1
|
Sapolsky R. 2022 ISPNE Bruce McEwen Lifetime Achievement award: Stress, from molecules to societies. Psychoneuroendocrinology 2023; 154:106274. [PMID: 37163880 DOI: 10.1016/j.psyneuen.2023.106274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
The International Society for Psychoneuroendocrinology meeting in Chicago in 2022 was thrilled to recognize Dr. Robert Sapolsky with the Bruce McEwen Lifetime Achievement award. This is the second year for the award to be named to honor Bruce McEwen and it marks the completion of a special issue edited by Blazej Miziak and Robert Paul Juster in the journal Psychoneuroendocrinology dedicated to Bruce's legacy and the unfathomable contribution of Allostatic Load to the stress field. Yet, as our award winner writes, Bruce's legacy is more than scientific as he was well known for mentorship and being an exemplary person, theorist, and scientist. Perhaps understandably for a career favored by humble introverts and shy reclusives, the science shines in the spotlight and personal reflections are cut to accommodate word count limits. For scholars entering the field, stargazing at larger than life luminaries in the field is thrilling yet intimidating as it feels impossible that these experts have the same doubts and distractions as the rest of us primates. Thus, Psychoneuroendocrinology is thrilled to kick off the first perspectives piece in the Cell to Selves series with Dr. Robert Sapolsky sharing that, like his Baboon troops in Kenya, he too sometimes has a bad-hair day. This paper is a written version of a lecture I gave on September 8th, 2022, when receiving the first Bruce McEwen Lifetime Achievement Award from the ISPNE. This was a bittersweet honor; Bruce was my graduate advisor at Rockefeller University and over the next forty years, he was my mentor, teacher and father figure. His death in 2020 left a hole in my life.
Collapse
Affiliation(s)
- Robert Sapolsky
- Departments of Biology, Neurology and Neurosurgery, Stanford University, Stanford, CA 94305-5020, USA.
| |
Collapse
|
2
|
Vannini E, Olimpico F, Middei S, Ammassari-Teule M, de Graaf EL, McDonnell L, Schmidt G, Fabbri A, Fiorentini C, Baroncelli L, Costa M, Caleo M. Electrophysiology of glioma: a Rho GTPase-activating protein reduces tumor growth and spares neuron structure and function. Neuro Oncol 2016; 18:1634-1643. [PMID: 27298309 DOI: 10.1093/neuonc/now114] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 04/22/2016] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Glioblastomas are the most aggressive type of brain tumor. A successful treatment should aim at halting tumor growth and protecting neuronal cells to prevent functional deficits and cognitive deterioration. Here, we exploited a Rho GTPase-activating bacterial protein toxin, cytotoxic necrotizing factor 1 (CNF1), to interfere with glioma cell growth in vitro and vivo. We also investigated whether this toxin spares neuron structure and function in peritumoral areas. METHODS We performed a microarray transcriptomic and in-depth proteomic analysis to characterize the molecular changes triggered by CNF1 in glioma cells. We also examined tumor cell senescence and growth in vehicle- and CNF1-treated glioma-bearing mice. Electrophysiological and morphological techniques were used to investigate neuronal alterations in peritumoral cortical areas. RESULTS Administration of CNF1 triggered molecular and morphological hallmarks of senescence in mouse and human glioma cells in vitro. CNF1 treatment in vivo induced glioma cell senescence and potently reduced tumor volumes. In peritumoral areas of glioma-bearing mice, neurons showed a shrunken dendritic arbor and severe functional alterations such as increased spontaneous activity and reduced visual responsiveness. CNF1 treatment enhanced dendritic length and improved several physiological properties of pyramidal neurons, demonstrating functional preservation of the cortical network. CONCLUSIONS Our findings demonstrate that CNF1 reduces glioma volume while at the same time maintaining the physiological and structural properties of peritumoral neurons. These data indicate a promising strategy for the development of more effective antiglioma therapies.
Collapse
Affiliation(s)
- Eleonora Vannini
- CNR Neuroscience Institute, Pisa, Italy (E.V., F.O., L.B., M.C., Mat.C.); CNR Cellular Biology and Neurobiology Institute, Rome, Italy (S.M., M.A.-T.); Fondazione Pisana per la Scienza, Mass Spectrometry and Proteomics, Pisa, Italy (E.L.d.G., L.M.); Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Freiburg, Germany (G.S.); Istituto Superiore di Sanità, Rome, Italy (A.F., C.F.); Scuola Normale Superiore, Pisa, Italy (M.C., Mat.C.)
| | - Francesco Olimpico
- CNR Neuroscience Institute, Pisa, Italy (E.V., F.O., L.B., M.C., Mat.C.); CNR Cellular Biology and Neurobiology Institute, Rome, Italy (S.M., M.A.-T.); Fondazione Pisana per la Scienza, Mass Spectrometry and Proteomics, Pisa, Italy (E.L.d.G., L.M.); Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Freiburg, Germany (G.S.); Istituto Superiore di Sanità, Rome, Italy (A.F., C.F.); Scuola Normale Superiore, Pisa, Italy (M.C., Mat.C.)
| | - Silvia Middei
- CNR Neuroscience Institute, Pisa, Italy (E.V., F.O., L.B., M.C., Mat.C.); CNR Cellular Biology and Neurobiology Institute, Rome, Italy (S.M., M.A.-T.); Fondazione Pisana per la Scienza, Mass Spectrometry and Proteomics, Pisa, Italy (E.L.d.G., L.M.); Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Freiburg, Germany (G.S.); Istituto Superiore di Sanità, Rome, Italy (A.F., C.F.); Scuola Normale Superiore, Pisa, Italy (M.C., Mat.C.)
| | - Martine Ammassari-Teule
- CNR Neuroscience Institute, Pisa, Italy (E.V., F.O., L.B., M.C., Mat.C.); CNR Cellular Biology and Neurobiology Institute, Rome, Italy (S.M., M.A.-T.); Fondazione Pisana per la Scienza, Mass Spectrometry and Proteomics, Pisa, Italy (E.L.d.G., L.M.); Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Freiburg, Germany (G.S.); Istituto Superiore di Sanità, Rome, Italy (A.F., C.F.); Scuola Normale Superiore, Pisa, Italy (M.C., Mat.C.)
| | - Erik L de Graaf
- CNR Neuroscience Institute, Pisa, Italy (E.V., F.O., L.B., M.C., Mat.C.); CNR Cellular Biology and Neurobiology Institute, Rome, Italy (S.M., M.A.-T.); Fondazione Pisana per la Scienza, Mass Spectrometry and Proteomics, Pisa, Italy (E.L.d.G., L.M.); Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Freiburg, Germany (G.S.); Istituto Superiore di Sanità, Rome, Italy (A.F., C.F.); Scuola Normale Superiore, Pisa, Italy (M.C., Mat.C.)
| | - Liam McDonnell
- CNR Neuroscience Institute, Pisa, Italy (E.V., F.O., L.B., M.C., Mat.C.); CNR Cellular Biology and Neurobiology Institute, Rome, Italy (S.M., M.A.-T.); Fondazione Pisana per la Scienza, Mass Spectrometry and Proteomics, Pisa, Italy (E.L.d.G., L.M.); Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Freiburg, Germany (G.S.); Istituto Superiore di Sanità, Rome, Italy (A.F., C.F.); Scuola Normale Superiore, Pisa, Italy (M.C., Mat.C.)
| | - Gudula Schmidt
- CNR Neuroscience Institute, Pisa, Italy (E.V., F.O., L.B., M.C., Mat.C.); CNR Cellular Biology and Neurobiology Institute, Rome, Italy (S.M., M.A.-T.); Fondazione Pisana per la Scienza, Mass Spectrometry and Proteomics, Pisa, Italy (E.L.d.G., L.M.); Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Freiburg, Germany (G.S.); Istituto Superiore di Sanità, Rome, Italy (A.F., C.F.); Scuola Normale Superiore, Pisa, Italy (M.C., Mat.C.)
