1
|
Laskowitz DT, Van Wyck DW. ApoE Mimetic Peptides as Therapy for Traumatic Brain Injury. Neurotherapeutics 2023; 20:1496-1507. [PMID: 37592168 PMCID: PMC10684461 DOI: 10.1007/s13311-023-01413-0] [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] [Accepted: 07/17/2023] [Indexed: 08/19/2023] Open
Abstract
The lack of targeted therapies for traumatic brain injury (TBI) remains a compelling clinical unmet need. Although knowledge of the pathophysiologic cascades involved in TBI has expanded rapidly, the development of novel pharmacological therapies has remained largely stagnant. Difficulties in creating animal models that recapitulate the different facets of clinical TBI pathology and flaws in the design of clinical trials have contributed to the ongoing failures in neuroprotective drug development. Furthermore, multiple pathophysiological mechanisms initiated early after TBI that progress in the subacute and chronic setting may limit the potential of traditional approaches that target a specific cellular pathway for acute therapeutic intervention. We describe a reverse translational approach that focuses on translating endogenous mechanisms known to influence outcomes after TBI to develop druggable targets. In particular, numerous clinical observations have demonstrated an association between apolipoprotein E (apoE) polymorphism and functional recovery after brain injury. ApoE has been shown to mitigate the response to acute brain injury by exerting immunomodulatory properties that reduce secondary tissue injury as well as protecting neurons from excitotoxicity. CN-105 represents an apoE mimetic peptide that can effectively penetrate the CNS compartment and retains the neuroprotective properties of the intact protein.
Collapse
Affiliation(s)
- Daniel T Laskowitz
- Department of Neurology, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Neurobiology, Duke University School of Medicine, Durham, NC, 27710, USA
- AegisCN LLC, 701 W Main Street, Durham, NC, 27701, USA
| | - David W Van Wyck
- Department of Neurology, Duke University School of Medicine, Durham, NC, 27710, USA.
| |
Collapse
|
2
|
Eskandari S, Rezayof A, Asghari SM, Hashemizadeh S. Neurobiochemical characteristics of arginine-rich peptides explain their potential therapeutic efficacy in neurodegenerative diseases. Neuropeptides 2023; 101:102356. [PMID: 37390744 DOI: 10.1016/j.npep.2023.102356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/02/2023]
Abstract
Neurodegenerative diseases, including Alzheimer̕ s disease (AD), Parkinson̕ s disease (PD), Huntington̕ s disease (HD), and Amyotrophic Lateral Sclerosis (ALS) require special attention to find new potential treatment methods. This review aims to summarize the current knowledge of the relationship between the biochemical properties of arginine-rich peptides (ARPs) and their neuroprotective effects to deal with the harmful effects of risk factors. It seems that ARPs have portrayed a promising and fantastic landscape for treating neurodegeneration-associated disorders. With multimodal mechanisms of action, ARPs play various unprecedented roles, including as the novel delivery platforms for entering the central nervous system (CNS), the potent antagonists for calcium influx, the invader molecules for targeting mitochondria, and the protein stabilizers. Interestingly, these peptides inhibit the proteolytic enzymes and block protein aggregation to induce pro-survival signaling pathways. ARPs also serve as the scavengers of toxic molecules and the reducers of oxidative stress agents. They also have anti-inflammatory, antimicrobial, and anti-cancer properties. Moreover, by providing an efficient nucleic acid delivery system, ARPs can play an essential role in developing various fields, including gene vaccines, gene therapy, gene editing, and imaging. ARP agents and ARP/cargo therapeutics can be raised as an emergent class of neurotherapeutics for neurodegeneration. Part of the aim of this review is to present recent advances in treating neurodegenerative diseases using ARPs as an emerging and powerful therapeutic tool. The applications and progress of ARPs-based nucleic acid delivery systems have also been discussed to highlight their usefulness as a broad-acting class of drugs.
Collapse
Affiliation(s)
- Sedigheh Eskandari
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran; Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Ameneh Rezayof
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran.
| | - S Mohsen Asghari
- Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran.
| | - Shiva Hashemizadeh
- School of Cognitive Sciences, Institute for Research in Fundamental Sciences, IPM, Tehran, Iran
| |
Collapse
|
3
|
Yan J, Zhang Y, Wang L, Li Z, Tang S, Wang Y, Gu N, Sun X, Li L. TREM2 activation alleviates neural damage via Akt/CREB/BDNF signalling after traumatic brain injury in mice. J Neuroinflammation 2022; 19:289. [PMID: 36463233 PMCID: PMC9719652 DOI: 10.1186/s12974-022-02651-3] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 11/21/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Neuroinflammation is one of the most important processes in secondary injury after traumatic brain injury (TBI). Triggering receptor expressed on myeloid cells 2 (TREM2) has been proven to exert neuroprotective effects in neurodegenerative diseases and stroke by modulating neuroinflammation, and promoting phagocytosis and cell survival. However, the role of TREM2 in TBI has not yet been elucidated. In this study, we are the first to use COG1410, an agonist of TREM2, to assess the effects of TREM2 activation in a murine TBI model. METHODS Adult male wild-type (WT) C57BL/6 mice and adult male TREM2 KO mice were subjected to different treatments. TBI was established by the controlled cortical impact (CCI) method. COG1410 was delivered 1 h after CCI via tail vein injection. Western blot analysis, immunofluorescence, laser speckle contrast imaging (LSCI), neurological behaviour tests, brain electrophysiological monitoring, Evans blue assays, magnetic resonance imaging (MRI), and brain water content measurement were performed in this study. RESULTS The expression of endogenous TREM2 peaked at 3 d after CCI, and it was mainly expressed on microglia and neurons. We found that COG1410 improved neurological functions within 3 d, as well as neurological functions and brain electrophysiological activity at 2 weeks after CCI. COG1410 exerted neuroprotective effects by inhibiting neutrophil infiltration and microglial activation, and suppressing neuroinflammation after CCI. In addition, COG1410 treatment alleviated blood brain barrier (BBB) disruption and brain oedema; furthermore, COG1410 promoted cerebral blood flow (CBF) recovery at traumatic injury sites after CCI. In addition, COG1410 suppressed neural apoptosis at 3 d after CCI. TREM2 activation upregulated p-Akt, p-CREB, BDNF, and Bcl-2 and suppressed TNF-α, IL-1β, Bax, and cleaved caspase-3 at 3 d after CCI. Moreover, TREM2 knockout abolished the effects of COG1410 on vascular phenotypes and microglial states. Finally, the neuroprotective effects of COG1410 were suppressed by TREM2 depletion. CONCLUSIONS Altogether, we are the first to demonstrate that TREM2 activation by COG1410 alleviated neural damage through activation of Akt/CREB/BDNF signalling axis in microglia after CCI. Finally, COG1410 treatment improved neurological behaviour and brain electrophysiological activity after CCI.
