1
|
Smith MJ, Penny T, Pham Y, Sutherland AE, Jenkin G, Fahey MC, Paton MCB, Finch-Edmondson M, Miller SL, McDonald CA. Neuroprotective Action of Tacrolimus before and after Onset of Neonatal Hypoxic-Ischaemic Brain Injury in Rats. Cells 2023; 12:2659. [PMID: 37998394 PMCID: PMC10669941 DOI: 10.3390/cells12222659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 11/25/2023] Open
Abstract
(1) Background: Neonatal brain injury can lead to permanent neurodevelopmental impairments. Notably, suppressing inflammatory pathways may reduce damage. To determine the role of neuroinflammation in the progression of neonatal brain injury, we investigated the effect of treating neonatal rat pups with the immunosuppressant tacrolimus at two time points: before and after hypoxic-ischaemic (HI)-induced injury. (2) Methods: To induce HI injury, postnatal day (PND) 10 rat pups underwent single carotid artery ligation followed by hypoxia (8% oxygen, 90 min). Pups received daily tacrolimus (or a vehicle) starting either 3 days before HI on PND 7 (pre-HI), or 12 h after HI (post-HI). Four doses were tested: 0.025, 0.05, 0.1 or 0.25 mg/kg/day. Pups were euthanised at PND 17 or PND 50. (3) Results: All tacrolimus doses administered pre-HI significantly reduced brain infarct size and neuronal loss, increased the number of resting microglia and reduced cellular apoptosis (p < 0.05 compared to control). In contrast, only the highest dose of tacrolimus administered post-HI (0.25 mg/kg/day) reduced brain infarct size (p < 0.05). All doses of tacrolimus reduced pup weight compared to the controls. (4) Conclusions: Tacrolimus administration 3 days pre-HI was neuroprotective, likely mediated through neuroinflammatory and cell death pathways. Tacrolimus post-HI may have limited capacity to reduce brain injury, with higher doses increasing rat pup mortality. This work highlights the benefits of targeting neuroinflammation during the acute injurious period. More specific targeting of neuroinflammation, e.g., via T-cells, warrants further investigation.
Collapse
Affiliation(s)
- Madeleine J. Smith
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; (M.J.S.); (T.P.); (Y.P.); (A.E.S.); (G.J.); (M.C.F.); (S.L.M.)
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia
| | - Tayla Penny
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; (M.J.S.); (T.P.); (Y.P.); (A.E.S.); (G.J.); (M.C.F.); (S.L.M.)
| | - Yen Pham
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; (M.J.S.); (T.P.); (Y.P.); (A.E.S.); (G.J.); (M.C.F.); (S.L.M.)
| | - Amy E. Sutherland
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; (M.J.S.); (T.P.); (Y.P.); (A.E.S.); (G.J.); (M.C.F.); (S.L.M.)
| | - Graham Jenkin
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; (M.J.S.); (T.P.); (Y.P.); (A.E.S.); (G.J.); (M.C.F.); (S.L.M.)
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia
| | - Michael C. Fahey
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; (M.J.S.); (T.P.); (Y.P.); (A.E.S.); (G.J.); (M.C.F.); (S.L.M.)
- Department of Paediatrics, Monash University, Clayton, VIC 3168, Australia
| | - Madison C. B. Paton
- Cerebral Palsy Alliance Research Institute, Speciality of Child and Adolescent Health, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2050, Australia; (M.C.B.P.); (M.F.-E.)
| | - Megan Finch-Edmondson
- Cerebral Palsy Alliance Research Institute, Speciality of Child and Adolescent Health, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2050, Australia; (M.C.B.P.); (M.F.-E.)
| | - Suzanne L. Miller
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; (M.J.S.); (T.P.); (Y.P.); (A.E.S.); (G.J.); (M.C.F.); (S.L.M.)
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia
| | - Courtney A. McDonald
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; (M.J.S.); (T.P.); (Y.P.); (A.E.S.); (G.J.); (M.C.F.); (S.L.M.)
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168, Australia
| |
Collapse
|
2
|
Zhao X, Gu R, Zhao Y, Wei F, Gao X, Zhuang Y, Xiao Z, Shen H, Dai J. Adult spinal cord tissue transplantation combined with local tacrolimus sustained-release collagen hydrogel promotes complete spinal cord injury repair. Cell Prolif 2023; 56:e13451. [PMID: 36916024 DOI: 10.1111/cpr.13451] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/26/2023] [Accepted: 03/03/2023] [Indexed: 03/16/2023] Open
Abstract
The strategy of replacing a completely damaged spinal cord with allogenic adult spinal cord tissues (aSCs) can potentially repair complete spinal cord injury (SCI) in combination with immunosuppressive drugs, such as tacrolimus (Tac), which suppress transplant rejection and improve graft survival. However, daily systemic administration of immunosuppressive agents may cause harsh side effects. Herein, a localized, sustained Tac-release collagen hydrogel (Col/Tac) was developed to maximize the immune regulatory efficacy but minimize the side effects of Tac after aSC transplantation in complete SCI recipients. Thoracic aSCs of rat donors were transplanted into the complete thoracic spinal cord transection rat recipients, after which Col/Tac hydrogel was implanted. The Tac-encapsulated collagen hydrogel exhibited suitable mechanical properties and long-term sustained Tac release behaviour. After Col/Tac hydrogel implantation in SCI rats with aSC transplantation, the recipients' survival rate significantly improved and the side effects on tissues were reduced compared with those with conventional Tac medication. Moreover, treatment with the Col/Tac hydrogel exhibited similarly reduced immune rejection levels by regulating immune responses and promoted neurogenesis compared to daily Tac injections, and thus improved functional restoration. Localized delivery of immunosuppressive agents by the Col/Tac hydrogel may be a promising strategy for overcoming immune rejection of transplants, with significant potential for clinical application in the future.