| | - Alessia Fabbri
- CNR Neuroscience Institute, Pisa, Italy (E.V., F.O., L.B., M.C., Mat.C.); CNR Cellular Biology and Neurobiology Institute, Rome, Italy (S.M., M.A.-T.); Fondazione Pisana per la Scienza, Mass Spectrometry and Proteomics, Pisa, Italy (E.L.d.G., L.M.); Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Freiburg, Germany (G.S.); Istituto Superiore di Sanità, Rome, Italy (A.F., C.F.); Scuola Normale Superiore, Pisa, Italy (M.C., Mat.C.)
| | - Carla Fiorentini
- CNR Neuroscience Institute, Pisa, Italy (E.V., F.O., L.B., M.C., Mat.C.); CNR Cellular Biology and Neurobiology Institute, Rome, Italy (S.M., M.A.-T.); Fondazione Pisana per la Scienza, Mass Spectrometry and Proteomics, Pisa, Italy (E.L.d.G., L.M.); Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Freiburg, Germany (G.S.); Istituto Superiore di Sanità, Rome, Italy (A.F., C.F.); Scuola Normale Superiore, Pisa, Italy (M.C., Mat.C.)
| | - Laura Baroncelli
- CNR Neuroscience Institute, Pisa, Italy (E.V., F.O., L.B., M.C., Mat.C.); CNR Cellular Biology and Neurobiology Institute, Rome, Italy (S.M., M.A.-T.); Fondazione Pisana per la Scienza, Mass Spectrometry and Proteomics, Pisa, Italy (E.L.d.G., L.M.); Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Freiburg, Germany (G.S.); Istituto Superiore di Sanità, Rome, Italy (A.F., C.F.); Scuola Normale Superiore, Pisa, Italy (M.C., Mat.C.)
| | - Mario Costa
- CNR Neuroscience Institute, Pisa, Italy (E.V., F.O., L.B., M.C., Mat.C.); CNR Cellular Biology and Neurobiology Institute, Rome, Italy (S.M., M.A.-T.); Fondazione Pisana per la Scienza, Mass Spectrometry and Proteomics, Pisa, Italy (E.L.d.G., L.M.); Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Freiburg, Germany (G.S.); Istituto Superiore di Sanità, Rome, Italy (A.F., C.F.); Scuola Normale Superiore, Pisa, Italy (M.C., Mat.C.)
| | - Matteo Caleo
- CNR Neuroscience Institute, Pisa, Italy (E.V., F.O., L.B., M.C., Mat.C.); CNR Cellular Biology and Neurobiology Institute, Rome, Italy (S.M., M.A.-T.); Fondazione Pisana per la Scienza, Mass Spectrometry and Proteomics, Pisa, Italy (E.L.d.G., L.M.); Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Freiburg, Germany (G.S.); Istituto Superiore di Sanità, Rome, Italy (A.F., C.F.); Scuola Normale Superiore, Pisa, Italy (M.C., Mat.C.)
| |
Collapse
|
3
|
|
4
|
Works MG, Koenig JB, Sapolsky RM. Soluble TNF receptor 1-secreting ex vivo-derived dendritic cells reduce injury after stroke. J Cereb Blood Flow Metab 2013; 33:1376-85. [PMID: 23756688 PMCID: PMC3764376 DOI: 10.1038/jcbfm.2013.100] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 05/15/2013] [Accepted: 05/23/2013] [Indexed: 01/16/2023]
Abstract
Inflammation is a major factor in the progression of damage after stroke and in the clinic, current therapies treat the clot, not the resulting damage. We have developed a novel method of protein delivery that exploits the migration ability of leukocytes after ischemic stroke (transient middle cerebral artery occlusion; tMCAO). In our studies, ex vivo-derived dendritic cells (exDCs) migrate to the inflamed rat brain soon after tMCAO onset and the number of cells that remain in the brain after injection is significantly correlated with the amount of local inflammation at the injury site. In addition, exDCs transduced to overexpress soluble tumor necrosis factor (TNF) receptor1 (sTNFR1) produce functional cargo that is secreted and that blocks TNF-α bioavailability in vitro. When delivered at 6 hours post-tMCAO reperfusion, sTNFR1-exDC-treated rats show significantly smaller infarct size and decreased inflammation compared with animals treated with exDCs transduced with GFP lentivirus. These studies indicate that cell-mediated delivery of proteins may be a promising new approach to reduce brain damage after acute neurologic insult.
Collapse
Affiliation(s)
- Melissa G Works
- Department of Biology, Stanford University, Stanford, CA 94305, USA.
| | | | | |
Collapse
|
5
|
Zhao H, Ren C, Chen X, Shen J. From rapid to delayed and remote postconditioning: the evolving concept of ischemic postconditioning in brain ischemia. Curr Drug Targets 2012; 13:173-87. [PMID: 22204317 DOI: 10.2174/138945012799201621] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2010] [Revised: 08/09/2011] [Accepted: 08/12/2011] [Indexed: 01/13/2023]
Abstract
Ischemic postconditioning is a concept originally defined to contrast with that of ischemic preconditioning. While both preconditioning and postconditioning confer a neuroprotective effect on brain ischemia, preconditioning is a sublethal insult performed in advance of brain ischemia, and postconditioning, which conventionally refers to a series of brief occlusions and reperfusions of the blood vessels, is conducted after ischemia/reperfusion. In this article, we first briefly review the history of preconditioning, including the experimentation that initially uncovered its neuroprotective effects and later revealed its underlying mechanisms-of-action. We then discuss how preconditioning research evolved into that of postconditioning--a concept that now represents a broad range of stimuli or triggers, including delayed postconditioning, pharmacological postconditioning, remote postconditioning--and its underlying protective mechanisms involving the Akt, MAPK, PKC and K(ATP) channel cell-signaling pathways. Because the concept of postconditioning is so closely associated with that of preconditioning, and both share some common protective mechanisms, we also discuss whether a combination of preconditioning and postconditioning offers greater protection than preconditioning or postconditioning alone.
Collapse
Affiliation(s)
- Heng Zhao
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305-5327, USA.
| | | | | | | |
Collapse
|
6
|
Wei D, Ren C, Chen X, Zhao H. The chronic protective effects of limb remote preconditioning and the underlying mechanisms involved in inflammatory factors in rat stroke. PLoS One 2012; 7:e30892. [PMID: 22347410 PMCID: PMC3275571 DOI: 10.1371/journal.pone.0030892] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Accepted: 12/23/2011] [Indexed: 11/19/2022] Open
Abstract
We recently demonstrated that limb remote preconditioning (LRP) protects against focal ischemia measured 2 days post-stroke. Here, we studied whether LRP provides long-term protection and improves neurological function. We also investigated whether LRP transmits its protective signaling via the afferent nerve pathways from the preconditioned limb to the ischemic brain and whether inflammatory factors are involved in LRP, including the novel galectin-9/Tim-3 inflammatory cell signaling pathway, which induces cell death in lymphocytes. LRP in the left hind femoral artery was performed immediately before stroke. LRP reduced brain injury size both at 2 days and 60 days post-stroke and improved behavioral outcomes for up to 2 months. The sensory nerve inhibitors capsaicin and hexamethonium, a ganglion blocker, abolished the protective effects of LRP. In addition, LRP inhibited edema formation and blood-brain barrier (BBB) permeability measured 2 days post-stroke. Western blot and immunostaining analysis showed that LRP inhibited protein expression of both galectin-9 and T-cell immunoglobulin domain and mucin domain 3 (Tim-3), which were increased after stroke. In addition, LRP decreased iNOS and nitrotyrosine protein expression after stroke. In conclusion, LRP executes long-term protective effects against stroke and may block brain injury by inhibiting activities of the galectin-9/Tim-3 pathway, iNOS, and nitrotyrosine.