Collapse
Affiliation(s)
- Jin Yan
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd, Chongqing, 400016, China
| | - Yuan Zhang
- Department of Neurosurgery, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
| | - Lin Wang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd, Chongqing, 400016, China
- Department of Neurosurgery, Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical College, Nanchong, China
| | - Zhao Li
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd, Chongqing, 400016, China
- Department of Neurosurgery, Chengdu Integrated TCM & Western Medicine Hospital, Chengdu, China
| | - Shuang Tang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd, Chongqing, 400016, China
- Department of Neurosurgery, Suining Central Hospital, Suining, China
| | - Yingwen Wang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd, Chongqing, 400016, China
| | - Nina Gu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd, Chongqing, 400016, China
| | - Xiaochuan Sun
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd, Chongqing, 400016, China.
| | - Lin Li
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, 1 Youyi Rd, Chongqing, 400016, China.
- Department of Neuro-oncology, Chongqing University Cancer Hospital, Chongqing, China.
- Department of Neurosurgery, The Affiliated Hospital of North Sichuan Medical College, Nanchong, China.
| |
Collapse
|
4
|
Van Wyck D, Kolls BJ, Wang H, Cantillana V, Maughan M, Laskowitz DT. Prophylactic treatment with CN-105 improves functional outcomes in a murine model of closed head injury. Exp Brain Res 2022; 240:2413-2423. [PMID: 35841411 DOI: 10.1007/s00221-022-06417-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 07/04/2022] [Indexed: 11/30/2022]
Abstract
The treatment of traumatic brain injury (TBI) in military populations is hindered by underreporting and underdiagnosis. Clinical symptoms and outcomes may be mitigated with an effective pre-injury prophylaxis. This study evaluates whether CN-105, a 5-amino acid apolipoprotein E (ApoE) mimetic peptide previously shown to modify the post-traumatic neuroinflammatory response, would maintain its neuroprotective effects if administered prior to closed-head injury in a clinically relevant murine model. CN-105 was synthesized by Polypeptide Inc. (San Diego, CA) and administered to C57-BL/6 mice intravenously (IV) and/or by intraperitoneal (IP) injection at various time points prior to injury while vehicle treated animals received IV and/or IP normal saline. Animals were randomized following injury and behavioral observations were conducted by investigators blinded to treatment. Vestibulomotor function was assessed using an automated Rotarod (Ugo Basile, Comerio, Italy), and hippocampal microglial activation was assessed using F4/80 immunohistochemical staining in treated and untreated mice 7 days post-TBI. Separate, in vivo assessments of the pharmacokinetics was performed in healthy CD-1. IV CN-105 administered prior to head injury improved vestibulomotor function compared to vehicle control-treated animals. CN-105 co-administered by IP and IV dosing 6 h prior to injury also improved vestibulomotor function up to 28 days following injury. Microglia counted in CN-105 treated specimens were significantly fewer (P = 0.03) than in vehicle specimens. CN-105 improves functional outcomes and reduces hippocampal microglial activation when administered prior to injury and could be adapted as a pre-injury prophylaxis for soldiers at high risk for TBI.
Collapse
Affiliation(s)
- David Van Wyck
- 3Rd Special Forces Group (A), U.S. Army Special Operations Command, 111 Enduring Freedom Drive (Stop A), Fort Bragg, NC, 28310, USA. .,Department of Neurology, Duke University School of Medicine, Durham, NC, 27710, USA.
| | - Bradley J Kolls
- Department of Neurology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Haichen Wang
- Department of Neurology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Viviana Cantillana
- Department of Neurology, Duke University School of Medicine, Durham, NC, 27710, USA
| | | | - Daniel T Laskowitz
- Department of Neurology, Duke University School of Medicine, Durham, NC, 27710, USA.,Department of Neurobiology, Duke University School of Medicine, Durham, NC, 27710, USA.,Aegis-CN LLC., 701 W Main Street, Durham, NC, 27701, USA
| |
Collapse
|
5
|
Ahmed S, Pande AH, Sharma SS. Therapeutic potential of ApoE-mimetic peptides in CNS disorders: Current perspective. Exp Neurol 2022; 353:114051. [DOI: 10.1016/j.expneurol.2022.114051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/23/2022] [Accepted: 03/14/2022] [Indexed: 02/07/2023]
|
6
|
Hodgetts SI, Lovett SJ, Baron-Heeris D, Fogliani A, Sturm M, Van den Heuvel C, Harvey AR. Effects of amyloid precursor protein peptide APP96-110, alone or with human mesenchymal stromal cells, on recovery after spinal cord injury. Neural Regen Res 2021; 17:1376-1386. [PMID: 34782585 PMCID: PMC8643048 DOI: 10.4103/1673-5374.327357] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Delivery of a peptide (APP96-110), derived from amyloid precursor protein (APP), has been shown to elicit neuroprotective effects following cerebral stroke and traumatic brain injury. In this study, the effect of APP96-110 or a mutant version of this peptide (mAPP96-110) was assessed following moderate (200 kdyn, (2 N)) thoracic contusive spinal cord injury (SCI) in adult Nude rats. Animals received a single tail vein injection of APP96-110 or mAPP96-110 at 30 minutes post-SCI and were then assessed for functional improvements over the next 8 weeks. A cohort of animals also received transplants of either viable or non-viable human mesenchymal stromal cells (hMSCs) into the SC lesion site at one week post-injury to assess the effect of combining intravenous APP96-110 delivery with hMSC treatment. Rats were perfused 8 weeks post-SCI and longitudinal sections of spinal cord analyzed for a number of factors including hMSC viability, cyst size, axonal regrowth, glial reactivity and macrophage activation. Analysis of sensorimotor function revealed occasional significant differences between groups using Ladderwalk or Ratwalk tests, however there were no consistent improvements in functional outcome after any of the treatments. mAPP96-110 alone, and APP96-110 in combination with both viable and non-viable hMSCs significantly reduced cyst size compared to SCI alone. Combined treatments with donor hMSCs also significantly increased βIII tubulin+, glial fibrillary acidic protein (GFAP+) and laminin+ expression, and decreased ED1+ expression in tissues. This preliminary study demonstrates that intravenous delivery of APP96-110 peptide has selective, modest neuroprotective effects following SCI, which may be enhanced when combined with hMSC transplantation. However, the effects are less pronounced and less consistent compared to the protective morphological and cognitive impact that this same peptide has on neuronal survival and behaviour after stroke and traumatic brain injury. Thus while the efficacy of a particular therapeutic approach in one CNS injury model may provide justification for its use in other neurotrauma models, similar outcomes may not necessarily occur and more targeted approaches suited to location and severity are required. All animal experiments were approved by The University of Western Australia Animal Ethics Committee (RA3/100/1460) on April 12, 2016.