Collapse
Affiliation(s)
- Xinhao Zhao
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of NanoTech and NanoBionics, Chinese Academy of Sciences, Suzhou, China.,China-Japan Union Hospital of Jilin University, Changchun, China.,State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Rui Gu
- China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Feng Wei
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of NanoTech and NanoBionics, Chinese Academy of Sciences, Suzhou, China.,School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, China
| | - Xu Gao
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of NanoTech and NanoBionics, Chinese Academy of Sciences, Suzhou, China.,China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yan Zhuang
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of NanoTech and NanoBionics, Chinese Academy of Sciences, Suzhou, China.,School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, China
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - He Shen
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of NanoTech and NanoBionics, Chinese Academy of Sciences, Suzhou, China.,School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, China
| | - Jianwu Dai
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of NanoTech and NanoBionics, Chinese Academy of Sciences, Suzhou, China.,State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.,School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, China
| |
Collapse
|
3
|
Dematteis G, Restelli E, Vanella VV, Manfredi M, Marengo E, Corazzari M, Genazzani AA, Chiesa R, Lim D, Tapella L. Calcineurin Controls Cellular Prion Protein Expression in Mouse Astrocytes. Cells 2022; 11:cells11040609. [PMID: 35203261 PMCID: PMC8870693 DOI: 10.3390/cells11040609] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 02/04/2022] [Accepted: 02/08/2022] [Indexed: 02/05/2023] Open
Abstract
Prion diseases arise from the conformational conversion of the cellular prion protein (PrPC) into a self-replicating prion isoform (PrPSc). Although this process has been studied mostly in neurons, a growing body of evidence suggests that astrocytes express PrPC and are able to replicate and accumulate PrPSc. Currently, prion diseases remain incurable, while downregulation of PrPC represents the most promising therapy due to the reduction of the substrate for prion conversion. Here we show that the astrocyte-specific genetic ablation or pharmacological inhibition of the calcium-activated phosphatase calcineurin (CaN) reduces PrPC expression in astrocytes. Immunocytochemical analysis of cultured CaN-KO astrocytes and isolation of synaptosomal compartments from the hippocampi of astrocyte-specific CaN-KO (ACN-KO) mice suggest that PrPC is downregulated both in vitro and in vivo. The downregulation occurs without affecting the glycosylation of PrPC and without alteration of its proteasomal or lysosomal degradation. Direct assessment of the protein synthesis rate and shotgun mass spectrometry proteomics analysis suggest that the reduction of PrPC is related to the impairment of global protein synthesis in CaN-KO astrocytes. When WT-PrP and PrP-D177N, a mouse homologue of a human mutation associated with the inherited prion disease fatal familial insomnia, were expressed in astrocytes, CaN-KO astrocytes showed an aberrant localization of both WT-PrP and PrP-D177N variants with predominant localization to the Golgi apparatus, suggesting that ablation of CaN affects both WT and mutant PrP proteins. These results provide new mechanistic details in relation to the regulation of PrP expression in astrocytes, suggesting the therapeutic potential of astroglial cells.
Collapse
Affiliation(s)
- Giulia Dematteis
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy; (G.D.); (A.A.G.)
| | - Elena Restelli
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (E.R.); (R.C.)
| | - Virginia Vita Vanella
- Department of Translational Medicine, Center for Translational Research on Autoimmune and Allergic Disease (CAAD), Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy; (V.V.V.); (M.M.)
| | - Marcello Manfredi
- Department of Translational Medicine, Center for Translational Research on Autoimmune and Allergic Disease (CAAD), Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy; (V.V.V.); (M.M.)
| | - Emilio Marengo
- Department of Sciences and Technological Innovation, Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy;
| | - Marco Corazzari
- Department of Health Science (DSS), Center for Translational Research on Autoimmune and Allergic Disease (CAAD) & Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy;
| | - Armando A. Genazzani
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy; (G.D.); (A.A.G.)
| | - Roberto Chiesa
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy; (E.R.); (R.C.)
| | - Dmitry Lim
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy; (G.D.); (A.A.G.)
- Correspondence: (D.L.); (L.T.); Tel.: +39-0321-375822 (L.T.)
| | - Laura Tapella
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale “Amedeo Avogadro”, 28100 Novara, Italy; (G.D.); (A.A.G.)
- Correspondence: (D.L.); (L.T.); Tel.: +39-0321-375822 (L.T.)
| |
Collapse
|
4
|
Yang Y, Liu Y, Zhu J, Song S, Huang Y, Zhang W, Sun Y, Hao J, Yang X, Gao Q, Ma Z, Zhang J, Gu X. Neuroinflammation-mediated mitochondrial dysregulation involved in postoperative cognitive dysfunction. Free Radic Biol Med 2022; 178:134-146. [PMID: 34875338 DOI: 10.1016/j.freeradbiomed.2021.12.004] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/20/2021] [Accepted: 12/02/2021] [Indexed: 12/22/2022]
Abstract
Neuroinflammation following peripheral surgery is a pivotal pathogenic mechanism of postoperative cognitive dysfunction (POCD). However, the key site of inflammation-mediated neural damage remains unclear. Impaired mitochondrial function is a vital feature of degenerated neurons. Dynamin-related protein 1 (DRP1), a crucial regulator of mitochondrial dynamics, has been shown to play an essential role in synapse formation. Here, we designed experiments to assess whether Drp1-regulated mitochondrial dynamics and function are involved in the pathological processes of POCD and elucidate its relationship with neuroinflammation. Aged mice were subjected to experimental laparotomy under isoflurane anesthesia. Primary neurons and SH-SY5Y cells were exposed to tumor necrosis factor (TNF). We found an increase in Drp1 activation as well as mitochondrial fragmentation both in the hippocampus of mice after surgery and primary neurons after TNF exposure. Pretreatment with Mdivi-1, a Drp1 specific inhibitor, reduced this mitochondrial fragmentation. Drp1 knockdown with small interfering RNA blocked TNF-induced mitochondrial fragmentation in SH-SY5Y cells. However, the application of Mdivi-1 exhibited a negative impact on mitochondrial function and neurite growth in primary neurons. Calcineurin activity was increased in primary neurons after TNF exposure and contributed to the Drp1 activation. The calcineurin inhibitor FK506 exhibited a Drp1-independent function that mitigated mitochondrial dysfunction. Finally, we found that FK506 pretreatment ameliorated the neurite growth in neurons treated with TNF and the learning ability of mice after surgery. Overall, our research indicated a crucial role of mitochondrial function in the pathological processes of POCD, and neuronal metabolic modulation may represent a novel and important target for POCD.