Collapse
Affiliation(s)
- Dingtai Wei
- Department of Neurosurgery, Stanford University, Stanford, California, United States of America
- Stroke Center, Stanford University, Stanford, California, United States of America
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin, China
- Department of Radiology, Fujian Medical University Ningde Hospital, Fujian, China
| | - Chuancheng Ren
- Department of Neurosurgery, Stanford University, Stanford, California, United States of America
- Stroke Center, Stanford University, Stanford, California, United States of America
- Shanghai No.5 Hospital, Fudan University, Shanghai, China
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Heng Zhao
- Department of Neurosurgery, Stanford University, Stanford, California, United States of America
- Stroke Center, Stanford University, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
7
|
Cheng MY, Lee IP, Jin M, Sun G, Zhao H, Steinberg GK, Sapolsky RM. An insult-inducible vector system activated by hypoxia and oxidative stress for neuronal gene therapy. Transl Stroke Res 2011; 2:92-100. [PMID: 21603078 PMCID: PMC3097421 DOI: 10.1007/s12975-010-0060-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Gene therapy has demonstrated the protective potential of a variety of genes against stroke. However, conventional gene therapy vectors are limited due to the inability to temporally control their expression, which can sometimes lead to deleterious side effects. Thus, an inducible vector that can be temporally controlled and activated by the insult itself would be advantageous. Using hypoxia responsive elements (HRE) and antioxidant responsive elements (ARE), we have constructed an insult-inducible vector activated by hypoxia and reactive oxygen species (ROS). In COS7 cells, the inducible ARE-HRE-luciferase vectors are highly activated by oxygen deprivation, hydrogen peroxide treatment, and the ROS-induced transcription factor NF-E2-related factor 2 (Nrf2). Using a defective herpes virus, the neuroprotective potential of this inducible vector was tested by over-expressing the transcription factor Nrf2. In primary cortical cultures, expression of the inducible ARE-HRE-Nrf2 protects against oxygen glucose deprivation, similar to that afforded by the constitutively expressed Nrf2. This ARE+HRE vector system is advantageous in that it allows the expression of a transgene to be activated not only during hypoxia but also maintained after reperfusion, thus prolonging the transgene expression during an ischemic insult. This insult-inducible vector system will be a valuable gene therapy tool for activating therapeutic/protective genes in cerebrovascular diseases.
Collapse
Affiliation(s)
- Michelle Y. Cheng
- Department of Biology, Stanford University, 371 Serra Mall, Stanford, CA 94305-5020, USA
- Department of Neurosurgery, Stanford University School of Medicine, 300 Pasteur Drive, Rm 281A, Stanford, CA 94305-5327, USA
| | - I-Ping Lee
- Department of Biology, Stanford University, 371 Serra Mall, Stanford, CA 94305-5020, USA
| | - Michael Jin
- Department of Biology, Stanford University, 371 Serra Mall, Stanford, CA 94305-5020, USA
| | - Guohua Sun
- Department of Neurosurgery, Stanford University School of Medicine, 300 Pasteur Drive, Rm 281A, Stanford, CA 94305-5327, USA
| | - Heng Zhao
- Department of Neurosurgery, Stanford University School of Medicine, 300 Pasteur Drive, Rm 281A, Stanford, CA 94305-5327, USA
| | - Gary K. Steinberg
- Department of Neurosurgery, Stanford University School of Medicine, 300 Pasteur Drive, Rm 281A, Stanford, CA 94305-5327, USA
| | - Robert M. Sapolsky
- Department of Biology, Stanford University, 371 Serra Mall, Stanford, CA 94305-5020, USA
- Department of Neurosurgery, Stanford University School of Medicine, 300 Pasteur Drive, Rm 281A, Stanford, CA 94305-5327, USA
| |
Collapse
|
8
|
Robin E, Simerabet M, Hassoun SM, Adamczyk S, Tavernier B, Vallet B, Bordet R, Lebuffe G. Postconditioning in focal cerebral ischemia: role of the mitochondrial ATP-dependent potassium channel. Brain Res 2010; 1375:137-46. [PMID: 21182830 DOI: 10.1016/j.brainres.2010.12.054] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2010] [Revised: 12/14/2010] [Accepted: 12/14/2010] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Ischemic postconditioning (IpostC) has been described in both heart and brain. The first aim of this study was to evaluate the effects of IpostC on brain infarct size and neurological function in the middle cerebral artery occlusion (MCAO) model. The second aim was to determine the involvement of the mitochondrial potassium ATP-dependent channel (mitoK(ATP)) opening and its capacity to improve mitochondrial dysfunction induced by ischemia-reperfusion. METHODS Wistar rats were subjected to 60min MCAO followed by 24-h reperfusion. Postconditioning was performed by 3 cycles of 30-s occlusion-reperfusion at the onset of reperfusion. Three behavioral tests were performed following 24h of reperfusion. Involvement of mitoK(ATP) was determined by the modulation of IpostC effects by 5-hydroxydecanoate (5-HD) and diazoxide. Mitochondrial function after 24h of reperfusion on isolated mitochondria was assessed through mitochondrial oxygen consumption, mitochondrial membrane potential and calcium retention capacity to evaluate impact of IpostC on mitochondrial permeability transition pore (MPTP) opening. RESULTS IpostC resulted in a 40% decrease in infarct size and improved neurological outcome. These effects were lost when IpostC was delayed by 5min. The administration of diazoxide resulted in a 60% in infarct size. The beneficial effects of IpostC and diazoxide were blocked by 5-HD. Furthermore, 5-HD also blocked the inhibition of MPTP opening by IpostC and diazoxide. The hyperpolarization induced by ischemia-reperfusion was corrected by IpostC without any effect on oxidative phosphorylation. CONCLUSION Our results confirm ischemic postconditioning-induced neuroprotection. They also support the involvement of mitoK(ATP) opening and its role in inhibiting the opening of MTPT induced by postconditioning.
Collapse
Affiliation(s)
- Emmanuel Robin
- Department of Pharmacology, Lille Medical School, University of Lille 2, Lille, France
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Mitra R, Sapolsky RM. Gene therapy in rodent amygdala against fear disorders. Expert Opin Biol Ther 2010; 10:1289-303. [DOI: 10.1517/14712598.2010.509341] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
10
|
Lee AL, Campbell LB, Sapolsky RM. Neighbor effects of neurons bearing protective transgenes. Brain Res 2010; 1339:70-5. [PMID: 20417625 PMCID: PMC2885846 DOI: 10.1016/j.brainres.2010.04.037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2010] [Revised: 04/14/2010] [Accepted: 04/16/2010] [Indexed: 11/29/2022]
Abstract
Viral vectors bearing protective transgenes can decrease neurotoxicity after varied necrotic insults. A neuron that dies necrotically releases glutamate, calcium and reactive oxygen species, thereby potentially damaging neighboring neurons. This raises the possibility that preventing such neuron death via gene therapy can secondarily protect neighboring neurons that, themselves, do not express a protective transgene. We determined whether such "good neighbor" effects occur, by characterizing neurons that, while uninfected themselves, are in close proximity to a transgene-bearing neuron. We tested two genes whose overexpression protects against excitotoxicity: anti-apoptotic Bcl-2, and a calcium-activated K(+) channel, SK2. Using herpes simplex virus type 2-mediated transgene delivery to hippocampal cultures, we observed "good neighbor" effects on neuronal survival following an excitotoxic insult. However, in the absence of insult, "bad neighbor" effects could also occur (i.e., where being in proximity to a neuron constitutively expressing one of those transgenes is deleterious). We also characterized the necessity for cell-cell contact for these effects. These phenomena may have broad implications for the efficacy of gene overexpression strategies in the CNS.
