenesis and problems arising with respect to exploration of new drug candidates are discussed.
Collapse
Affiliation(s)
- Hiroshi Katsuki
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Kumamoto 862-0973, Japan.
| |
Collapse
|
37
|
Sanli AM, Serbes G, Calişkan M, Kaptanoğlu E, Sargon MF, Kilinç K, Beşalti O, Sekerci Z. Effect of granulocyte-colony stimulating factor on spinal cord tissue after experimental contusion injury. J Clin Neurosci 2010; 17:1548-52. [PMID: 20801040 DOI: 10.1016/j.jocn.2010.03.043] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2010] [Revised: 03/09/2010] [Accepted: 03/10/2010] [Indexed: 12/01/2022]
Abstract
The purpose of this study was to investigate the early effects of granulocyte-colony stimulating factor (G-CSF) on myeloperoxidase (MPO) activity, lipid peroxidation (LPO) and ultrastructural findings in rats after spinal cord injury (SCI). We also compared the effects of G-CSF and methylprednisolone sodium succinate (MPSS). Wistar rats were divided into four groups: control, SCI alone (50 g/cm weight drop trauma), SCI+MPSS (30 mg/kg), and SCI+G-CSF (50 μg/kg). Administration of G-CSF and MPSS significantly decreased LPO (p < 0.05) and MPO activity (p < 0.05) in the first 24 hours. MPSS was more effective than G-CSF in reducing LPO (p < 0.05) and in minimizing ultrastructure changes. The results of this study indicate that G-CSF exerts a beneficial effect by decreasing MPO activity and LPO and may reduce tissue damage in the first 24 hours after SCI. Our findings do not exclude the possibility that G-CSF has a protective effect on spinal cord ultrastructure after the first 24 hours following SCI.
Collapse
Affiliation(s)
- A Metin Sanli
- Department of Neurosurgery, Dişkapi Yildirım Beyazit Research and Education Hospital, Ankara, Turkey.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Beray-Berthat V, Delifer C, Besson VC, Girgis H, Coqueran B, Plotkine M, Marchand-Leroux C, Margaill I. Long-term histological and behavioural characterisation of a collagenase-induced model of intracerebral haemorrhage in rats. J Neurosci Methods 2010; 191:180-90. [DOI: 10.1016/j.jneumeth.2010.06.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Accepted: 06/19/2010] [Indexed: 10/19/2022]
|
39
|
Ohnishi M, Katsuki H, Unemura K, Izumi Y, Kume T, Takada-Takatori Y, Akaike A. Heme oxygenase-1 contributes to pathology associated with thrombin-induced striatal and cortical injury in organotypic slice culture. Brain Res 2010; 1347:170-8. [PMID: 20515663 DOI: 10.1016/j.brainres.2010.05.077] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2010] [Revised: 05/24/2010] [Accepted: 05/25/2010] [Indexed: 12/14/2022]
Abstract
The blood coagulation factor thrombin that leaks from ruptured vessels initiates brain tissue damage after intracerebral hemorrhage. We have recently shown that mitogen-activated protein kinases (MAPKs) activated by thrombin exacerbate hemorrhagic brain injury via supporting survival of neuropathic microglia. Here, we investigated whether induction of heme oxygenase (HO)-1 is involved in these events. Zinc protoporphyrin IX (ZnPP IX), a HO-1 inhibitor, attenuated thrombin-induced injury of cortical cells in a concentration-dependent manner (0.3-3 microM) and tended to inhibit shrinkage of the striatal tissue at 0.3 microM. HO-1 expression was induced by thrombin in microglia and astrocytes in both the cortex and the striatum. The increase of HO-1 protein was suppressed by a p38 MAPK inhibitor SB203580, and early activation of p38 MAPK after thrombin treatment was observed in neurons and microglia in the striatum. Notably, concomitant application of a low concentration (0.3 microM) of ZnPP IX with thrombin induced apoptotic cell death in striatal microglia and significantly decreased the number of activated microglia in the striatal region. On the other hand, a carbon monoxide releaser reversed the protective effect of ZnPP IX on thrombin-induced injury of cortical cells. Overall, these results suggest that p38 MAPK-dependent induction of HO-1 supports survival of striatal microglia during thrombin insults. Thrombin-induced cortical injury may be also regulated by the expression of HO-1 and the resultant production of heme degradation products such as carbon monoxide.