Collapse
Affiliation(s)
- Yan Yang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, 210008, China
| | - Yue Liu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, 210008, China
| | - Jixiang Zhu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, 210008, China
| | - Shiyu Song
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu Province, 210093, China
| | - Yulin Huang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, 210008, China
| | - Wei Zhang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, 210008, China
| | - Yu'e Sun
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, 210008, China
| | - Jing Hao
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, 210008, China
| | - Xuli Yang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, 210008, China
| | - Qian Gao
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu Province, 210093, China
| | - Zhengliang Ma
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, 210008, China.
| | - Juan Zhang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, 210008, China.
| | - Xiaoping Gu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, 210008, China.
| |
Collapse
|
5
|
Afshari K, Momeni Roudsari N, Lashgari NA, Haddadi NS, Haj-Mirzaian A, Hassan Nejad M, Shafaroodi H, Ghasemi M, Dehpour AR, Abdolghaffari AH. Antibiotics with therapeutic effects on spinal cord injury: a review. Fundam Clin Pharmacol 2020; 35:277-304. [PMID: 33464681 DOI: 10.1111/fcp.12605] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 08/06/2020] [Accepted: 09/08/2020] [Indexed: 12/17/2022]
Abstract
Accumulating evidence indicates that a considerable number of antibiotics exert anti-inflammatory and neuroprotective effects in different central and peripheral nervous system diseases including spinal cord injury (SCI). Both clinical and preclinical studies on SCI have found therapeutic effects of antibiotics from different families on SCI. These include macrolides, minocycline, β-lactams, and dapsone, all of which have been found to improve SCI sequels and complications. These antibiotics may target similar signaling pathways such as reducing inflammatory microglial activity, promoting autophagy, inhibiting neuronal apoptosis, and modulating the SCI-related mitochondrial dysfunction. In this review paper, we will discuss the mechanisms underlying therapeutic effects of these antibiotics on SCI, which not only could supply vital information for investigators but also guide clinicians to consider administering these antibiotics as part of a multimodal therapeutic approach for management of SCI and its complications.
Collapse
Affiliation(s)
- Khashayar Afshari
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, 1419733141, Iran.,Experimental Medicine Research Center, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran.,Department of Dermatology, University of Massachusetts Medical School, Worcester, MA, 01655, USA
| | - Nazanin Momeni Roudsari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, No. 99, Yakhchal, Gholhak, Shariati St., Tehran, P. O. Box: 19419-33111, Iran
| | - Naser-Aldin Lashgari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, No. 99, Yakhchal, Gholhak, Shariati St., Tehran, P. O. Box: 19419-33111, Iran
| | - Nazgol-Sadat Haddadi
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, 1419733141, Iran.,Experimental Medicine Research Center, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran.,Department of Dermatology, University of Massachusetts Medical School, Worcester, MA, 01655, USA
| | - Arvin Haj-Mirzaian
- Experimental Medicine Research Center, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
| | - Malihe Hassan Nejad
- Department of Infectious Diseases, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, 1419733141, Iran
| | - Hamed Shafaroodi
- Experimental Medicine Research Center, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
| | - Mehdi Ghasemi
- Department of Neurology, University of Massachusetts School of Medicine, Worcester, MA, 01655, USA
| | - Ahmad Reza Dehpour
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, 1419733141, Iran.,Experimental Medicine Research Center, Tehran University of Medical Sciences, P.O. Box 13145-784, Tehran, Iran
| | - Amir Hossein Abdolghaffari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, No. 99, Yakhchal, Gholhak, Shariati St., Tehran, P. O. Box: 19419-33111, Iran.,Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, 31375-1369, Iran.,Gastrointestinal Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, 1419733151, Iran
| |
Collapse
|
6
|
Spinal Cord Injury: Pathophysiology, Multimolecular Interactions, and Underlying Recovery Mechanisms. Int J Mol Sci 2020; 21:ijms21207533. [PMID: 33066029 PMCID: PMC7589539 DOI: 10.3390/ijms21207533] [Citation(s) in RCA: 692] [Impact Index Per Article: 138.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/17/2020] [Accepted: 09/24/2020] [Indexed: 12/30/2022] Open
Abstract
Spinal cord injury (SCI) is a destructive neurological and pathological state that causes major motor, sensory and autonomic dysfunctions. Its pathophysiology comprises acute and chronic phases and incorporates a cascade of destructive events such as ischemia, oxidative stress, inflammatory events, apoptotic pathways and locomotor dysfunctions. Many therapeutic strategies have been proposed to overcome neurodegenerative events and reduce secondary neuronal damage. Efforts have also been devoted in developing neuroprotective and neuro-regenerative therapies that promote neuronal recovery and outcome. Although varying degrees of success have been achieved, curative accomplishment is still elusive probably due to the complex healing and protective mechanisms involved. Thus, current understanding in this area must be assessed to formulate appropriate treatment modalities to improve SCI recovery. This review aims to promote the understanding of SCI pathophysiology, interrelated or interlinked multimolecular interactions and various methods of neuronal recovery i.e., neuroprotective, immunomodulatory and neuro-regenerative pathways and relevant approaches.
Collapse
|
7
|
Ahmed T, Zulfiqar A, Arguelles S, Rasekhian M, Nabavi SF, Silva AS, Nabavi SM. Map kinase signaling as therapeutic target for neurodegeneration. Pharmacol Res 2020; 160:105090. [PMID: 32707231 DOI: 10.1016/j.phrs.2020.105090] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 06/14/2020] [Accepted: 07/16/2020] [Indexed: 12/11/2022]
Abstract
Aging is known to be one of the major risk factors in many neurodegenerative diseases (ND) whose prevalence is estimated to rise in the coming years due to the increase in life expectancy. Examples of neurodegenerative diseases include Huntington's, Parkinson's, and Alzheimer's diseases, along with Amyotrophic Lateral Sclerosis, Spinocerebellar ataxias and Frontotemporal Dementia. Given that so far these ND do not have effective pharmacological therapies, a better understanding of the molecular and cellular mechanisms can contribute to development of effective treatments. During the previous decade, the data indicated that dysregulation of MAP kinases [which included c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinase 1 and 2 (ERK1/2), and p38] are associated with several stages of the inflammatory process which in turn contributes to age-related neurodegenerative diseases. This evidence suggests that control of inflammation through regulation of MAP kinase could be a worthwhile approach against neurodegenerative diseases. In this review we summarize the pathways of MAP kinase signal transduction and different pharmacological inhibitors that can be used in its modulation against ND.