Collapse
Affiliation(s)
- Angela L Lee
- Department of Biology, Stanford University, Stanford, CA 94305-5020, USA.
| | | | | |
Collapse
|
11
|
Boison D. Cell and gene therapies for refractory epilepsy. Curr Neuropharmacol 2010; 5:115-25. [PMID: 18615179 DOI: 10.2174/157015907780866938] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2006] [Revised: 03/07/2007] [Accepted: 03/08/2007] [Indexed: 12/20/2022] Open
Abstract
Despite recent advances in the development of antiepileptic drugs, refractory epilepsy remains a major clinical problem affecting up to 35% of patients with partial epilepsy. Currently, there are few therapies that affect the underlying disease process. Therefore, novel therapeutic concepts are urgently needed. The recent development of experimental cell and gene therapies may offer several advantages compared to conventional systemic pharmacotherapy: (i) Specificity to underlying pathogenetic mechanisms by rational design; (ii) specificity to epileptogenic networks by focal delivery; and (iii) avoidance of side effects. A number of naturally occurring brain substances, such as GABA, adenosine, and the neuropeptides galanin and neuropeptide Y, may function as endogenous anticonvulsants and, in addition, may interact with the process of epileptogenesis. Unfortunately, the systemic application of these compounds is compromised by limited bioavailability, poor penetration of the blood-brain barrier, or the widespread systemic distribution of their respective receptors. Therefore, in recent years a new field of cell and gene-based neuropharmacology has emerged, aimed at either delivering endogenous anticonvulsant compounds by focal intracerebral transplantation of bioengineered cells (ex vivo gene therapy), or by inducing epileptogenic brain areas to produce these compounds in situ (in vivo gene therapy). In this review, recent efforts to develop GABA-, adenosine-, galanin-, and neuropeptide Y- based cell and gene therapies are discussed. The neurochemical rationales for using these compounds are discussed, the advantages of focal applications are highlighted and preclinical cell transplantation and gene therapy studies are critically evaluated. Although many promising data have been generated recently, potential problems, such as long-term therapeutic efficacy, long-term safety, and efficacy in clinically relevant animal models, need to be addressed before clinical applications can be contemplated.
Collapse
Affiliation(s)
- Detlev Boison
- RS Dow Neurobiology Laboratories, Legacy Research, Portland, OR 97232, USA.
| |
Collapse
|
12
|
Mitra R, Sapolsky RM. Expression of chimeric estrogen-glucocorticoid-receptor in the amygdala reduces anxiety. Brain Res 2010; 1342:33-8. [DOI: 10.1016/j.brainres.2010.03.092] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2009] [Revised: 03/12/2010] [Accepted: 03/27/2010] [Indexed: 11/28/2022]
|
13
|
Mitra R, Ferguson D, Sapolsky RM. Mineralocorticoid receptor overexpression in basolateral amygdala reduces corticosterone secretion and anxiety. Biol Psychiatry 2009; 66:686-90. [PMID: 19500777 DOI: 10.1016/j.biopsych.2009.04.016] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2008] [Revised: 03/17/2009] [Accepted: 04/02/2009] [Indexed: 11/15/2022]
Abstract
BACKGROUND The amygdala plays a critical role in the development of anxiety and the regulation of stress hormone secretion. Reciprocally, stress and stress hormones can induce amygdala hypertrophy, a phenomenon related to enhanced anxiety. As such, modulating amygdaloid function can potentially reduce maladaptive features of the stress response. The amygdala contains two kind of receptor for corticosteroids, the adrenal steroid hormone released during stress: glucocorticoid receptors (GRs) and mineralocorticoid receptors (MRs). Although high-affinity MRs are heavily occupied during basal conditions, low-affinity GRs are heavily occupied only by stress levels of glucocorticoids. Prolonged and heavy occupancy of GRs tends to mediate the deleterious effects of glucocorticoids on neurons, whereas MR occupancy tends to mediate beneficial effects. METHODS In this report, we overexpress MR in neurons of adult rat basolateral amygdala, with a herpes simplex viral vector coding for two copies of MR. RESULTS Such overexpression reduced anxiety, as measured on an elevated plus-maze, and reduced the magnitude of glucocorticoid secretion after an acute stressor. CONCLUSIONS Thus, increasing MR signaling in basolateral amygdala could be valuable in management of stress disorders.
Collapse
Affiliation(s)
- Rupshi Mitra
- Department of Biology, Stanford University, Stanford, California 94305, USA.
| | | | | |
Collapse
|
14
|
Mitra R, Ferguson D, Sapolsky RM. SK2 potassium channel overexpression in basolateral amygdala reduces anxiety, stress-induced corticosterone secretion and dendritic arborization. Mol Psychiatry 2009; 14:847-55, 827. [PMID: 19204724 PMCID: PMC2763614 DOI: 10.1038/mp.2009.9] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2008] [Revised: 12/19/2008] [Accepted: 01/07/2009] [Indexed: 02/01/2023]
Abstract
The basolateral amygdala is critical for generation of anxiety. In addition, exposure to both stress and glucocorticoids induces anxiety. Demonstrated ability of the amygdala to change in response to stress and glucocorticoids could thus be important therapeutic target for anxiety management. Several studies have reported a relationship between anxiety and dendritic arborization of the amygdaloid neurons. In this study we employed a gene therapeutic approach to reduce anxiety and dendritic arborization of the amygdala neurons. Specifically, we overexpressed SK2 potassium channel in the basolateral amygdala using a herpes simplex viral system. Our choice of therapeutic cargo was guided by the indications that activation of the amygdala might underlie anxiety and that SK2 could reduce neuronal activation by exerting inhibitory influence on action potentials. We report that SK2 overexpression reduced anxiety and stress-induced corticosterone secretion at a systemic level. SK2 overexpression also reduced dendritic arborization of the amygdala neurons. Hence, SK2 is a potential gene therapy candidate molecule that can be used against stress-related neuropsychiatric disorders such as anxiety.
Collapse
Affiliation(s)
- R Mitra
- Department of Biology, Stanford University, Stanford, CA 94305, USA.
| | | | | |
Collapse
|
15
|
Abstract
Ischemic postconditioning initially referred to a stuttering reperfusion performed immediately after reperfusion, for preventing ischemia/reperfusion injury in both myocardial and cerebral infarction. It has evolved into a concept that can be induced by a broad range of stimuli or triggers, and may even be performed as late as 6 h after focal ischemia and 2 days after transient global ischemia. The concept is thought to be derived from ischemic preconditioning or partial/gradual reperfusion, but in fact the first experiment for postconditioning was carried out much earlier than that of preconditioning or partial/gradual reperfusion, in the research on myocardial ischemia. This review first examines the protective effects and parameters of postconditioning in various cerebral ischemic models. Thereafter, it provides insights into the protective mechanisms of postconditioning associated with reperfusion injury and the Akt, mitogen-activated protein kinase (MAPK), protein kinase C (PKC), and ATP-sensitive K+ (K(ATP)) channel cell signaling pathways. Finally, some open issues and future challenges regarding clinical translation of postconditioning are discussed.
Collapse
Affiliation(s)
- Heng Zhao
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California 94305-5327, USA.
| |
Collapse
|
16
|
Rodrigues SM, Sapolsky RM. Disruption of fear memory through dual-hormone gene therapy. Biol Psychiatry 2009; 65:441-4. [PMID: 18973875 PMCID: PMC2660393 DOI: 10.1016/j.biopsych.2008.09.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2007] [Revised: 08/12/2008] [Accepted: 09/03/2008] [Indexed: 01/14/2023]
Abstract
BACKGROUND The basolateral complex of the amygdala (BLA) is uniquely affected by steroid hormones. Whereas glucocorticoids (GCs)--the adrenal hormones released during stress--increase the excitability of BLA neurons, estrogen decreases it. METHODS In this study, we used a vector designed to express a chimeric gene that contains the GC-binding domain of the GC receptor (GR) and the DNA binding domain of the estrogen receptor (ER) ("ER/GR") in infected neurons; as a result, it transduces GC signals into estrogenic ones. We microinfused ER/GR bilaterally into the BLA of rats to determine whether it would impair fear conditioning, a valuable BLA-dependent paradigm for studying the neural basis of emotional memory. RESULTS Expression of ER/GR in the BLA caused robust expression of the transgene and a significant disruption of both auditory and contextual long-term fear memory consolidation, whereas fear learning and post-shock freezing remained intact. CONCLUSIONS These data show that dual gene therapy with ER/GR might be a useful tool for understanding the role of steroid hormones in the storage of traumatic memories.