Collapse
Affiliation(s)
- Masatoshi Ohnishi
- Department of Pharmacotherapeutics, Faculty of Pharmacy and Pharmaceutical Sciences, Fukuyama University, Gakuencho-1, Fukuyama, Hiroshima 729-0292, Japan
| | | | | | | | | | | | | |
Collapse
|
40
|
Liu SP, Lee SD, Lee HT, Liu DD, Wang HJ, Liu RS, Lin SZ, Shyu WC. Granulocyte colony-stimulating factor activating HIF-1alpha acts synergistically with erythropoietin to promote tissue plasticity. PLoS One 2010; 5:e10093. [PMID: 20404921 PMCID: PMC2852409 DOI: 10.1371/journal.pone.0010093] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2009] [Accepted: 03/14/2010] [Indexed: 12/29/2022] Open
Abstract
Stroke and peripheral limb ischemia are serious clinical problems with poor prognosis and limited treatment. The cytokines erythropoietin (EPO) and granulocyte-colony stimulating factor (G-CSF) have been used to induce endogenous cell repair and angiogenesis. Here, we demonstrated that the combination therapy of EPO and G-CSF exerted synergistic effects on cell survival and functional recovery from cerebral and peripheral limbs ischemia. We observed that even under normoxic conditions, G-CSF activates hypoxia-inducible factor-1alpha (HIF-1alpha), which then binds to the EPO promoter and enhances EPO expression. Serum EPO level was significantly increased by G-CSF injection, with the exception of Tg-HIF-1alpha(+f/+f) mice. The neuroplastic mechanisms exerted by EPO combined with G-CSF included enhanced expression of the antiapoptotic protein of Bcl-2, augmented neurotrophic factors synthesis, and promoted neovascularization. Further, the combination therapy significantly increased homing and differentiation of bone marrow stem cells (BMSCs) and intrinsic neural progenitor cells (INPCs) into the ischemic area. In summary, EPO in combination with G-CSF synergistically enhanced angiogenesis and tissue plasticity in ischemic animal models, leading to greater functional recovery than either agent alone.
Collapse
Affiliation(s)
- Shih-Ping Liu
- Center for Neuropsychiatry, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Shin-Da Lee
- Department of Physical Therapy, Graduate Institute of Rehabilitation Science, China Medical University, Taichung, Taiwan
- Department of Healthcare Administration, Asia University, Taichung, Taiwan
| | - Hsu-Tung Lee
- Department of Neurosurgery, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Demeral David Liu
- Department of Dentistry, China Medical University Hospital, Taichung, Taiwan
| | - Hsiao-Jung Wang
- Center for Neuropsychiatry, China Medical University Hospital, Taichung, Taiwan
| | - Ren-Shyan Liu
- Department of Nuclear Medicine, Taipei Veterans General Hospital, National Yang-Ming University, Taipei, Taiwan
| | - Shinn-Zong Lin
- Center for Neuropsychiatry, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Immunology, China Medical University, Taichung, Taiwan
| | - Woei-Cherng Shyu
- Center for Neuropsychiatry, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Immunology, China Medical University, Taichung, Taiwan
| |
Collapse
|
41
|
Shin H, Yoo SR, Ko JY, Kim KS, Ro YS. Granulocyte colony-stimulating factor for recalcitrant postherpetic neuralgia: a prospective, open, pilot study in three patients. Clin Exp Dermatol 2010. [DOI: 10.1111/j.1365-2230.2009.03570.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
42
|
Tarella C, Rutella S, Gualandi F, Melazzini M, Scimè R, Petrini M, Moglia C, Ulla M, Omedé P, Bella VL, Corbo M, Silani V, Siciliano G, Mora G, Caponnetto C, Sabatelli M, Chiò A. Consistent bone marrow-derived cell mobilization following repeated short courses of granulocyte–colony-stimulating factor in patients with amyotrophic lateral sclerosis: results from a multicenter prospective trial. Cytotherapy 2010; 12:50-9. [DOI: 10.3109/14653240903300682] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
43
|