Collapse
Affiliation(s)
- Touqeer Ahmed
- Neurobiology Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan.
| | - Abida Zulfiqar
- Neurobiology Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Sandro Arguelles
- Department of Physiology, Faculty of Pharmacy, University of Seville, Seville, Spain.
| | - Mahsa Rasekhian
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyed Fazel Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran; Division of Translational Medicine, Baqiyatallah Hospital, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ana Sanches Silva
- National Institute for Agricultural and Veterinary Research (INIAV), Vila Do Conde, Portugal; Center for Study in Animal Science (CECA), ICETA, University of Oporto, Oporto, Portugal
| | - Seyed Mohammad Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran; Division of Translational Medicine, Baqiyatallah Hospital, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Effect of Different Doses and Times of FK506 on Different Areas of the Hippocampus in the Rat Model of Transient Global Cerebral Ischemia-Reperfusion. Neurol Res Int 2019; 2019:8047672. [PMID: 31467711 PMCID: PMC6699343 DOI: 10.1155/2019/8047672] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 06/26/2019] [Accepted: 07/07/2019] [Indexed: 12/14/2022] Open
Abstract
Background Stroke is a major worldwide problem that is leading to a high mortality rate in humans. Ischemia, as the most common type of stroke, is characterized by tissue damage that can occur due to insufficient blood flow to the brain even for a brief duration, leading to the release of inflammatory factors, cytokines, and free radicals. In this study, we investigated the effective dose and injection time of FK506 as an immunophilin ligand for providing a suitable effect on cells of CA2, CA3, and dentate gyrus of the hippocampus. Methods In this in vivo study, a total of 48 male Wistar rats were divided into nine groups. The ischemia model was induced by the ligation of bilateral common carotid arteries. The doses of FK506 (3, 6, and 10 mg/kg) were administered intravenously (IV) at the beginning of reperfusion, followed by repeated injections (10 mg/kg) at 6, 24, 48, and 72 hours after ischemia, respectively. Brains were removed and prepared for Nissl staining and the TdT-mediated dUTP Nick End Labeling method. Results Data showed that global ischemia did not decrease the number of viable pyramidal cells in CA2 and CA3 regions, but significant differences were observed in the number of viable granular cells and apoptotic bodies in the dentate gyrus between the control and ischemia groups. Repeated doses of 6 mg/kg of FK506 at an interval of 48 hours were deemed to be the suitable dose and best time of injection. Conclusions It seems that FK506 can ameliorate the extent of apoptosis and may be a good candidate for the treatment of ischemia-induced brain damage.
Collapse
|
9
|
Yin Y, Xiao G, Zhang K, Ying G, Xu H, De Melo BAG, Li S, Liu F, Yetisen AK, Jiang N. Tacrolimus- and Nerve Growth Factor-Treated Allografts for Neural Tissue Regeneration. ACS Chem Neurosci 2019; 10:1411-1419. [PMID: 30525428 DOI: 10.1021/acschemneuro.8b00452] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Treatment of injured peripheral nerves, especially long-distance nerve defects, remains a significant challenge in regenerative medicine due to complex biological conditions and a lack of biomaterials for effective nerve reconstruction. Without proper treatment, nerve injury leads to motor and sensory dysfunction. Here, we have developed an efficacious nerve allograft treated with a dual drug containing acrolimus and nerve growth factor to bridge the nerve gap and achieve rapid neural tissue recovery without immunological rejection. The recovery of the structure, activity, and function of rats treated with the dual drug-treated allograft was investigated by walking track analysis and electrophysiological measurement. The sciatic functional index was measured to be -3.0 after a 12-week treatment. The nerve conduction velocity, peak latency, and peak amplitude of the nerve action potentials demonstrate the functional recovery of the nerve. To study the synergistic effect of the dual drug on the growth of neurites, a neural cell hypoxia model was created. The dual drug exhibited a high efficiency in promoting the growth of nerve cells under the nerve injury-induced hypoxic condition. The dual drug could protect the cells against antioxidative damage from hypoxia by the expression of heat shock protein, hypoxia-inducible factor, β-tubulin, and vimentin.
Collapse
Affiliation(s)
- Yixia Yin
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, China
- Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Gao Xiao
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts 02138, United States
- College of Environment and Resources, Fuzhou University, Fuzhou 350108, China
| | - Kaiming Zhang
- Department of Orthopedics, Second Hospital of Yueyang, Yueyang 414000, China
| | - Guoliang Ying
- Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- School of Materials Science and Engineering, Wuhan Institute of Technology, Wuhan 430205, China
| | - Haixing Xu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, China
| | - Bruna A. G. De Melo
- Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Shipu Li
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, China
| | - Fang Liu
- Department of Orthopedics, Second Hospital of Yueyang, Yueyang 414000, China
| | - Ali K. Yetisen
- Department of Chemical Engineering, Imperial College London, London SW7 2AZ, U.K
| | - Nan Jiang
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts 02138, United States
| |
Collapse
|
10
|
Yuan H, Hu Y, Jiang L, Wang T. The research progress of miRNA/lncRNA associated with spinal cord injury. IBRAIN 2019. [DOI: 10.1002/j.2769-2795.2019.tb00042.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Hao Yuan
- Department of Spinal SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Yue Hu
- Department of Anesthesiology and Institute of Neurological DiseaseTranslational Neuroscience Center, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Ling Jiang
- Department of Anesthesiology and Institute of Neurological DiseaseTranslational Neuroscience Center, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Ting‐Hua Wang
- Department of Anesthesiology and Institute of Neurological DiseaseTranslational Neuroscience Center, West China Hospital, Sichuan UniversityChengduSichuanChina
- Institute of Neuroscience, Kunming Medical UniversityKunmingYunnanChina
| |
Collapse
|
11
|
Mizuma A, Kim JY, Kacimi R, Stauderman K, Dunn M, Hebbar S, Yenari MA. Microglial Calcium Release-Activated Calcium Channel Inhibition Improves Outcome from Experimental Traumatic Brain Injury and Microglia-Induced Neuronal Death. J Neurotrauma 2018; 36:996-1007. [PMID: 30351197 DOI: 10.1089/neu.2018.5856] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Store-operated Ca2+ entry (SOCE) mediated by calcium release-activated calcium (CRAC) channels contributes to calcium signaling. The resulting intracellular calcium increases activate calcineurin, which in turn activates immune transcription factor nuclear factor of activated T cells (NFAT). Microglia contain CRAC channels, but little is known whether these channels play a role in acute brain insults. We studied a novel CRAC channel inhibitor to explore the therapeutic potential of this compound in microglia-mediated injury. Cultured microglial BV2 cells were activated by Toll-like receptor agonists or IFNγ. Some cultures were treated with a novel CRAC channel inhibitor (CM-EX-137). Western blots revealed the presence of CRAC channel proteins STIM1 and Orai1 in BV2 cells. CM-EX-137 decreased nitric oxide (NO) release and inducible nitric oxide synthase (iNOS) expression in activated microglia and reduced agonist-induced intracellular calcium accumulation in microglia, while suppressing inflammatory transcription factors nuclear factor kappa B (NF-κB) and nuclear factor of activated T cells (NFAT). Male C57/BL6 mice exposed to experimental brain trauma and treated with CM-EX-137 had decreased lesion size, brain hemorrhage, and improved neurological deficits with decreased microglial activation, iNOS and Orai1 and STIM1 levels. We suggest a novel anti-inflammatory approach for managing acute brain injury. Our observations also shed light on new calcium signaling pathways not described previously in brain injury models.