Collapse
Affiliation(s)
- Sarina M Rodrigues
- Department of Biological Sciences, Stanford University, Stanford, California, USA.
| | | |
Collapse
|
17
|
Blocking glucocorticoid and enhancing estrogenic genomic signaling protects against cerebral ischemia. J Cereb Blood Flow Metab 2009; 29:130-6. [PMID: 18797472 DOI: 10.1038/jcbfm.2008.105] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Glucocorticoids (GCs) and estrogen can modulate neuron death and dysfunction during neurological insults. Glucocorticoids are adrenal steroids secreted during stress, and hypersecretion of GCs during cerebral ischemia compromises the ability of hippocampal and cortical neurons to survive. In contrast, estrogen can be neuroprotective after cerebral ischemia. Here we evaluate the protective potential of a herpes viral vector expressing a chimeric receptor (ER/GR), which is composed of the ligand-binding domain of the GC receptor (GR) and the DNA-binding domain of the estrogen receptor-alpha (ER). This novel receptor can transduce an endangering GC signal into a protective estrogenic one. Using an in vitro oxygen glucose deprivation model (OGD), GCs exacerbated neuron death in primary cortical cultures, and this worsening effect was completely blocked by ER/GR expression. Moreover, blocking GC actions with a vector expressing a dominant negative GC receptor promoted neuron survival during postischemia, but not preischemia. Thus, gene therapeutic strategies to modulate GC and estrogen signaling can be beneficial during an ischemic insult.
Collapse
|
18
|
Tsai LK, Tsai MS, Ting CH, Wang SH, Li H. Restoring Bcl-x(L) levels benefits a mouse model of spinal muscular atrophy. Neurobiol Dis 2008; 31:361-7. [PMID: 18590823 DOI: 10.1016/j.nbd.2008.05.014] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2008] [Revised: 05/13/2008] [Accepted: 05/19/2008] [Indexed: 10/22/2022] Open
Abstract
Currently, no curative treatment is available for spinal muscular atrophy (SMA). Since the degeneration of spinal motor neurons in SMA is mediated by apoptosis, over-expression of an anti-apoptotic factor, Bcl-x(L), may benefit SMA. Here, we crossed a mouse model of SMA with Bcl-x(L) transgenic mice to create SMA/Bcl-x(L) mice. The Bcl-x(L) expression in the spinal neurons of SMA/Bcl-x(L) mice was nearly double that in SMA mice. SMA/Bcl-x(L) mice showed preserved motor function, normalized electrophysiological tests, diminished muscle atrophy, and less motor neuron degeneration. In addition, the life span of SMA/Bcl-x(L) mice was 1.5 times longer than that of SMA mice. Therefore, over-expression of Bcl-x(L) has a potential for amelioration of SMA, and Bcl-x(L) may be another attractive therapeutic target other than survival motor neuron (SMN) protein for use in future drug screening for SMA.
Collapse
Affiliation(s)
- Li-Kai Tsai
- Department of Neurology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei 100, Taiwan
| | | | | | | | | |
Collapse
|
19
|
Gao X, Zhang H, Takahashi T, Hsieh J, Liao J, Steinberg GK, Zhao H. The Akt signaling pathway contributes to postconditioning's protection against stroke; the protection is associated with the MAPK and PKC pathways. J Neurochem 2008; 105:943-55. [PMID: 18182053 PMCID: PMC2746404 DOI: 10.1111/j.1471-4159.2008.05218.x] [Citation(s) in RCA: 145] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
We previously reported that ischemic postconditioning with a series of mechanical interruptions of reperfusion reduced infarct volume 2 days after focal ischemia in rats. Here, we extend this data by examining long-term protection and exploring underlying mechanisms involving the Akt, mitogen-activated protein kinase (MAPK) and protein kinase C (PKC) signaling pathways. Post-conditioning reduced infarct and improved behavioral function assessed 30 days after stroke. Additionally, postconditioning increased levels of phosphorylated Akt (Ser473) as measured by western blot and Akt activity as measured by an in vitro kinase assay. Inhibiting Akt activity by a phosphoinositide 3-kinase inhibitor, LY294002, enlarged infarct in postconditioned rats. Postconditioning did not affect protein levels of phosphorylated-phosphatase and tensin homologue deleted on chromosome 10 or -phosphoinositide-dependent protein kinase-1 (molecules upstream of Akt) but did inhibit an increase in phosphorylated-glycogen synthase kinase 3beta, an Akt effector. In addition, postconditioning blocked beta-catenin phosphorylation subsequent to glycogen synthase kinase, but had no effect on total or non-phosphorylated active beta-catenin protein levels. Furthermore, postconditioning inhibited increases in the amount of phosphorylated-c-Jun N-terminal kinase and extracellular signal-regulated kinase 1/2 in the MAPK pathway. Finally, postconditioning blocked death-promoting deltaPKC cleavage and attenuated reduction in phosphorylation of survival-promoting epsilonPKC. In conclusion, our data suggest that postconditioning provides long-term protection against stroke in rats. Additionally, we found that Akt activity contributes to postconditioning's protection; furthermore, increases in epsilonPKC activity, a survival-promoting pathway, and reductions in MAPK and deltaPKC activity; two putative death-promoting pathways correlate with postconditioning's protection.
Collapse
Affiliation(s)
- Xuwen Gao
- Department of Neurosurgery, Stanford University, Palo Alto, California, USA
- Stanford Stroke Center, Stanford University, Palo Alto, California, USA
| | - Hanfeng Zhang
- Department of Neurosurgery, Stanford University, Palo Alto, California, USA
- Stanford Stroke Center, Stanford University, Palo Alto, California, USA
| | - Tetsuya Takahashi
- Department of Neurosurgery, Stanford University, Palo Alto, California, USA
- Stanford Stroke Center, Stanford University, Palo Alto, California, USA
| | - Jason Hsieh
- Department of Neurosurgery, Stanford University, Palo Alto, California, USA
| | - Janette Liao
- Department of Neurosurgery, Stanford University, Palo Alto, California, USA
| | - Gary K. Steinberg
- Department of Neurosurgery, Stanford University, Palo Alto, California, USA
- Stanford Stroke Center, Stanford University, Palo Alto, California, USA
| | - Heng Zhao
- Department of Neurosurgery, Stanford University, Palo Alto, California, USA
- Stanford Stroke Center, Stanford University, Palo Alto, California, USA
| |
Collapse
|
20
|
The Protective Effect of Ischemic Postconditioning Against Ischemic Injury: From the Heart to the Brain. J Neuroimmune Pharmacol 2007; 2:313-8. [DOI: 10.1007/s11481-007-9089-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2006] [Accepted: 08/21/2007] [Indexed: 12/01/2022]
|
21
|
Royo NC, LeBold D, Magge SN, Chen I, Hauspurg A, Cohen AS, Watson DJ. Neurotrophin-mediated neuroprotection of hippocampal neurons following traumatic brain injury is not associated with acute recovery of hippocampal function. Neuroscience 2007; 148:359-70. [PMID: 17681695 PMCID: PMC2579330 DOI: 10.1016/j.neuroscience.2007.06.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2007] [Revised: 06/12/2007] [Accepted: 06/18/2007] [Indexed: 11/23/2022]
Abstract
Traumatic brain injury (TBI) causes selective hippocampal cell death which is believed to be associated with the cognitive impairment observed in both clinical and experimental settings. The endogenous neurotrophin-4/5 (NT-4/5), a TrkB ligand, has been shown to be neuroprotective for vulnerable CA3 pyramidal neurons after experimental brain injury. In this study, infusion of recombinant NT-4/5 increased survival of CA2/3 pyramidal neurons to 71% after lateral fluid percussion brain injury in rats, compared with 55% in vehicle-treated controls. The functional outcome of this NT-4/5-mediated neuroprotection was examined using three hippocampal-dependent behavioral tests. Injury-induced impairment was evident in all three tests, but interestingly, there was no treatment-related improvement in any of these measures. Similarly, injury-induced decreased excitability in the Schaffer collaterals was not affected by NT-4/5 treatment. We propose that a deeper understanding of the factors that link neuronal survival to recovery of function will be important for future studies of potentially therapeutic agents.