High serum levels of growth factors are associated with good outcome in intracerebral hemorrhage. J Cereb Blood Flow Metab 2009; 29:1968-74. [PMID: 19756022 DOI: 10.1038/jcbfm.2009.182] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In experimental models, growth factors (GFs) such as vascular endothelial growth factor (VEGF), Angiopoietin 1 (Ang-1), or granulocyte-colony stimulating factor (G-CSF) mediate brain recovery after intracerebral hemorrhage (ICH). Our aim was to study the association between serum levels of GF and clinical outcome in patients with ICH. A total of 95 patients with primary ICH (male, 66.3%; mean age, 67.8+/-9.8 years) were prospectively included in the study within 12 h from symptoms onset. The main outcome variable was good functional outcome at 3 months (modified Rankin scale < or =2). Median serum levels of GF at 72 h from stroke onset were significantly higher in patients with good outcome (n=39) compared with those with poor outcome (all P<0.0001). Serum levels of VEGF > or =330 pg/mL, G-CSF > or =413 pg/mL, and Ang-1 > or =35 ng/mL at 72 h were independently associated with good functional outcome (odds ratio (OR), 11.2; 95% confidence interval (CI): 2.9 to 43.0; OR, 19.6; 95% CI: 3.9 to 97.9; and OR, 14.7; 95% CI: 3.6 to 60.0, respectively), neurologic improvement (all P<0.0001) and reduced residual cavity at 3 months (all P<0.01). These results illustrate that high serum levels of GF are associated with good functional outcome and reduced lesion volume in ICH.
Collapse
|
44
|
Granulocyte-colony stimulating factor for stroke treatment: mechanisms of action and efficacy in preclinical studies. EXPERIMENTAL & TRANSLATIONAL STROKE MEDICINE 2009; 1:2. [PMID: 20142989 PMCID: PMC2816868 DOI: 10.1186/2040-7378-1-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2009] [Accepted: 10/21/2009] [Indexed: 01/22/2023]
Abstract
G-CSF is widely employed for the treatment of chemotherapy-induced neutropenia. Recently, neuroprotective effects of G-CSF in animal stroke models were discovered including infarct size reduction and enhancement of functional recovery. The underlying mechanisms of action of G-CSF in ischemia appear to be a direct anti-apoptotic activity in neurons and a neurogenesis inducing capacity. Additional effects may be based on the stimulation of new blood-vessel formation, the stimulation of immunocompetence and -modulation as well as on bone marrow mobilization. In addition to a discussion of these mechanisms, we will review the available preclinical studies and analyze their impact on the overall efficacy of G-CSF in experimental stroke.
Collapse
|
45
|
The role of granulocyte-colony stimulating factor (G-CSF) in the healthy brain: a characterization of G-CSF-deficient mice. J Neurosci 2009; 29:11572-81. [PMID: 19759304 DOI: 10.1523/jneurosci.0453-09.2009] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Granulocyte-colony stimulating factor (G-CSF) is a hematopoietic growth factor that controls proliferation and differentiation of neural stem cells. Although recent studies have begun to explore G-CSF-related mechanisms of action in various disease models, little is known about its function in the healthy brain. In the present study, the effect of G-CSF deficiency on memory formation and motor skills was investigated. The impact of G-CSF deficiency on the structural integrity of the hippocampus was evaluated by analyzing the generation of doublecortin-expressing cells, the amount of bromodeoxyurine-labeled cells, the dendritic complexity in hippocampal neurons, the binding densities of NMDA and GABA(A) receptors and the induction of long-term potentiation (LTP). G-CSF deficiency caused a disruption in memory formation and in the development of motor skills. These impairments were associated with reduced ligand binding densities of NMDA receptors in hippocampal subfields CA3 and the dentate gyrus. The reduced excitation was potentiated by increased ligand binding densities of GABA(A) receptors resulting in a relative shift in favor of inhibition and impaired behavioral performance. These alterations were accompanied by impaired induction of LTP in the CA1 region. Moreover, G-CSF deficiency led to decreased dendritic complexity in hippocampal neurons in the dentate gyrus and the CA1 region. G-CSF deficiency also caused a reduction of neuronal precursor cells in the dentate gyrus. These findings confirm G-CSF as an essential neurotrophic factor, and point to a role in the proliferation, differentiation and functional integration of neural cells necessary for the structural and functional integrity of the hippocampal formation.