Collapse
Affiliation(s)
- Atsushi Mizuma
- 1 Department of Neurology, University of California, San Francisco; the San Francisco VA Medical Center, San Francisco, California.,2 Department of Neurology, Tokai University School of Medicine, Isehara, Japan
| | - Jong Youl Kim
- 1 Department of Neurology, University of California, San Francisco; the San Francisco VA Medical Center, San Francisco, California.,3 Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea
| | - Rachid Kacimi
- 1 Department of Neurology, University of California, San Francisco; the San Francisco VA Medical Center, San Francisco, California
| | | | | | | | - Midori A Yenari
- 1 Department of Neurology, University of California, San Francisco; the San Francisco VA Medical Center, San Francisco, California
| |
Collapse
|
12
|
Panahi Y, Mojtahedzadeh M, Najafi A, Rajaee SM, Torkaman M, Sahebkar A. Neuroprotective Agents in the Intensive Care Unit: -Neuroprotective Agents in ICU. J Pharmacopuncture 2018; 21:226-240. [PMID: 30652049 PMCID: PMC6333194 DOI: 10.3831/kpi.2018.21.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 08/09/2018] [Accepted: 11/14/2018] [Indexed: 01/31/2023] Open
Abstract
Neuroprotection or prevention of neuronal loss is a complicated molecular process that is mediated by various cellular pathways. Use of different pharmacological agents as neuroprotectants has been reported especially in the last decades. These neuroprotective agents act through inhibition of inflammatory processes and apoptosis, attenuation of oxidative stress and reduction of free radicals. Control of this injurious molecular process is essential to the reduction of neuronal injuries and is associated with improved functional outcomes and recovery of the patients admitted to the intensive care unit. This study reviews neuroprotective agents and their mechanisms of action against central nervous system damages.
Collapse
Affiliation(s)
- Yunes Panahi
- Clinical Pharmacy Department, Faculty of Pharmacy, Baqiyatallah University of Medical Sciences, Tehran,
Iran
- Research Center for Rational Use of Drugs, Tehran University of Medical Sciences, Tehran,
Iran
| | - Mojtaba Mojtahedzadeh
- Research Center for Rational Use of Drugs, Tehran University of Medical Sciences, Tehran,
Iran
- Department of Anesthesiology and Critical Care Medicine, Faculty of Medicine, Sina Hospital, Tehran University of Medical Sciences, Tehran,
Iran
| | - Atabak Najafi
- Gastrointestinal Pharmacology Interest Group(GPIG), Universal Scientific Education and Research Network(USERN), Tehran,
Iran
| | - Seyyed Mahdi Rajaee
- Gastrointestinal Pharmacology Interest Group(GPIG), Universal Scientific Education and Research Network(USERN), Tehran,
Iran
| | - Mohammad Torkaman
- Department of Pediatrics, School of Medicine, Baqiyatallah University of Medical Sciences, Tehran,
Iran
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad,
Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad,
Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad,
Iran
| |
Collapse
|
13
|
Calcineurin/P-ERK/Egr-1 Pathway is Involved in Fear Memory Impairment after Isoflurane Exposure in Mice. Sci Rep 2017; 7:13947. [PMID: 29066839 PMCID: PMC5654981 DOI: 10.1038/s41598-017-13975-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 10/03/2017] [Indexed: 11/28/2022] Open
Abstract
Isoflurane exposure adversely influences subsequent fear memory formation in mice. Calcineurin (CaN), a phosphatase, prevents the establishment of emotional memory by dephosphorylating substrates and inhibiting the expression of learning and memory related genes. We investigated whether isoflurane impairment of fear memory formation was associated with altered CaN activity and downstream phosphorylated-extracellular signal-regulated kinases (p-ERK) and early growth response gene-1 (Egr-1) expression in hippocampus and amygdala. We also tested whether memory performance can be rescued by the CaN inhibitor FK506. Adult C57BL/6 mice were injected FK506 or vehicle after being exposed to 1.3% isoflurane or air for 1 h. After a 1 h- recovery, mice underwent classical fear conditioning (FC) training. Fear memory were tested 30 min, 48 h and 7 days after training. The activity of CaN, and expression of p-ERK and Egr-1 in hippocampus and amygdala were analyzed. Isoflurane exposure reduced mice freezing time in contextual and tone FC tests 30 min and 48 h after training. Hippocampus and amygdala from isoflurane-exposed mice had enhanced CaN activity, reduced p-ERK/ERK and Egr-1 expression. All these changes in isoflurane-exposed mice were attenuated by FK506 treatment. These results indicate calcineurin/p-ERK/Egr-1 Pathway is involved in fear memory impairment after isoflurane exposure in mice.