Collapse
Affiliation(s)
- N C Royo
- Department of Neurosurgery, 371A Stemmler Hall/6071, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA.
| | | | | | | | | | | | | |
Collapse
|
22
|
Ferguson D, Sapolsky R. Mineralocorticoid receptor overexpression differentially modulates specific phases of spatial and nonspatial memory. J Neurosci 2007; 27:8046-52. [PMID: 17652595 PMCID: PMC6672723 DOI: 10.1523/jneurosci.1187-07.2007] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Glucocorticoids (GCs) and stress modulate specific phases of information processing. The modulatory affects of GCs on hippocampal function are thought to be mediated by the mineralocorticoid receptor (MR) and the glucocorticoid receptor (GR). The GR plays a critical role in mediating the impairing effects of GCs on hippocampal function. Conversely, activation of MR facilitates hippocampal function. The high affinity of MR for GCs suggests that the receptor protein levels play a key role in regulating the beneficial effects of MR-mediated gene transcription. Using herpes simplex vectors, we transiently increased MR levels in dentate gyrus granule cells, which in turn enhanced MR signaling. We then examined its effects on spatial and nonspatial memory consolidation and retrieval using the object placement and object recognition task. Additionally, we assessed whether an increased MR signal could block the impairing effects of high GCs on memory retrieval. Rats overexpressing MR displayed an enhancement in the consolidation of nonspatial memory relative to rats expressing green fluorescent protein and suggest the potential for gene transfer techniques for enhancing cognition during stress. Moreover, rats overexpressing MR were spared from the disruptive effects of high GCs on the retrieval of nonspatial memory. Thus, this study illustrates the critical role of MR in mediating the retrieval and consolidation of nonspatial memory.
Collapse
Affiliation(s)
- Deveroux Ferguson
- Department of Biological Sciences, Stanford University, Stanford, California 94305, USA.
| | | |
Collapse
|
23
|
Dietrich J, Kempermann G. Role of Endogenous Neural Stem Cells in Neurological Disease and Brain Repair. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 557:191-220. [PMID: 16955712 DOI: 10.1007/0-387-30128-3_12] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
These examples show that stem-cell-based therapy of neuro-psychiatric disorders will not follow a single scheme, but rather include widely different approaches. This is in accordance with the notion that the impact of stem cell biology on neurology will be fundamental, providing a shift in perspective, rather than introducing just one novel therapeutic tool. Stem cell biology, much like genomics and proteomics, offers a "view from within" with an emphasis on a theoretical or real potential and thereby the inherent openness, which is central to the concept of stem cells. Thus, stem cell biology influences many other, more traditional therapeutic approaches, rather than introducing one distinct novel form of therapy. Substantial advances have been made i n neural stemcell research during the years. With the identification of stem and progenitor cells in the adult brain and the complex interaction of different stem cell compartments in the CNS--both, under physiological and pathological conditions--new questions arise: What is the lineage relationship between t he different progenitor cells in the CNS and how much lineage plasticity exists? What are the signals controlling proliferation and differentiation of neural stem cells and can these be utilized to allow repair of the CNS? Insights in these questions will help to better understand the role of stem cells during development and aging and the possible relation of impaired or disrupted stem cell function and their impact on both the development and treatment of neurological disease. A number o f studies have indicated a limited neuronal and glial regeneration certain pathological conditions. These fundamental observations have already changed our view on understanding neurological disease and the brain's capacity for endogenous repair. The following years will have to show how we can influence andmodulate endogenous repair nisms by increasing the cellular plasticity in the young and aged CNS.
Collapse
Affiliation(s)
- Jörg Dietrich
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | | |
Collapse
|
24
|
Noe' F, Nissinen J, Pitkänen A, Gobbi M, Sperk G, During M, Vezzani A. Gene therapy in epilepsy: the focus on NPY. Peptides 2007; 28:377-83. [PMID: 17196301 DOI: 10.1016/j.peptides.2006.07.025] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2006] [Accepted: 07/24/2006] [Indexed: 01/16/2023]
Abstract
Gene therapy represents an innovative and promising alternative for the treatment of epileptic patients who are resistant to conventional antiepileptic drugs. Among the various approaches for the application of gene therapy in the treatment of CNS disorders, recombinant viral vectors have been most widely used so far. Several gene targets could be used to correct the compromized balance between inhibitory and excitatory transmission in epilepsy. Transduction of neuropeptide genes such as galanin and neuropeptide Y (NPY) in specific brain areas in experimental models of seizures resulted in significant anticonvulsant effects. In particular, the long-lasting NPY over-expression obtained in the rat hippocampus using intracerebral application of recombinant adeno-associated viral (AAV) vectors reduced the generalization of seizures from their site of onset, delayed acquisition of fully kindled seizures and afforded neuroprotection. These results establish a proof-of-principle for the applicability of AAV-NPY vectors for the inhibition of seizures in epilepsy. Additional investigations are required to demonstrate a therapeutic role of gene therapy in chronic models of seizures and to address in more detail safety concerns and possible side-effects.
Collapse
Affiliation(s)
- Francesco Noe'
- Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Via Eritrea 62, 20157 Milano, Italy
| | | | | | | | | | | | | |
Collapse
|
25
|
Golembewski EK, Wales SQ, Aurelian L, Yarowsky PJ. The HSV-2 protein ICP10PK prevents neuronal apoptosis and loss of function in an in vivo model of neurodegeneration associated with glutamate excitotoxicity. Exp Neurol 2006; 203:381-93. [PMID: 17046754 PMCID: PMC1994904 DOI: 10.1016/j.expneurol.2006.08.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2006] [Revised: 08/16/2006] [Accepted: 08/22/2006] [Indexed: 12/13/2022]
Abstract
Excessive glutamate receptor activation results in neuronal death, a process known as excitotoxicity. Intrastriatal injection of N-methyl-d-aspartate (NMDA) is a model of excitotoxicity. We used this model to examine whether excitotoxic injury is inhibited by the anti-apoptotic herpes simplex virus type 2 (HSV-2) protein, ICP10PK, delivered by the replication incompetent HSV-2 vector, DeltaRR. Intrastriatal DeltaRR administration (2500 plaque forming units) was nontoxic and did not induce microglial activation 5 days after injection. Intrastriatal injection of DeltaRR with NMDA or 4 h after NMDA injection showed increased neuronal survival and decreased mitochondrial damage compared to injection of NMDA alone. Neuroprotection was due to the inhibition of NMDA-induced apoptosis through ERK activation. DeltaRR-treated mice did not develop NMDA-associated behavioral deficits. The data suggest that DeltaRR is a promising platform for treatment of acute neuronal injury.
Collapse
Affiliation(s)
- Erin K Golembewski
- Department of Pharmacology and Experimental Therapeutics, University of Maryland, Baltimore, MD 20742, USA
| | | | | | | |
Collapse
|
26
|
Peluffo H, Acarin L, Arís A, González P, Villaverde A, Castellano B, González B. Neuroprotection from NMDA excitotoxic lesion by Cu/Zn superoxide dismutase gene delivery to the postnatal rat brain by a modular protein vector. BMC Neurosci 2006; 7:35. [PMID: 16638118 PMCID: PMC1462999 DOI: 10.1186/1471-2202-7-35] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2005] [Accepted: 04/25/2006] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Superoxide mediated oxidative stress is a key neuropathologic mechanism in acute central nervous system injuries. We have analyzed the neuroprotective efficacy of the transient overexpression of antioxidant enzyme Cu/Zn Superoxide dismutase (SOD) after excitotoxic injury to the immature rat brain by using a recently constructed modular protein vector for non-viral gene delivery termed NLSCt. For this purpose, animals were injected with the NLSCt vector carrying the Cu/Zn SOD or the control GFP transgenes 2 hours after intracortical N-methyl-D-aspartate (NMDA) administration, and daily functional evaluation was performed. Moreover, 3 days after, lesion volume, neuronal degeneration and nitrotyrosine immunoreactivity were evaluated. RESULTS Overexpression of Cu/Zn SOD transgene after NMDA administration showed improved functional outcome and a reduced lesion volume at 3 days post lesion. In secondary degenerative areas, increased neuronal survival as well as decreased numbers of degenerating neurons and nitrotyrosine immunoreactivity was seen. Interestingly, injection of the NLSCt vector carrying the control GFP transgene also displayed a significant neuroprotective effect but less pronounced. CONCLUSION When the appropriate levels of Cu/Zn SOD are expressed transiently after injury using the non-viral modular protein vector NLSCt a neuroprotective effect is seen. Thus recombinant modular protein vectors may be suitable for in vivo gene therapy, and Cu/Zn SOD should be considered as an interesting therapeutic transgene.