Collapse
|
46
|
Lan X, Qu H, Yao W, Zhang C. Granulocyte-colony stimulating factor inhibits neuronal apoptosis in a rat model of diabetic cerebral ischemia. TOHOKU J EXP MED 2009; 216:117-26. [PMID: 18832793 DOI: 10.1620/tjem.216.117] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Incidence of cerebral vascular disease (CVD) is higher in patients with diabetes mellitus (DM) than that in individuals without DM, and neuronal apoptosis determines the severity of cerebral infarction. However, there is no effective therapy for CVD. Granulocyte-colony stimulating factor (G-CSF), a potent hematopoietic factor, could inhibit apoptosis of hematopoietic progenitor cells. However, its effect on neuronal cells is still unclear. In this study, we investigated the anti-apoptosis properties of G-CSF in neurons following focal cerebral ischemia in diabetic rats. The diabetic condition was generated in rats by intravenous injection of streptozotocin. After 6 weeks, diabetic rats underwent middle cerebral artery occlusion (MCAO) and received subcutaneous administration of G-CSF (50 microg/kg) daily for 7, 14 or 21 days. We analyzed the changes in neurological severity scores, infarct volume, number of apoptotic neurons, and the expression of G-CSF receptor, phosphorylated signal transducer and activator of transcription 3 (pSTAT3), cellular inhibitor of apoptosis protein 2 (cIAP2), Bcl-2, and Bax in the brain tissue. Bax is a pro-apoptotic member of the Bcl-2 protein family. The DM rats treated with G-CSF not only showed the reduced infarct volume and decreased apoptosis cell number, but also presented improved neurological scores. The G-CSF also increased the expression of pSTAT3, Bcl-2, and cIAP2 proteins as well as Bcl-2 mRNA, but inhibited Bax protein expression in the brain. These results indicate that G-CSF partially increases neuronal survival by affecting apoptosis pathways. G-CSF provides a potential treatment for stroke and other neurological dysfunction accompanied by neuronal apoptosis.
Collapse
Affiliation(s)
- Xifa Lan
- Department of Neurology, The First Affiliated Hospital, China Medical University, Shenyang, PR China
| | | | | | | |
Collapse
|
47
|
Gautier S, Ouk T, Petrault O, Caron J, Bordet R. Neutrophils contribute to intracerebral haemorrhages after treatment with recombinant tissue plasminogen activator following cerebral ischaemia. Br J Pharmacol 2009; 156:673-9. [PMID: 19210512 DOI: 10.1111/j.1476-5381.2009.00068.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND AND PURPOSE Polymorphonuclear neutrophils (PMNs) contribute to the vascular damage caused by transient cerebral ischaemia. Here we have evaluated the role of PMNs in intracerebral haemorrhage (ICH) induced in a model of thrombolysis with recombinant tissue plasminogen activator (t-PA) during the acute phase of cerebral ischaemia. EXPERIMENTAL APPROACH The middle cerebral artery (MCA) of male spontaneously hypertensive rats was occluded for 1 h followed by reperfusion and, 5 h later, infusion of thrombolytic products (generated in vitro by t-PA on autologous clots). Effects of pretreatment (before the MCA occlusion) with vinblastine (4 days before; 0.5 mg.kg(-1)), monoclonal anti-neutrophil antibody (mAbRP3; 12 h, 0.3 mg.kg(-1)) or saline on ICH, neutrophil infiltration, MCA vascular reactivity and brain infarct volume were assessed, 24 h after the beginning of reperfusion. KEY RESULTS Depletion of circulating neutrophils significantly reduced t-PA-induced ICH (vinblastine, 4.6 +/- 1.0; mAbRP3, 5.2 +/- 1.0 vs. saline, 10.8 +/- 2.7 haemorrhages; P < 0.05). This depletion was associated with a decrease in cerebral infiltration by neutrophils and a decrease of endothelium-dependent, vascular dysfunction in isolated MCA, induced by the ischaemia/reperfusion and t-PA treatment. Brain infarct volume was significantly decreased after vinblastine treatment (159 +/- 13 mm(3) vs. 243 +/- 16 mm(3) with saline; P < 0.01) but not after depletion with mAbRP3 (221 +/- 22 mm(3)). CONCLUSIONS AND IMPLICATIONS Our results showed that pharmacological depletion of PMNs prevented t-PA-induced ICH, in parallel with a decrease in cerebral infiltration by PMNs and a decreased endothelial dysfunction in cerebral blood vessels.