Collapse
|
14
|
Liu G, Fan G, Guo G, Kang W, Wang D, Xu B, Zhao J. FK506 Attenuates the Inflammation in Rat Spinal Cord Injury by Inhibiting the Activation of NF-κB in Microglia Cells. Cell Mol Neurobiol 2017; 37:843-855. [PMID: 27572744 PMCID: PMC11482064 DOI: 10.1007/s10571-016-0422-8] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 08/23/2016] [Indexed: 02/08/2023]
Abstract
FK-506 (Tacrolimus) is a very commonly used immunomodulatory agent that plays important roles in modulating the calcium-dependent phosphoserine-phosphothreonine protein phosphatase calcineurin and thus inhibits calcineurin-mediated secondary neuronal damage. The biological function of FK-506 in the spinal cord has not been fully elucidated. To clarify the anti-inflammatory action of FK-506 in spinal cord injury (SCI), we performed an acute spinal cord contusion injury model in adult rats and hypoxia-treated primary spinal cord microglia cultures. This work studied the activation of NF-κB and proinflammatory cytokine (TNF-a, IL-1b, and IL-6) expression. ELISA and q-PCR analysis revealed that TNF-a, IL-1b, and IL-6 levels significantly increased 3 days after spinal cord contusion and decreased after 14 days, accompanied by the increased activation of NF-κB. This increase was reversed by an FK-506 treatment. Double immunofluorescence labeling suggested that NF-κB activation was especially prominent in microglia. Immunohistochemistry confirmed no alteration in the number of microglia. Moreover, the results in hypoxia-treated primary spinal cord microglia confirmed the effect of FK-506 on TNF-a, IL-1b, and IL-6 expression and NF-κB activation. These findings suggest that FK-506 may be involved in microglial activation after SCI.
Collapse
Affiliation(s)
- Gang Liu
- Department of Orthopedic Surgery, Jinling Hospital, 305# East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Gentao Fan
- Department of Orthopedic Surgery, Jinling Hospital, 305# East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Guodong Guo
- Department of Orthopedic Surgery, Jinling Hospital, 305# East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Wenbo Kang
- Department of Orthopedic Surgery, Jinling Hospital, 305# East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Dongsheng Wang
- Department of Orthopedic Surgery, Jinling Hospital, 305# East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Bin Xu
- Department of Orthopedic Surgery, Jinling Hospital, 305# East Zhongshan Road, Nanjing, 210002, Jiangsu, China
| | - Jianning Zhao
- Department of Orthopedic Surgery, Jinling Hospital, 305# East Zhongshan Road, Nanjing, 210002, Jiangsu, China.
| |
Collapse
|
15
|
Fields JA, Overk C, Adame A, Florio J, Mante M, Pineda A, Desplats P, Rockenstein E, Achim C, Masliah E. Neuroprotective effects of the immunomodulatory drug FK506 in a model of HIV1-gp120 neurotoxicity. J Neuroinflammation 2016; 13:120. [PMID: 27220536 PMCID: PMC4879748 DOI: 10.1186/s12974-016-0585-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 05/16/2016] [Indexed: 11/12/2022] Open
Abstract
Background HIV-associated neurocognitive disorders (HAND) continue to be a common morbidity associated with chronic HIV infection. It has been shown that HIV proteins (e.g., gp120) released from infected microglial/macrophage cells can cause neuronal damage by triggering inflammation and oxidative stress, activating aberrant kinase pathways, and by disrupting mitochondrial function and biogenesis. Previous studies have shown that FK506, an immunophilin ligand that modulates inflammation and mitochondrial function and inhibits calcineurin, is capable of rescuing the neurodegenerative pathology in models of Parkinson’s disease, Alzheimer’s disease, and Huntington’s disease. In this context, the main objective of this study was to evaluate if FK506 could rescue the neuronal degeneration and mitochondrial alterations in a transgenic (tg) animal model of HIV1-gp120 neurotoxicity. Methods GFAP-gp120 tg mice were treated with FK506 and analyzed for neuropathology, behavior, mitochondrial markers, and calcium flux by two-photon microscopy. Results We found that FK506 reduced the neuronal cell loss and neuro-inflammation in the gp120 tg mice. Moreover, while vehicle-treated gp120 tg mice displayed damaged mitochondria and increased neuro-inflammatory markers, FK506 rescued the morphological mitochondrial alterations and neuro-inflammation while increasing levels of optic atrophy 1 and mitofusin 1. By two-photon microscopy, calcium levels were not affected in the gp120 tg mice and no effects of FK506 were detected. However, at a functional level, FK506 ameliorated the gp120 tg mice hyperactivity in the open field. Conclusions Together, these results suggest that FK506 might be potentially neuroprotective in patients with HAND by mitigating inflammation and mitochondrial alterations.
Collapse
Affiliation(s)
- Jerel A Fields
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Cassia Overk
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Anthony Adame
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Jazmin Florio
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Michael Mante
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Andrea Pineda
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Paula Desplats
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Edward Rockenstein
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Cristian Achim
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Eliezer Masliah
- Department of Pathology, University of California San Diego, La Jolla, CA, USA. .,Department of Neurosciences, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
16
|
Ahmad M, Zakaria A, Almutairi KM. Effectiveness of minocycline and FK506 alone and in combination on enhanced behavioral and biochemical recovery from spinal cord injury in rats. Pharmacol Biochem Behav 2016; 145:45-54. [PMID: 27106204 DOI: 10.1016/j.pbb.2016.04.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 04/15/2016] [Accepted: 04/18/2016] [Indexed: 11/15/2022]
Abstract
Injury to the spinal cord results in immediate physical damage (primary injury) followed by a prolonged posttraumatic inflammatory disorder (secondary injury). The present study aimed to investigate the neuroprotective effects of minocycline and FK506 (Tacrolimus) individually and in combination on recovery from experimental spinal cord injury (SCI). Young adult male rats were subjected to experimental SCI by weight compression method. Minocycline (50mg/kg) and FK506 (1mg/kg) were administered orally in combination and individually to the SCI group daily for three weeks. During these three weeks, the recovery was measured using behavioral motor parameters (including BBB, Tarlov and other scorings) every other day for 29days after SCI. Thereafter, the animals were sacrificed and the segment of the spinal cord centered at the injury site was removed for the histopathological studies as well as for biochemical analysis of monoamines such as 5-hydroxytryptamine (5-HT) and 5-hydroxy-indolacetic acid (5-HIAA) and some oxidative stress indices, such as thiobarbituric acid-reactive substances (TBARS), total glutathione (GSH) and myeloperoxidase (MPO). All behavioral results indicated that both drugs induced significant recovery from SCI with respect to time. The biochemical and histopathological results supported the behavioral findings, revealing significant recovery in the regeneration of the injured spinal tissues, the monoamine levels, and the oxidative stress indices. Overall, the effects of the tested drugs for SCI recovery were as follows: FK506+minocycline>minocycline>FK506 in all studied parameters. Thus, minocycline and FK506 may prove to be a potential therapy cocktail to treat acute SCI. However, further studies are warranted.