Collapse
Affiliation(s)
- Stuart I Hodgetts
- School of Human Sciences, The University of Western Australia (UWA); Perron Institute for Neurological and Translational Science, Perth, WA, Australia
| | - Sarah J Lovett
- School of Human Sciences, The University of Western Australia (UWA), Perth, WA, Australia
| | - D Baron-Heeris
- School of Human Sciences, The University of Western Australia (UWA), Perth, WA, Australia
| | - A Fogliani
- School of Human Sciences, The University of Western Australia (UWA), Perth, WA, Australia
| | - Marian Sturm
- Cell and Tissue Therapies WA (CTTWA), Royal Perth Hospital, Perth, WA, Australia
| | - C Van den Heuvel
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Alan R Harvey
- School of Human Sciences, The University of Western Australia (UWA); Perron Institute for Neurological and Translational Science, Perth, WA, Australia
| |
Collapse
|
7
|
James ML, Troy J, Nowacki N, Komisarow J, Swisher CB, Tucker K, Hatton K, Babi MA, Worrall BB, Andrews C, Woo D, Kranz PG, Lascola C, Maughan M, Laskowitz DT. CN-105 in Participants with Acute Supratentorial Intracerebral Hemorrhage (CATCH) Trial. Neurocrit Care 2021; 36:216-225. [PMID: 34424490 DOI: 10.1007/s12028-021-01287-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 05/21/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Endogenous apolipoprotein (apo) E mediates neuroinflammatory responses and recovery after brain injury. Exogenously administered apoE-mimetic peptides effectively penetrate the central nervous system compartment and downregulate acute inflammation. CN-105 is a novel apoE-mimetic pentapeptide with excellent evidence of functional and histological improvement in preclinical models of intracerebral hemorrhage (ICH). The CN-105 in participants with Acute supraTentorial intraCerebral Hemorrhage (CATCH) trial is a first-in-disease-state multicenter open-label trial evaluating safety and feasability of CN-105 administration in patients with acute primary supratentorial ICH. METHODS Eligible patients were aged 30-80 years, had confirmed primary supratentorial ICH, and were able to intiate CN-105 administration (1.0 mg/kg every 6 h for 72 h) within 12 h of symptom onset. A priori defined safety end points, including hematoma volume, pharmacokinetics, and 30-day neurological outcomes, were analyzed. For clinical outcomes, CATCH participants were compared 1:1 with a closely matched contemporary ICH cohort through random selection. Hematoma volumes determined from computed tomography images on days 0, 1, 2, and 5 and ordinal modified Rankin Scale score at 30 days after ICH were compared. RESULTS In 38 participants enrolled across six study sites in the United States, adverse events occurred at an expected rate without increase in hematoma expansion or neurological deterioration. CN-105 treatment had an odds ratio (95% confidence interval) of 2.69 (1.31-5.51) for lower 30-day modified Rankin Scale score, after adjustment for ICH score, sex, and race/ethnicity, as compared with a matched contemporary cohort. CONCLUSIONS CN-105 administration represents an excellent translational candidate for treatment of acute ICH because of its safety, dosing feasibility, favorable pharmacokinetics, and possible improvement in neurological recovery.
Collapse
Affiliation(s)
- Michael L James
- Department of Anesthesiology, Duke University, Durham, NC, USA. .,Department of Neurology, Duke University, Durham, NC, USA. .,Duke Clinical Research Institute, Duke University, Durham, NC, USA.
| | - Jesse Troy
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | | | | | | | - Kristi Tucker
- Department of Neurology, Wake Forest-Baptist Health, Winston-Salem, NC, USA
| | - Kevin Hatton
- Department of Anesthesiology, University of Kentucky, Lexington, KY, USA
| | - Marc A Babi
- Departments of Neurology and Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Bradford B Worrall
- Departments of Neurology and Public Health Sciences, University of Virginia, Charlottesvile, VA, USA
| | - Charles Andrews
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, USA
| | - Daniel Woo
- Department of Neurology, University of Cincinnati, Cincinnati, OH, USA
| | - Peter G Kranz
- Department of Radiology, Duke University, Durham, NC, USA
| | | | | | - Daniel T Laskowitz
- Department of Anesthesiology, Duke University, Durham, NC, USA.,Department of Neurology, Duke University, Durham, NC, USA.,Duke Clinical Research Institute, Duke University, Durham, NC, USA.,AegisCN, LLC, Durham, NC, USA
| | | |
Collapse
|
8
|
Klegeris A. Targeting neuroprotective functions of astrocytes in neuroimmune diseases. Expert Opin Ther Targets 2021; 25:237-241. [PMID: 33836642 DOI: 10.1080/14728222.2021.1915993] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Andis Klegeris
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, British Columbia, Canada
| |
Collapse
|
9
|
James ML, Komisarow JM, Wang H, Laskowitz DT. Therapeutic Development of Apolipoprotein E Mimetics for Acute Brain Injury: Augmenting Endogenous Responses to Reduce Secondary Injury. Neurotherapeutics 2020; 17:475-483. [PMID: 32318912 PMCID: PMC7283431 DOI: 10.1007/s13311-020-00858-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Over the last few decades, increasing evidence demonstrates that the neuroinflammatory response is a double-edged sword. Although overly robust inflammatory responses may exacerbate secondary tissue injury, inflammatory processes are ultimately necessary for recovery. Traditional drug discovery often relies on reductionist approaches to isolate and modulate specific intracellular pathways believed to be involved in disease pathology. However, endogenous brain proteins are often pleiotropic in order to regulate neuroinflammation and recovery mechanisms. Thus, a process of "backward translation" aims to harness the adaptive properties of endogenous proteins to promote earlier and greater recovery after acute brain injury. One such endogenous protein is apolipoprotein E (apoE), the primary apolipoprotein produced in the brain. Robust preclinical and clinical evidence demonstrates that endogenous apoE produced within the brain modulates the neuroinflammatory response of the acutely injured brain. Thus, one innovative approach to improve outcomes following acute brain injury is administration of exogenous apoE-mimetic drugs optimized to cross the blood-brain barrier. In particular, one promising apoE mimetic peptide, CN-105, has demonstrated efficacy across a wide variety of preclinical models of brain injury and safety and feasibility in early-phase clinical trials. Preclinical and clinical evidence for apoE's neuroprotective effects and downregulation of neuroinflammatory and the resulting translational therapeutic development strategy for an apoE-based therapeutic are reviewed.