Collapse
Affiliation(s)
- Sophie Gautier
- EA1046, Département de Pharmacologie médicale, Institut de Médecine Prédictive et de Recherche Thérapeutique, Université de Lille 2, Faculté de Médecine, Centre Hospitalier Universitaire, Lille, France
| | | | | | | | | |
Collapse
|
48
|
Orakcioglu B, Becker K, Sakowitz OW, Herweh C, Köhrmann M, Huttner HB, Steiner T, Unterberg A, Schellinger PD. MRI of the perihemorrhagic zone in a rat ICH model: effect of hematoma evacuation. Neurocrit Care 2008; 8:448-55. [PMID: 18188706 DOI: 10.1007/s12028-007-9047-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND Perihemorrhagic pathophysiology of spontaneous intracerebral hemorrhages (ICH) remains unclear. Recently, ischemic changes in the perihemorrhagic zone (PHZ) have been discussed as a potential source of secondary damage. In this study, we focussed on diffusion and perfusion characteristics of experimental ICH. METHODS Experimental ICH was induced with a double injection model in rats. In total, 49 animals were examined at three timepoints within 3.5 h after ICH with a 2.35T animal scanner. We investigated perihemorrhagic relative apparent diffusion coefficients (rADC) and relative mean transit time (rMTT). Animals were divided into 2 groups; controls (gr1, n = 27) and facilitated hematoma evacuation with recombinant tissue plasminogen activator (rt-PA) after the first of 3 imaging time points (gr2, n = 22). Diffusion (rADC) and perfusion (rMTT) characteristics were analyzed in 3 regions of interest surrounding the hematoma (ROI1-3). RESULTS Overall rADC and rMTT values in ROI3 (normal tissue) did not show any changes. There was mild edema--not ischemia--in ROIs1 and 2 at TP1 with rADC of 1.05-1.18 in both groups indicating vasogenic edema (not ischemia). This did not change with hematoma evacuation. There was mild (non-critical) perfusion reduction in ROIs1 and 2 at TP1, which disappeared after clot evacuation in group 2 (P < 0.05 for TP3). Multifactorial ANOVA showed a solid trend (0.06 < P < 0.1) for clot evacuation associated normalization of perfusion in ROIs 1 and 2 within and in between groups 1 and 2. CONCLUSIONS We demonstrated vasogenic edema and mild perfusion reduction in the PHZ above the ischemic threshold. The existence of a perihemorrhagic "penumbra" indicating critically ischemic tissue analogous to ischemic stroke is unlikely.
Collapse
Affiliation(s)
- Berk Orakcioglu
- Department of Neurosurgery, University Hospital Heidelberg, Im Neuenheimer Feld 400, Heidelberg, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Enhanced regeneration in spinal cord injury by concomitant treatment with granulocyte colony-stimulating factor and neuronal stem cells. J Clin Neurosci 2008; 15:656-64. [DOI: 10.1016/j.jocn.2007.03.020] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2006] [Revised: 03/12/2007] [Accepted: 03/13/2007] [Indexed: 01/15/2023]
|
50
|
Granulocyte-colony stimulating factor attenuates striatal degeneration with activating survival pathways in 3-nitropropionic acid model of Huntington's disease. Brain Res 2008; 1194:130-7. [DOI: 10.1016/j.brainres.2007.11.058] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2007] [Revised: 11/24/2007] [Accepted: 11/27/2007] [Indexed: 12/11/2022]
|