Collapse
Affiliation(s)
- Mohammad Ahmad
- Department of Medical Surgical Nursing, College of Nursing, King Saud University, Riyadh, Saudi Arabia.
| | - Abdulrahim Zakaria
- Department of Rehabilitation Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Khalid M Almutairi
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
17
|
Setkowicz Z, Kosonowska E, Kaczyńska M, Gzieło-Jurek K, Janeczko K. Physical training decreases susceptibility to pilocarpine-induced seizures in the injured rat brain. Brain Res 2016; 1642:20-32. [PMID: 26972533 DOI: 10.1016/j.brainres.2016.03.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 01/27/2016] [Accepted: 03/08/2016] [Indexed: 10/22/2022]
Abstract
There is growing evidence that physical activity ameliorates the course of epilepsy in animal models as well as in clinical conditions. Since traumatic brain injury is one of the strongest determinants of epileptogenesis, the present study focuses on the question whether a moderate long-term physical training can decrease susceptibility to seizures evoked following brain damage. Wistar rats received a mechanical brain injury and were subjected to daily running sessions on a treadmill for 21 days. Thereafter, seizures were induced by pilocarpine injections in trained and non-trained, control groups. During the acute period of status epilepticus, the intensity of seizures was assessed within the six-hour observation period. The trained rats showed considerable amelioration of pilocarpine-induced motor symptoms when compared with their non-trained counterparts. Histological investigations of effects of the brain injury and of physical training detected significant quantitative changes in parvalbumin-, calretinin- and NPY-immunopositive neuronal populations. Some of the injury-induced changes, especially those shoved by parvalbumin-immunopositive neurons, were abolished by the subsequent physical training procedure and could, therefore, be considered as neuronal correlates of the observed functional amelioration of the injured brain.
Collapse
Affiliation(s)
- Zuzanna Setkowicz
- Department of Neuroanatomy, Institute of Zoology, Jagiellonian University, 9 Gronostajowa St., 30-387 Kraków, Poland
| | - Emilia Kosonowska
- Department of Neuroanatomy, Institute of Zoology, Jagiellonian University, 9 Gronostajowa St., 30-387 Kraków, Poland
| | - Małgorzata Kaczyńska
- Department of Neuroanatomy, Institute of Zoology, Jagiellonian University, 9 Gronostajowa St., 30-387 Kraków, Poland
| | - Kinga Gzieło-Jurek
- Department of Neuroanatomy, Institute of Zoology, Jagiellonian University, 9 Gronostajowa St., 30-387 Kraków, Poland
| | - Krzysztof Janeczko
- Department of Neuroanatomy, Institute of Zoology, Jagiellonian University, 9 Gronostajowa St., 30-387 Kraków, Poland.
| |
Collapse
|
18
|
Torres-Espín A, Redondo-Castro E, Hernandez J, Navarro X. Immunosuppression of allogenic mesenchymal stem cells transplantation after spinal cord injury improves graft survival and beneficial outcomes. J Neurotrauma 2015; 32:367-80. [PMID: 25203134 DOI: 10.1089/neu.2014.3562] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Cell therapy for spinal cord injury (SCI) is a promising strategy for clinical application. Mesenchymal stem cells (MSC) have demonstrated beneficial effects following transplantation in animal models of SCI. However, despite the immunoprivilege properties of the MSC, their survival in the injured spinal cord is reduced due to the detrimental milieu in the damaged tissue and immune rejection of the cells. The limited survival of the engrafted cells may determine the therapy success. Therefore, we compared two strategies to increase the presence of the cells in the injured spinal cord in rats: increasing the amount of MSC transplants and using immunosuppressive treatment with FK506 after transplantation. Functional outcomes for locomotion and electrophysiological responses were assessed. The grafted cells survival and the amount of cavity and spared tissue were studied. The findings indicate that immunosuppression improved grafted cells survival. A cell-dose effect was found regarding locomotion recovery and tissue protection independent of immunosuppression. Nevertheless, immunosuppression enhanced the electrophysiological outcomes and allowed filling of the cavity formed after injury by new regenerative tissue and axons. These results indicate that MSC transplantation combined with immunosuppression prolongs the survival of engrafted cells and improves functional and morphological outcomes after SCI.
Collapse
Affiliation(s)
- Abel Torres-Espín
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona , and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Bellaterra, Spain
| | | | | | | |
Collapse
|
19
|
Jóźwiak-Bębenista M, Kowalczyk E, Nowak JZ. The cyclic AMP effects and neuroprotective activities of PACAP and VIP in cultured astrocytes and neurons exposed to oxygen-glucose deprivation. Pharmacol Rep 2014; 67:332-8. [PMID: 25712659 DOI: 10.1016/j.pharep.2014.10.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 09/24/2014] [Accepted: 10/02/2014] [Indexed: 01/30/2023]
Abstract
BACKGROUND Pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal peptide (VIP) are endogenous peptides, widely expressed in the central and peripheral nervous system. The adenylyl cyclase (AC)/cyclic AMP (cAMP) is their main intracellular signal transduction pathway. Numerous data suggest that PACAP and VIP have considerable neuroprotective potential, indicating the possibility for their use as new therapeutic strategies in stroke treatment. The aim of this study was to evaluate the effect of oxygen-glucose deprivation (OGD) - an established in vitro model for ischemic cell stress - on PACAP and VIP-evoked receptor-mediated cAMP generation in glial and neuronal cells, and to determine whether PACAP and VIP have neuroprotective activity under these conditions. METHODS The formation of [(3)H]cAMP by PACAP, VIP and forskolin (a direct activator of AC) was measured in [(3)H]adenine prelabeled primary rat glial and neuronal cells under normoxia and OGD conditions. The effects of PACAP and VIP on cell viability were measured using the MTT conversion method, and were compared to tacrolimus (FK506), a well known neuroprotective agent. RESULTS The OGD model inhibited the PACAP and VIP-induced cAMP formation in rat astrocytes and neurons. Incubation of neuronal cells with PACAP prevented OGD-induced cell death, more efficiently than VIP and FK506. CONCLUSION The obtained results showed that hypoxia/ischemia may trigger down-regulation of the brain AC-coupled PACAP/VIP receptors, with a consequent decrease of PACAP- and/or VIP-ergic-dependent cAMP-driven signaling. Moreover, our findings indicate that PACAP and VIP can prevent the deleterious effect of OGD on rat neuronal cells.