Collapse
Affiliation(s)
- Hugo Peluffo
- Unitat d'Histologia, Torre M5, Facultat de Medicina, Departament de Biologia Cel.lular, Fisiologia i Immunologia, and Institut de Neurociències, Universitat Autònoma de Barcelona, 08193, Spain
| | - Laia Acarin
- Unitat d'Histologia, Torre M5, Facultat de Medicina, Departament de Biologia Cel.lular, Fisiologia i Immunologia, and Institut de Neurociències, Universitat Autònoma de Barcelona, 08193, Spain
| | - Anna Arís
- Institut de Biotecnologia i de Biomedicina and Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08193, Spain
| | - Pau González
- Unitat d'Histologia, Torre M5, Facultat de Medicina, Departament de Biologia Cel.lular, Fisiologia i Immunologia, and Institut de Neurociències, Universitat Autònoma de Barcelona, 08193, Spain
| | - Antoni Villaverde
- Institut de Biotecnologia i de Biomedicina and Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08193, Spain
| | - Bernardo Castellano
- Unitat d'Histologia, Torre M5, Facultat de Medicina, Departament de Biologia Cel.lular, Fisiologia i Immunologia, and Institut de Neurociències, Universitat Autònoma de Barcelona, 08193, Spain
| | - Berta González
- Unitat d'Histologia, Torre M5, Facultat de Medicina, Departament de Biologia Cel.lular, Fisiologia i Immunologia, and Institut de Neurociències, Universitat Autònoma de Barcelona, 08193, Spain
| |
Collapse
|
27
|
Garrity-Moses ME, Teng Q, Liu J, Tanase D, Boulis NM. Neuroprotective adeno-associated virus Bcl-xL gene transfer in models of motor neuron disease. Muscle Nerve 2006; 32:734-44. [PMID: 16116646 DOI: 10.1002/mus.20418] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Recent work implicates excitotoxicity-induced apoptosis as the mechanism triggering motor neuron death in amyotrophic lateral sclerosis (ALS). Our laboratory has previously utilized glutamate excitotoxicity in vitro to study this process. The present experiment tests whether overexpression of the gene for Bcl-xL can inhibit excitotoxicity in this model system. To track Bcl-xL expression, the gene for green fluorescent protein (GFP) was inserted in-frame, upstream of the Bcl-xL gene. The GFP-Bcl-xL gene was then cloned into an adeno-associated viral (AAV2) vector. GFP expression in both SH-SY5Y and embryonic day 15 (E15) motor neurons (MNs) peaked 48 hours after infection. Bcl-xL expression in SH-SY5Y cells significantly reduced terminal deoxy-UTP nick-end labeling (TUNEL)-positive cells and maintained cell density after glutamate exposure. Similarly, Bcl-xL expression inhibited the development of TUNEL staining in E15 MNs and supported cell density after glutamate exposure. These findings suggest that AAV-mediated expression of genes for antiapoptotic proteins may provide a means for ALS gene therapy.
Collapse
Affiliation(s)
- Mary E Garrity-Moses
- Department of Neurological Surgery, Lerner Research Institute, Cleveland Clinic Foundation, NB 2 120, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | | | | | | | | |
Collapse
|
28
|
Abstract
Herpes simplex virus (HSV)-based vectors have primarily been developed for neuronal gene delivery, taking advantage of the virus' natural neurotropism. Two types of vector are available: replication defective viruses, whose cytotoxicity has been abolished by deleting viral gene products, and amplicon vectors, which are plasmids packaged into HSV particles with the aid of a helper virus. In this review I discuss how the cytotoxicity of the wild-type virus has been abolished, the progress which has been made toward defining promoter elements capable of directing long-term transgene expression form the latent viral genome and some of the potential clinical uses of these versatile vectors.
Collapse
Affiliation(s)
- Robin Lachmann
- Department of Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
29
|
Caleo M, Cenni MC. Anterograde transport of neurotrophic factors: possible therapeutic implications. Mol Neurobiol 2004; 29:179-96. [PMID: 15126685 DOI: 10.1385/mn:29:2:179] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2003] [Accepted: 10/06/2003] [Indexed: 12/14/2022]
Abstract
The actions of neurotrophic factors are classically thought to be mediated by their retrograde transport from target tissues to the cell bodies. There is now evidence that specific trophic factors are trafficked anterogradely along peripheral and central axons and released to postsynaptic cells. This review focuses on recent experiments that demonstrate the involvement of the anterograde transfer of neurotrophic factors in various physiological processes, including the regulation of developmental neuronal death, the modulation of synaptic transmission, and the control of axonal and dendritic architecture. The authors also discuss whether anterograde transport of exogenous trophic factors can be exploited to protect damaged postsynaptic neurons and spare their function. This issue has clear implications for possible therapeutic applications of neurotrophic factors.
Collapse
Affiliation(s)
- Matteo Caleo
- Istituto di Neuroscienze del CNR and Scuola Normale Superiore, via G. Moruzzi, 1 - 56100 Pisa, Italy.
| | | |
Collapse
|
30
|
Lee AL, Dumas TC, Tarapore PE, Webster BR, Ho DY, Kaufer D, Sapolsky RM. Potassium channel gene therapy can prevent neuron death resulting from necrotic and apoptotic insults. J Neurochem 2003; 86:1079-88. [PMID: 12911616 DOI: 10.1046/j.1471-4159.2003.01880.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Necrotic insults such as seizure are excitotoxic. Logically, membrane hyperpolarization by increasing outwardly conducting potassium channel currents should attenuate hyperexcitation and enhance neuron survival. Therefore, we overexpressed a small-conductance calcium-activated (SK2) or voltage-gated (Kv1.1) channel via viral vectors in cultured hippocampal neurons. We found that SK2 or Kv1.1 protected not only against kainate or glutamate excitotoxicity but also increased survival after sodium cyanide or staurosporine. In vivo overexpression of either channel in dentate gyrus reduced kainate-induced CA3 lesions. In hippocampal slices, the kainate-induced increase in granule cell excitability was reduced by overexpression of either channel, suggesting that these channels exert their protective effects during hyperexcitation. It is also important to understand any functional disturbances created by transgene overexpression alone. In the absence of insult, overexpression of Kv1.1, but not SK2, reduced baseline excitability in dentate gyrus granule cells. Furthermore, while no behavioral disturbances during spatial acquisition in the Morris water maze were observed with overexpression of either channel, animals overexpressing SK2, but not Kv1.1, exhibited a memory deficit post-training. This difference raises the possibility that the means by which these channel subtypes protect may differ. With further development, potassium channel vectors may be an effective pre-emptive strategy against necrotic insults.
Collapse
Affiliation(s)
- Angela L Lee
- Department of Biological Sciences, Stanford University, Stanford, CA 94305-5020, USA.
| | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Alzheimer's disease (AD) is a polygenic/complex disorder in which more than 50 genetic loci are involved. Primary and secondary loci are potentially responsible for the phenotypic expression of the disease under the influence of both environmental factors and epigenetic phenomena. The construction of haplotypes as genomic clusters integrating the different genotype combinations of AD-related genes is a suitable strategy to investigate functional genomics in AD. It appears that AD patients show about 3-5 times higher genetic variation than the control population. The analysis of genotype-phenotype correlations has revealed that the presence of the APOE-4 allele in AD, in conjunction with other loci distributed across the genome, influence disease onset, brain atrophy, cerebrovascular perfusion, blood pressure, beta-amyloid deposition, ApoE secretion, lipid metabolism, brain bioelectrical activity, cognition, apoptosis and treatment outcome. Pharmacogenomics studies also indicate that the therapeutic response in AD is genotype-specific and that approximately 15% of the cases with efficacy and/or safety problems are associated with a defective CYP2D6 gene. Consequently, the understanding of functional genomics in AD will foster productive pharmacogenomic studies in the search for effective medications and preventive strategies in AD.