Collapse
Affiliation(s)
- Michael L James
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
- Department of Neurology, Duke University School of Medicine, Durham, NC, USA
- Department of Anesthesiology, Duke University School of Medicine, Durham, NC, USA
| | - Jordan M Komisarow
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC, USA
| | - Haichen Wang
- Department of Neurology, Duke University School of Medicine, Durham, NC, USA
| | - Daniel T Laskowitz
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA.
- Department of Neurology, Duke University School of Medicine, Durham, NC, USA.
- Department of Anesthesiology, Duke University School of Medicine, Durham, NC, USA.
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
10
|
Meloni BP, Mastaglia FL, Knuckey NW. Cationic Arginine-Rich Peptides (CARPs): A Novel Class of Neuroprotective Agents With a Multimodal Mechanism of Action. Front Neurol 2020; 11:108. [PMID: 32158425 PMCID: PMC7052017 DOI: 10.3389/fneur.2020.00108] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 01/30/2020] [Indexed: 12/17/2022] Open
Abstract
There are virtually no clinically available neuroprotective drugs for the treatment of acute and chronic neurological disorders, hence there is an urgent need for the development of new neuroprotective molecules. Cationic arginine-rich peptides (CARPs) are an expanding and relatively novel class of compounds, which possess intrinsic neuroprotective properties. Intriguingly, CARPs possess a combination of biological properties unprecedented for a neuroprotective agent including the ability to traverse cell membranes and enter the CNS, antagonize calcium influx, target mitochondria, stabilize proteins, inhibit proteolytic enzymes, induce pro-survival signaling, scavenge toxic molecules, and reduce oxidative stress as well as, having a range of anti-inflammatory, analgesic, anti-microbial, and anti-cancer actions. CARPs have also been used as carrier molecules for the delivery of other putative neuroprotective agents across the blood-brain barrier and blood-spinal cord barrier. However, there is increasing evidence that the neuroprotective efficacy of many, if not all these other agents delivered using a cationic arginine-rich cell-penetrating peptide (CCPPs) carrier (e.g., TAT) may actually be mediated largely by the properties of the carrier molecule, with overall efficacy further enhanced according to the amino acid composition of the cargo peptide, in particular its arginine content. Therefore, in reviewing the neuroprotective mechanisms of action of CARPs we also consider studies using CCPPs fused to a putative neuroprotective peptide. We review the history of CARPs in neuroprotection and discuss in detail the intrinsic biological properties that may contribute to their cytoprotective effects and their usefulness as a broad-acting class of neuroprotective drugs.
Collapse
Affiliation(s)
- Bruno P Meloni
- Department of Neurosurgery, QEII Medical Centre, Sir Charles Gairdner Hospital, Nedlands, WA, Australia.,Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, WA, Australia
| | - Frank L Mastaglia
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, WA, Australia
| | - Neville W Knuckey
- Department of Neurosurgery, QEII Medical Centre, Sir Charles Gairdner Hospital, Nedlands, WA, Australia.,Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|
11
|
Chiu LS, Anderton RS, Cross JL, Clark VW, Knuckey NW, Meloni BP. Poly-arginine Peptide R18D Reduces Neuroinflammation and Functional Deficits Following Traumatic Brain Injury in the Long-Evans Rat. Int J Pept Res Ther 2019. [DOI: 10.1007/s10989-018-09799-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
12
|
Asam K, Staniszewski A, Zhang H, Melideo SL, Mazzeo A, Voronkov M, Huber KL, Pérez E, Stock M, Stock JB, Arancio O, Nicholls RE. Eicosanoyl-5-hydroxytryptamide (EHT) prevents Alzheimer's disease-related cognitive and electrophysiological impairments in mice exposed to elevated concentrations of oligomeric beta-amyloid. PLoS One 2017; 12:e0189413. [PMID: 29253878 PMCID: PMC5734769 DOI: 10.1371/journal.pone.0189413] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 11/24/2017] [Indexed: 02/04/2023] Open
Abstract
Soluble forms of oligomeric beta-amyloid (Aβ) are thought to play a central role in Alzheimer's disease (AD). Transgenic manipulation of methylation of the serine/threonine protein phosphatase, PP2A, was recently shown to alter the sensitivity of mice to AD-related impairments resulting from acute exposure to elevated levels of Aβ. In addition, eicosanoyl-5-hydroxytryptamide (EHT), a naturally occurring component from coffee beans that modulates PP2A methylation, was shown to confer therapeutic benefits in rodent models of AD and Parkinson's disease. Here, we tested the hypothesis that EHT protects animals from the pathological effects of exposure to elevated levels of soluble oligomeric Aβ. We treated mice with EHT-containing food at two different doses and assessed the sensitivity of these animals to Aβ-induced behavioral and electrophysiological impairments. We found that EHT administration protected animals from Aβ-induced cognitive impairments in both a radial-arm water maze and contextual fear conditioning task. We also found that both chronic and acute EHT administration prevented Aβ-induced impairments in long-term potentiation. These data add to the accumulating evidence suggesting that interventions with pharmacological agents, such as EHT, that target PP2A activity may be therapeutically beneficial for AD and other neurological conditions.