Collapse
Affiliation(s)
- Marta Jóźwiak-Bębenista
- Department of Pharmacology and Toxicology, The Interfaculty Chair of Basic and Clinical Pharmacology, Medical University of Lodz, Łódź, Poland.
| | - Edward Kowalczyk
- Department of Pharmacology and Toxicology, The Interfaculty Chair of Basic and Clinical Pharmacology, Medical University of Lodz, Łódź, Poland
| | - Jerzy Z Nowak
- Department of Pharmacology and Toxicology, The Interfaculty Chair of Basic and Clinical Pharmacology, Medical University of Lodz, Łódź, Poland
| |
Collapse
|
20
|
Diaz-Arrastia R, Kochanek PM, Bergold P, Kenney K, Marx CE, Grimes CJB, Loh LTCY, Adam LTCGE, Oskvig D, Curley KC, Salzer W. Pharmacotherapy of traumatic brain injury: state of the science and the road forward: report of the Department of Defense Neurotrauma Pharmacology Workgroup. J Neurotrauma 2014; 31:135-58. [PMID: 23968241 DOI: 10.1089/neu.2013.3019] [Citation(s) in RCA: 193] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Despite substantial investments by government, philanthropic, and commercial sources over the past several decades, traumatic brain injury (TBI) remains an unmet medical need and a major source of disability and mortality in both developed and developing societies. The U.S. Department of Defense neurotrauma research portfolio contains more than 500 research projects funded at more than $700 million and is aimed at developing interventions that mitigate the effects of trauma to the nervous system and lead to improved quality of life outcomes. A key area of this portfolio focuses on the need for effective pharmacological approaches for treating patients with TBI and its associated symptoms. The Neurotrauma Pharmacology Workgroup was established by the U.S. Army Medical Research and Materiel Command (USAMRMC) with the overarching goal of providing a strategic research plan for developing pharmacological treatments that improve clinical outcomes after TBI. To inform this plan, the Workgroup (a) assessed the current state of the science and ongoing research and (b) identified research gaps to inform future development of research priorities for the neurotrauma research portfolio. The Workgroup identified the six most critical research priority areas in the field of pharmacological treatment for persons with TBI. The priority areas represent parallel efforts needed to advance clinical care; each requires independent effort and sufficient investment. These priority areas will help the USAMRMC and other funding agencies strategically guide their research portfolios to ensure the development of effective pharmacological approaches for treating patients with TBI.
Collapse
Affiliation(s)
- Ramon Diaz-Arrastia
- 1 Department of Neurology, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Xu J, Lacoske MH, Theodorakis EA. Neurotrophic natural products: chemistry and biology. Angew Chem Int Ed Engl 2014; 53:956-87. [PMID: 24353244 PMCID: PMC3945720 DOI: 10.1002/anie.201302268] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Indexed: 12/12/2022]
Abstract
Neurodegenerative diseases and spinal cord injury affect approximately 50 million people worldwide, bringing the total healthcare cost to over 600 billion dollars per year. Nervous system growth factors, that is, neurotrophins, are a potential solution to these disorders, since they could promote nerve regeneration. An average of 500 publications per year attests to the significance of neurotrophins in biomedical sciences and underlines their potential for therapeutic applications. Nonetheless, the poor pharmacokinetic profile of neurotrophins severely restricts their clinical use. On the other hand, small molecules that modulate neurotrophic activity offer a promising therapeutic approach against neurological disorders. Nature has provided an impressive array of natural products that have potent neurotrophic activities. This Review highlights the current synthetic strategies toward these compounds and summarizes their ability to induce neuronal growth and rehabilitation. It is anticipated that neurotrophic natural products could be used not only as starting points in drug design but also as tools to study the next frontier in biomedical sciences: the brain activity map project.
Collapse
Affiliation(s)
- Jing Xu
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0358 (USA), Homepage: http://theodorakisgroup.ucsd.edu
| | - Michelle H. Lacoske
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0358 (USA), Homepage: http://theodorakisgroup.ucsd.edu
| | - Emmanuel A. Theodorakis
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0358 (USA), Homepage: http://theodorakisgroup.ucsd.edu
| |
Collapse
|
22
|
Xu J, Lacoske MH, Theodorakis EA. Neurotrophe Naturstoffe - ihre Chemie und Biologie. Angew Chem Int Ed Engl 2013. [DOI: 10.1002/ange.201302268] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
23
|
Chen G, Zhang Z, Wang S, Lv D. Combined treatment with FK506 and nerve growth factor for spinal cord injury in rats. Exp Ther Med 2013; 6:868-872. [PMID: 24137280 PMCID: PMC3797285 DOI: 10.3892/etm.2013.1254] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Accepted: 07/17/2013] [Indexed: 01/21/2023] Open
Abstract
Following spinal cord injury in rats, FK506 is able to protect local nerve tissue, promote neural regeneration, reduce neuronal apoptosis and accelerate the recovery of spinal cord functions. Nerve growth factor (NGF) is important in the regulation of central and peripheral nerve cell regeneration, growth differentiation and functions. Previous studies have shown that FK506 and NGF exhibit a synergistic effect in the treatment of peripheral nerve injury; however, it remains unclear whether the synergistic effect is present in the treatment of spinal cord injury. In this study, we combined FK506 and NGF for the treatment of spinal cord injury in rats. The NF200 protein expression in rats with spinal cord injury was determined using immunohistochemical staining and NF200 mRNA expression levels were observed using the reverse transcription-polymerase chain reaction method. The restoration of spinal cord functions was evaluated using the Basso, Beattie and Bresnahan score. The results demonstrated that the combined treatment significantly enhanced the expression of NF200 and improved spinal cord functions compared with the results of the single treatment. Our experimental observations indicated that FK506 and NGF exhibit a synergistic effect in the treatment of spinal cord injury in rats and that the combined treatment may effectively promote neural regeneration and functional recovery in rats following spinal cord injury.
Collapse
Affiliation(s)
- Guang Chen
- Department of Orthopedic Surgery, First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | | | | | | |
Collapse
|