Collapse
Affiliation(s)
- Ramón Cacabelos
- EuroEspes Biomedical Research Center, Institute for CNS Disorders, 15166-Bergondo, Coruña, Spain.
| |
Collapse
|
32
|
Liou AKF, Clark RS, Henshall DC, Yin XM, Chen J. To die or not to die for neurons in ischemia, traumatic brain injury and epilepsy: a review on the stress-activated signaling pathways and apoptotic pathways. Prog Neurobiol 2003; 69:103-42. [PMID: 12684068 DOI: 10.1016/s0301-0082(03)00005-4] [Citation(s) in RCA: 216] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
After a severe episode of ischemia, traumatic brain injury (TBI) or epilepsy, it is typical to find necrotic cell death within the injury core. In addition, a substantial number of neurons in regions surrounding the injury core have been observed to die via the programmed cell death (PCD) pathways due to secondary effects derived from the various types of insults. Apart from the cell loss in the injury core, cell death in regions surrounding the injury core may also contribute to significant losses in neurological functions. In fact, it is the injured neurons in these regions around the injury core that treatments are targeting to preserve. In this review, we present our cumulated understanding of stress-activated signaling pathways and apoptotic pathways in the research areas of ischemic injury, TBI and epilepsy and that gathered from concerted research efforts in oncology and other diseases. However, it is obvious that our understanding of these pathways in the context of acute brain injury is at its infancy stage and merits further investigation. Hopefully, this added research effort will provide a more detailed knowledge from which better therapeutic strategies can be developed to treat these acute brain injuries.
Collapse
Affiliation(s)
- Anthony K F Liou
- Department of Neurology, University of Pittsburgh School of Medicine, S526 Biomedical Science Tower, 3500 Terrace Street, Pittsburgh, PA 15261, USA
| | | | | | | | | |
Collapse
|
33
|
Provision of brain-derived neurotrophic factor via anterograde transport from the eye preserves the physiological responses of axotomized geniculate neurons. J Neurosci 2003. [PMID: 12514226 DOI: 10.1523/jneurosci.23-01-00287.2003] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The neurotrophic factors of the nerve growth factor family (neurotrophins) have been shown to promote neuronal survival after brain injury and in various models of neurodegenerative conditions. However, it has not been determined whether neurotrophin treatment results in the maintenance of function of the rescued cells. Here we have used the retrograde degeneration of geniculate neurons as a model system to evaluate neuronal rescue and sparing of function after administration of brain-derived neurotrophic factor (BDNF). Death of geniculate neurons was induced by a visual cortex lesion in adult rats, and exogenous BDNF was delivered to the axotomized geniculate cells via anterograde transport after injection into the eye. By microelectrode recordings from the geniculate in vivo we have measured several physiological parameters such as contrast threshold, spatial resolution (visual acuity), signal-to-noise ratio, temporal resolution, and response latency. In control lesioned animals we found that geniculate cell dysfunction precedes the onset of neuronal death, indicating that an assessment of neuronal number per se is not predictive of functional performance. The administration of BDNF resulted in a highly significant cell-saving effect up to 2 weeks after the cortical damage and maintained nearly normal physiological responses in the geniculate. This preservation of function in adult axotomized neurons suggests possible therapeutic applications of BDNF.
Collapse
|
34
|
Affiliation(s)
- Robert M Sapolsky
- Department of Biological Sciences, Stanford University, Gilbert Laboratory, Stanford, California 94305-5020, USA.
| |
Collapse
|
35
|
Abstract
To illuminate the function of the thousands of genes that make up the complexity of the nervous system, it is critical to be able to introduce and express DNA in neurons. Over the past two decades, many gene transfer methods have been developed, including viral vectors, liposomes and electroporation. Although the perfect gene transfer technique for every application has not yet been developed, recent technical advances have facilitated the ease of neuronal gene transfer and have increased the accessibility of these techniques to all laboratories. In order to select a transfection method for any particular experiment, the specific advantages and disadvantages of each technique must be considered.
Collapse
Affiliation(s)
- Philip Washbourne
- Center for Neuroscience, University of California, Davis, Davis, California 95616, USA.
| | | |
Collapse
|
36
|
Harvey AR, Kamphuis W, Eggers R, Symons NA, Blits B, Niclou S, Boer GJ, Verhaagen J. Intravitreal injection of adeno-associated viral vectors results in the transduction of different types of retinal neurons in neonatal and adult rats: a comparison with lentiviral vectors. Mol Cell Neurosci 2002; 21:141-57. [PMID: 12359157 DOI: 10.1006/mcne.2002.1168] [Citation(s) in RCA: 89] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Replication-deficient viral vectors encoding the marker gene green fluorescent protein (GFP) were injected into the vitreous of newborn, juvenile (P14), and adult rats. We tested two different types of modified virus: adeno-associated viral-2-GFP (AAV-GFP) and lentiviral-GFP vectors (LV-GFP). The extent of retinal cell transduction in different-aged animals was compared 7, 21, and 70 days after eye injections. At all postinjection times, LV-GFP transduction was mostly limited to pigment epithelium and cells in sclera and choroid. In contrast, transduction of large numbers of neural retinal cells was seen 21 and 70 days after AAV-GFP injections. AAV-GFP predominantly transduced neurons, although GFP-positive Müller cells were seen. All neuronal classes were labeled, but the extent of transduction for a given class varied depending on injection age. After P0 injections about 50% of transduced cells were photoreceptors and 30-40% were amacrine or bipolar cells. After adult injections 60-70% of transduced cells were retinal ganglion cells. In adults many GFP-positive retinal axons were traced through the optic nerve/tract and terminal arbors were visualized in central targets.
Collapse
Affiliation(s)
- A R Harvey
- School of Anatomy and Human Biology and Western Australian Institute for Medical Research, The University of Western Australia, Crawley, Western Australia 6009, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Caleo M, Cenni MC, Costa M, Menna E, Zentilin L, Giadrossi S, Giacca M, Maffei L. Expression of BCL-2 via adeno-associated virus vectors rescues thalamic neurons after visual cortex lesion in the adult rat. Eur J Neurosci 2002; 15:1271-7. [PMID: 11994121 DOI: 10.1046/j.1460-9568.2002.01962.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Lesions of the mammalian visual cortex cause the retrograde degeneration of the thalamic neurons projecting to the damaged cortex. The proto-oncogene bcl-2 is known to inhibit neuronal apoptosis induced by a variety of noxious stimuli and preserve the functional integrity of the injured cells. Here we have tested whether the overexpression of bcl-2 via adeno-associated virus (AAV) vectors is able to protect the neurons in the lateral geniculate nucleus after visual cortex ablation in adult rats. Recombinant AAV vectors encoding Bcl-2 (AAV-Bcl-2) or green fluorescent protein (AAV-GFP) as a control were stereotaxically injected into the geniculate. Three weeks after vector injection, the ipsilateral visual cortex was removed by aspiration, and cell survival was assessed 2 weeks later. We found that 20% of the geniculate neurons were transduced by the Bcl-2 vector. These cells were completely protected from death following cortical ablation. Delivery of AAV-GFP transduced an identical number of geniculate neurons but had no effect on cell survival after lesion. The total number of surviving geniculate neurons was found to be significantly higher in animals injected with AAV-Bcl-2 than in rats injected with AAV-GFP or in control lesioned rats. These data indicate that Bcl-2 gene therapy with AAV vectors represents an effective treatment to promote neuronal survival after central nervous system insults.
Collapse
Affiliation(s)
- Matteo Caleo
- Scuola Normale Superiore, P.zza dei Cavalieri 7, 56126 Pisa, Italy.
| | | | | | | | | | | | | | | |
Collapse
|