Collapse
Affiliation(s)
- Kesava Asam
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States of America
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, United States of America
| | - Agnieszka Staniszewski
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States of America
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, United States of America
| | - Hong Zhang
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States of America
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, United States of America
| | - Scott L. Melideo
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Adolfo Mazzeo
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States of America
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, United States of America
| | - Michael Voronkov
- Signum Biosciences, 133 Wall Street, Princeton, New Jersey, United States of America
| | - Kristen L. Huber
- Signum Biosciences, 133 Wall Street, Princeton, New Jersey, United States of America
| | - Eduardo Pérez
- Signum Biosciences, 133 Wall Street, Princeton, New Jersey, United States of America
| | - Maxwell Stock
- Signum Biosciences, 133 Wall Street, Princeton, New Jersey, United States of America
| | - Jeffry B. Stock
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
- Signum Biosciences, 133 Wall Street, Princeton, New Jersey, United States of America
| | - Ottavio Arancio
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States of America
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, United States of America
- Department of Medicine, Columbia University, New York, NY, United States of America
| | - Russell E. Nicholls
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States of America
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, United States of America
- * E-mail:
| |
Collapse
|
13
|
Chiu LS, Anderton RS, Cross JL, Clark VW, Edwards AB, Knuckey NW, Meloni BP. Assessment of R18, COG1410, and APP96-110 in Excitotoxicity and Traumatic Brain Injury. Transl Neurosci 2017; 8:147-157. [PMID: 29177102 PMCID: PMC5700203 DOI: 10.1515/tnsci-2017-0021] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 10/25/2017] [Indexed: 01/02/2023] Open
Abstract
Cationic arginine-rich and poly-arginine peptides (referred to as CARPs) have potent neuroprotective properties in in vitro excitotoxicity and in vivo models of stroke. Traumatic brain injury (TBI) shares many pathophysiological processes as stroke, including excitotoxicity. Therefore, we evaluated our lead peptide, poly-arginine R18, with the COG1410 and APP96-110 peptides, which have neuroprotective actions following TBI. In an in vitro cortical neuronal glutamic acid excitotoxicity injury model, R18 was highly neuroprotective and reduced neuronal calcium influx, while COG1410 and APP96-110 displayed modest neuroprotection and were less effective at reducing calcium influx. In an impact-acceleration closed-head injury model (Marmarou model), R18, COG1410, and APP96-110 were administered intravenously (300 nmol/kg) at 30 minutes after injury in male Sprague-Dawley rats. When compared to vehicle, no peptide significantly improved functional outcomes, however the R18 and COG1410 treatment groups displayed positive trends in the adhesive tape test and rotarod assessments. Similarly, no peptide had a significant effect on hippocampal neuronal loss, however a significant reduction in axonal injury was observed for R18 and COG1410. In conclusion, this study has demonstrated that R18 is significantly more effective than COG1410 and APP96-110 at reducing neuronal injury and calcium influx following excitotoxicity, and that both R18 and COG1410 reduce axonal injury following TBI. Additional dose response and treatment time course studies are required to further assess the efficacy of R18 in TBI.
Collapse
Affiliation(s)
- Li Shan Chiu
- Perron Institute for Neurological and Translational Sciences, Nedlands, Western Australia, 6009, Australia.,Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, Western Australia, 6009, Australia
| | - Ryan S Anderton
- Perron Institute for Neurological and Translational Sciences, Nedlands, Western Australia, 6009, Australia.,Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, Western Australia, 6009, Australia.,School of Heath Sciences, The University Notre Dame Australia, Fremantle, Western Australia, 6160, Australia.,Institute for Health Research, The University Notre Dame Australia, Fremantle, Western Australia, 6160, Australia
| | - Jane L Cross
- Perron Institute for Neurological and Translational Sciences, Nedlands, Western Australia, 6009, Australia.,Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, Western Australia, 6009, Australia.,Department of Neurosurgery, Sir Charles Gairdner Hospital, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia
| | - Vince W Clark
- Perron Institute for Neurological and Translational Sciences, Nedlands, Western Australia, 6009, Australia.,Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, Western Australia, 6009, Australia.,Department of Neurosurgery, Sir Charles Gairdner Hospital, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia
| | - Adam B Edwards
- Perron Institute for Neurological and Translational Sciences, Nedlands, Western Australia, 6009, Australia.,School of Heath Sciences, The University Notre Dame Australia, Fremantle, Western Australia, 6160, Australia
| | - Neville W Knuckey
- Perron Institute for Neurological and Translational Sciences, Nedlands, Western Australia, 6009, Australia.,Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, Western Australia, 6009, Australia.,Department of Neurosurgery, Sir Charles Gairdner Hospital, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia
| | - Bruno P Meloni
- Perron Institute for Neurological and Translational Sciences, Nedlands, Western Australia, 6009, Australia.,Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, Western Australia, 6009, Australia.,Department of Neurosurgery, Sir Charles Gairdner Hospital, QEII Medical Centre, Nedlands, Western Australia, 6009, Australia
| |
Collapse
|
14
|
Lam PK, Wang KKW, Lo AWI, Tong CSW, Ching DWC, Wong K, Yang Z, Kong T, Lo KKY, Choy RKW, Lai PBS, Wong GKC, Poon WS. Interactome and reciprocal activation of pathways in topical mesenchymal stem cells and the recipient cerebral cortex following traumatic brain injury. Sci Rep 2017; 7:5017. [PMID: 28694468 PMCID: PMC5504061 DOI: 10.1038/s41598-017-01772-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 03/31/2017] [Indexed: 11/12/2022] Open
Abstract
In this study, GFP-MSCs were topically applied to the surface of cerebral cortex within 1 hour of experimental TBI. No treatment was given to the control group. Three days after topical application, the MSCs homed to the injured parenchyma and improved the neurological function. Topical MSCs triggered earlier astrocytosis and reactive microglia. TBI penumbra and hippocampus had higher cellular proliferation. Apoptosis was suppressed at hippocampus at 1 week and reduced neuronal damaged was found in the penumbral at day 14 apoptosis. Proteolytic neuronal injury biomarkers (alphaII-spectrin breakdown products, SBDPs) and glial cell injury biomarker, glial fibrillary acidic protein (GFAP)-breakdown product (GBDPs) in injured cortex were also attenuated by MSCs. In the penumbra, six genes related to axongenesis (Erbb2); growth factors (Artn, Ptn); cytokine (IL3); cell cycle (Hdac4); and notch signaling (Hes1) were up-regulated three days after MSC transplant. Transcriptome analysis demonstrated that 7,943 genes were differentially expressed and 94 signaling pathways were activated in the topical MSCs transplanted onto the cortex of brain injured rats with TBI. In conclusion, topical application offers a direct and efficient delivery of MSCs to the brain.
Collapse
Affiliation(s)
- Ping K Lam
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong, SAR, China
- Chow Tai Fook-Cheng Yu Tung Surgical Stem Cell Research Center, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Kevin K W Wang
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Departments of Psychiatry, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Anthony W I Lo
- Department of Anatomical & Cellular Pathology, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Cindy S W Tong
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong, SAR, China
- Chow Tai Fook-Cheng Yu Tung Surgical Stem Cell Research Center, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Don W C Ching
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong, SAR, China
- Department of Anatomical & Cellular Pathology, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Kenneth Wong
- Department of Obstetrics and Gynecology, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Zhihui Yang
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Departments of Psychiatry, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Themis Kong
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong, SAR, China
- Chow Tai Fook-Cheng Yu Tung Surgical Stem Cell Research Center, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Kin K Y Lo
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong, SAR, China
- Chow Tai Fook-Cheng Yu Tung Surgical Stem Cell Research Center, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Richard K W Choy
- Department of Obstetrics and Gynecology, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Paul B S Lai
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong, SAR, China
- Chow Tai Fook-Cheng Yu Tung Surgical Stem Cell Research Center, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - George K C Wong
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong, SAR, China
- Chow Tai Fook-Cheng Yu Tung Surgical Stem Cell Research Center, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | - Wai S Poon
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong, SAR, China.
- Chow Tai Fook-Cheng Yu Tung Surgical Stem Cell Research Center, The Chinese University of Hong Kong, Hong Kong, SAR, China.
| |
Collapse
|
15
|
Laskowitz DT, Wang H, Chen T, Lubkin DT, Cantillana V, Tu TM, Kernagis D, Zhou G, Macy G, Kolls BJ, Dawson HN. Neuroprotective pentapeptide CN-105 is associated with reduced sterile inflammation and improved functional outcomes in a traumatic brain injury murine model. Sci Rep 2017; 7:46461. [PMID: 28429734 PMCID: PMC5399447 DOI: 10.1038/srep46461] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 03/16/2017] [Indexed: 12/31/2022] Open
Abstract
At present, there are no proven pharmacological treatments demonstrated to improve long term functional outcomes following traumatic brain injury(TBI). In the setting of non-penetrating TBI, sterile brain inflammatory responses are associated with the development of cerebral edema, intracranial hypertension, and secondary neuronal injury. There is increasing evidence that endogenous apolipoprotein E(apoE) modifies the neuroinflammatory response through its role in downregulating glial activation, however, the intact apoE holoprotein does not cross the blood-brain barrier due to its size. To address this limitation, we developed a small 5 amino acid apoE mimetic peptide(CN-105) that mimics the polar face of the apoE helical domain involved in receptor interactions. The goal of this study was to investigate the therapeutic potential of CN-105 in a murine model of closed head injury. Treatment with CN-105 was associated with a durable improvement in functional outcomes as assessed by Rotarod and Morris Water Maze and a reduction in positive Fluoro-Jade B stained injured neurons and microglial activation. Administration of CN-105 was also associated with reduction in mRNA expression of a subset of inflammatory and immune-related genes.
Collapse
Affiliation(s)
- Daniel T Laskowitz
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Neurobiology, Duke University School of Medicine, Durham, NC 27710, USA.,Aegis-CN LLC., Durham, NC, USA
| | - Haichen Wang
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Tony Chen
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
| | - David T Lubkin
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Viviana Cantillana
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Tian Ming Tu
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore
| | - Dawn Kernagis
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Guanen Zhou
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Gary Macy
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Bradley J Kolls
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Hana N Dawson
- Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
16
|
Tu TM, Kolls BJ, Soderblom EJ, Cantillana V, Ferrell PD, Moseley MA, Wang H, Dawson HN, Laskowitz DT. Apolipoprotein E mimetic peptide, CN-105, improves outcomes in ischemic stroke. Ann Clin Transl Neurol 2017; 4:246-265. [PMID: 28382306 PMCID: PMC5376751 DOI: 10.1002/acn3.399] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 01/09/2017] [Accepted: 02/06/2017] [Indexed: 01/05/2023] Open
Abstract
Objective At present, the absence of a pharmacological neuroprotectant represents an important unmet clinical need in the treatment of ischemic and traumatic brain injury. Recent evidence suggests that administration of apolipoprotein E mimetic therapies represent a viable therapeutic strategy in this setting. We investigate the neuroprotective and anti‐inflammatory properties of the apolipoprotein E mimetic pentapeptide, CN‐105, in a microglial cell line and murine model of ischemic stroke. Methods Ten to 13‐week‐old male C57/BL6 mice underwent transient middle cerebral artery occlusion and were randomly selected to receive CN‐105 (0.1 mg/kg) in 100 μL volume or vehicle via tail vein injection at various time points. Survival, motor‐sensory functional outcomes using rotarod test and 4‐limb wire hanging test, infarct volume assessment using 2,3,5‐Triphenyltetrazolium chloride staining method, and microglial activation in the contralateral hippocampus using F4/80 immunostaining were assessed at various time points. In vitro assessment of tumor necrosis factor‐alpha secretion in a microglial cell line was performed, and phosphoproteomic analysis conducted to explore early mechanistic pathways of CN‐105 in ischemic stroke. Results Mice receiving CN‐105 demonstrated improved survival, improved functional outcomes, reduced infarct volume, and reduced microglial activation. CN‐105 also suppressed inflammatory cytokines secretion in microglial cells in vitro. Phosphoproteomic signals suggest that CN‐105 reduces proinflammatory pathways and lower oxidative stress. Interpretation CN‐105 improves functional and histological outcomes in a murine model of ischemic stroke via modulation of neuroinflammatory pathways. Recent clinical trial of this compound has demonstrated favorable pharmacokinetic and safety profile, suggesting that CN‐105 represents an attractive candidate for clinical translation.
Collapse
Affiliation(s)
- Tian Ming Tu
- Department of Neurology Duke University School of Medicine Durham North Carolina; Department of Neurology National Neuroscience Institute Tan Tock Seng Campus Singapore
| | - Brad J Kolls
- Department of Neurology Duke University School of Medicine Durham North Carolina
| | - Erik J Soderblom
- Duke Proteomics Core Facility Center for Genomic and Computational Biology Duke University Durham North Carolina
| | - Viviana Cantillana
- Department of Neurology Duke University School of Medicine Durham North Carolina
| | - Paul Durham Ferrell
- Department of Pathology Duke University School of Medicine Durham North Carolina
| | - M Arthur Moseley
- Duke Proteomics Core Facility Center for Genomic and Computational Biology Duke University Durham North Carolina
| | - Haichen Wang
- Department of Neurology Duke University School of Medicine Durham North Carolina
| | - Hana N Dawson
- Department of Neurology Duke University School of Medicine Durham North Carolina
| | - Daniel T Laskowitz
- Department of Neurology Duke University School of Medicine Durham North Carolina
| |
Collapse
|
17
|
Guptill JT, Raja SM, Boakye-Agyeman F, Noveck R, Ramey S, Tu TM, Laskowitz DT. Phase 1 Randomized, Double-Blind, Placebo-Controlled Study to Determine the Safety, Tolerability, and Pharmacokinetics of a Single Escalating Dose and Repeated Doses of CN-105 in Healthy Adult Subjects. J Clin Pharmacol 2016; 57:770-776. [PMID: 27990643 DOI: 10.1002/jcph.853] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 11/10/2016] [Indexed: 11/11/2022]
Abstract
Spontaneous intracranial hemorrhage (ICH) remains a devastating stroke subtype, affecting as many as 80,000 people annually in the United States and associated with extremely high mortality. In the absence of any pharmacological interventions demonstrated to improve outcome, care for patients with ICH remains largely supportive. Thus, despite advances in the understanding of ICH and brain injury, there remains an unmet need for interventions that improve neurologic recovery and outcomes. Recent research suggesting inflammation and APOE genotype play a role in modifying neurologic outcome after brain injury has led to the development of an APOE-derived peptide agent (CN-105). Preclinical studies have demonstrated that CN-105 effectively downregulates the inflammatory response in acute brain injury, including ICH. Following Investigational New Drug (IND) enabling studies in murine models, this first-in-human single escalating dose and multiple dose placebo-controlled clinical trial was performed to define the safety and pharmacokinetics (PK) of CN-105. A total of 48 subjects (12 control, 36 active) were randomized in this study; all subjects completed the study. No significant safety issues were identified with both dosing regimens, and PK analysis revealed linearity without significant drug accumulation. The median half-life in the terminal elimination phase of CN-105 following a single or repeated dosing regimen did not change (approximately 3.6 hours). With the PK and preliminary safety of CN-105 established, the drug is now poised to begin first-in-disease phase 2 clinical trials in patients with ICH who urgently need new therapeutic options.
Collapse
Affiliation(s)
- Jeffrey T Guptill
- Department of Neurology, Duke University, Durham, NC, USA.,Duke Clinical Research Institute, Durham, NC, USA
| | - Shruti M Raja
- Department of Neurology, Duke University, Durham, NC, USA.,Duke Clinical Research Institute, Durham, NC, USA
| | | | | | - Sarah Ramey
- Duke Clinical Research Institute, Durham, NC, USA
| | - Tian Ming Tu
- SingHealth, National Neuroscience Institute, Singapore, Republic of Singapore
| | - Daniel T Laskowitz
- Department of Neurology, Duke University, Durham, NC, USA.,Duke Clinical Research Institute, Durham, NC, USA
| |
Collapse
|
18
|
Peptide Pharmacological Approaches to Treating Traumatic Brain Injury: a Case for Arginine-Rich Peptides. Mol Neurobiol 2016; 54:7838-7857. [PMID: 27844291 DOI: 10.1007/s12035-016-0287-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 11/02/2016] [Indexed: 01/25/2023]
Abstract
Traumatic brain injury (TBI) has a devastating effect on victims and their families, and has profound negative societal and economic impacts, a situation that is further compounded by the lack of effective treatments to minimise injury after TBI. The current strategy for managing TBI is partly through preventative measures and partly through surgical and rehabilitative interventions. Secondary brain damage remains the principal focus for the development of a neuroprotective therapeutic. However, the complexity of TBI pathophysiology has meant that single-action pharmacological agents have been largely unsuccessful in combatting the associated brain injury cascades, while combination therapies to date have proved equally ineffective. Peptides have recently emerged as promising lead agents for the treatment of TBI, especially those rich in the cationic amino acid, arginine. Having been shown to lessen the impact of ischaemic stroke in animal models, there are reasonable grounds to believe that arginine-rich peptides may have neuroprotective therapeutic potential in TBI. Here, we review a range of peptides previously examined as therapeutic agents for TBI. In particular, we focus on cationic arginine-rich peptides -- a new class of agents that growing evidence suggests acts through multiple neuroprotective mechanisms.
Collapse
|
19
|
Neuroprotective pentapeptide CN-105 improves functional and histological outcomes in a murine model of intracerebral hemorrhage. Sci Rep 2016; 6:34834. [PMID: 27713572 PMCID: PMC5054364 DOI: 10.1038/srep34834] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 08/24/2016] [Indexed: 01/18/2023] Open
Abstract
Presently, no pharmacological treatments have been demonstrated to improve long-term functional outcomes following intracerebral hemorrhage (ICH). Clinical evidence associates apolipoprotein E (apoE) genotype with ICH incidence and outcome. While apoE modifies neuroinflammatory responses through its adaptive role in glial downregulation, intact apoE holoprotein is too large to cross the blood-brain barrier (BBB). Therefore, we developed a 5-amino acid peptide – CN-105 – that mimics the polar face of the apoE helical domain involved in receptor interactions. In the current study, we investigated the therapeutic potential of CN-105 in a mouse model of ICH. Three doses of CN-105 (0.05 mg/kg) was administered by tail vein injection within 24 hours after ICH induction. Functional assessment showed durable improvement in vestibulomotor performance after CN-105 treatment, as quantified by increased Rotarod latencies on Days 1–5 post-ICH, and long-term improvement in neurocognitive performance, as quantified by reduced Morris water maze latencies on Days 29–32 post-ICH. Further, brain water content was significantly reduced, neuroinflammation was decreased and hippocampal CA3 neuronal survival was increased, although hemorrhage volume was not affected by CN-105. We concluded, therefore, that pentapeptide CN-105 improved short- and long-term neurobehavioral outcomes in a murine model of ICH, suggesting therapeutic potential for patients with acute ICH.
Collapse
|
20
|
Cao F, Jiang Y, Wu Y, Zhong J, Liu J, Qin X, Chen L, Vitek MP, Li F, Xu L, Sun X. Apolipoprotein E-Mimetic COG1410 Reduces Acute Vasogenic Edema following Traumatic Brain Injury. J Neurotrauma 2016; 33:175-82. [PMID: 26192010 PMCID: PMC4722604 DOI: 10.1089/neu.2015.3887] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The degree of post-traumatic brain edema and dysfunction of the blood-brain barrier (BBB) influences the neurofunctional outcome after a traumatic brain injury (TBI). Previous studies have demonstrated that the administration of apolipoprotein E-mimetic peptide COG1410 reduces the brain water content after subarachnoid hemorrhage, intra-cerebral hemorrhage, and focal brain ischemia. However, the effects of COG1410 on vasogenic edema following TBI are not known. The current study evaluated the effects of 1 mg/kg daily COG1410 versus saline administered intravenously after a controlled cortical impact (CCI) injury on BBB dysfunction and vasogenic edema at an acute stage in mice. The results demonstrated that treatment with COG1410 suppressed the activity of matrix metalloproteinase-9, reduced the disruption of the BBB and Evans Blue dye extravasation, reduced the TBI lesion volume and vasogenic edema, and decreased the functional deficits compared with mice treated with vehicle, at an acute stage after CCI. These findings suggest that COG1410 is a promising preclinical therapeutic agent for the treatment of traumatic brain injury.
Collapse
Affiliation(s)
- Fang Cao
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yong Jiang
- Department of Neurosurgery, the Affiliated Hospital of Luzhou Medical College, Luzhou, China
| | - Yue Wu
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jianjun Zhong
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jieshi Liu
- Department of Neurosurgery, the Affiliated Hospital of Luzhou Medical College, Luzhou, China
| | - Xinghu Qin
- Department of Neurosurgery, the Affiliated Hospital of Luzhou Medical College, Luzhou, China
| | - Ligang Chen
- Department of Neurosurgery, the Affiliated Hospital of Luzhou Medical College, Luzhou, China
| | - Michael P. Vitek
- Department of Medicine (Neurology), Duke University Medical Center, Durham, North Carolina
| | - Fengqiao Li
- Cognosci Inc., Research Triangle Park, North Carolina
| | - Lu Xu
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China
| | - Xiaochuan Sun
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
21
|
Pedachenko EG, Biloshytsky VV, Mikhal'sky SA, Gridina NY, Kvitnitskaya-Ryzhova TY. [The effect of gene therapy with the APOE3 Gene on structural and functional manifestations of secondary hippocampal damages in experimental traumatic brain injury]. ZHURNAL VOPROSY NEĬROKHIRURGII IMENI N. N. BURDENKO 2015; 79:21-32. [PMID: 26146041 DOI: 10.17116/neiro201579221-32] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
AIM OF THE STUDY to study the efficiency of gene therapy following traumatic brain injury (TBI) by evaluating the influences of liposomal transfection of the brain tissue by APOE3-containing plasmid vector on the structural and functional manifestations of development of secondary brain injuries after acute experimental TBI in the rats of different age. MATERIAL AND METHODS Severe diffuse TBI in rats was inflicted under overall anesthesia by free load weighing 450 g, falling from a 1.5 m elevation. The mixture of DOTAP liposome and 25 μg of plasmid vector pCMV·SPORT6 with cDNA of APOE3 gene was infused intraventricularly using ALZET osmotic pumps. Combined morphological, electron microscopic, immunohistochemical and morphometric studies of СА1 hippocampal region were conducted in rats at days 5 and 10 following TBI and gene therapy after investigation of motor functions (using composite neurological motor score) and cognitive functions in Morris water maze. RESULTS Significant changes in the morphofunctional state of hippocampus, as well as in the neurological and cognitive functions were shown on the model of severe TBI in the adult and old Wistar rats. Gene therapy, specifically cationic-liposome mediated APOE3 gene transfer to the CNS cells by plasmid vector, decreased a TBI-induced death of neurons and improved qualitative composition of neuronal population, normalized neuron-glial relations, decreased gliosis and microglial activation, axonal damage, myelin destruction and lipofuscin accumulation, all these having age-related peculiarities. After gene therapy observed in the animal brain was a lower intensity of the processes of apoptosis and a decrease of its rate in old animals. The above changes were accompanied with a more fast and expressed regress of neurological and cognitive disturbances typical for TBI. Administration of plasmid vector after TBI resulted in an increase of survival rate of old animals vs. old animals which got no gene therapy. CONCLUSION APOE3 gene therapy has therapeutic potential in the treatment of severe TBI.
Collapse
Affiliation(s)
- E G Pedachenko
- State Institution 'A.P. Romodanov Institute of Neurosurgery' NAMS Ukraine
| | - V V Biloshytsky
- State Institution 'A.P. Romodanov Institute of Neurosurgery' NAMS Ukraine
| | - S A Mikhal'sky
- State Institution 'D.F. Chebotarev Institute of Gerontology' NAMS Ukraine, Kyiv
| | - N Ya Gridina
- State Institution 'A.P. Romodanov Institute of Neurosurgery' NAMS Ukraine
| | | |
Collapse
|
22
|
A behavioral and histological comparison of fluid percussion injury and controlled cortical impact injury to the rat sensorimotor cortex. Behav Brain Res 2015; 294:254-63. [PMID: 26275924 DOI: 10.1016/j.bbr.2015.08.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 08/05/2015] [Accepted: 08/08/2015] [Indexed: 11/20/2022]
Abstract
Our primary goal was to evaluate the behavioral and histological outcome of fluid percussion injury (FPI) and cortical contusion injury (CCI) to the sensorimotor cortex (SMC). The SMC has been used to evaluate neuroplasticity following CCI, but has not been extensively examined with FPI. In both the CCI and FPI models, a mechanical force of 4mm in diameter was applied over the SMC, allowing for a direct comparison to measure the relative rates of histology and recovery of function in these models. Gross behavioral deficits were found on the sensory task (tactile adhesive removal task) and multiple motor assessments (forelimb asymmetry task, forelimb placing task, and rotorod). These sensorimotor deficits occurred in the absence of cognitive deficits in the water maze. The CCI model creates focal damage with a localized injury wheras the FPI model creates a more diffuse injury causing widespread damage. Both behavioral and histological deficits ensued following both models of injury to the SMC. The neuroplastic changes and ease at which damage to this area can be measured behaviorally make this an excellent location to assess traumatic brain injury (TBI) treatments. No injury model can completely mimic the full spectrum of human TBI and any potential treatments should be validated across both focal and diffuse injury models. Both of these injury models to the SMC produce severe and enduring behavioral deficits, which are ideal for evaluating treatment options.
Collapse
|
23
|
Meloni BP, Milani D, Edwards AB, Anderton RS, O'Hare Doig RL, Fitzgerald M, Palmer TN, Knuckey NW. Neuroprotective peptides fused to arginine-rich cell penetrating peptides: Neuroprotective mechanism likely mediated by peptide endocytic properties. Pharmacol Ther 2015; 153:36-54. [PMID: 26048328 DOI: 10.1016/j.pharmthera.2015.06.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 05/29/2015] [Indexed: 12/22/2022]
Abstract
Several recent studies have demonstrated that TAT and other arginine-rich cell penetrating peptides (CPPs) have intrinsic neuroprotective properties in their own right. Examples, we have demonstrated that in addition to TAT, poly-arginine peptides (R8 to R18; containing 8-18 arginine residues) as well as some other arginine-rich peptides are neuroprotective in vitro (in neurons exposed to glutamic acid excitotoxicity and oxygen glucose deprivation) and in the case of R9 in vivo (after permanent middle cerebral artery occlusion in the rat). Based on several lines of evidence, we propose that this neuroprotection is related to the peptide's endocytosis-inducing properties, with peptide charge and arginine residues being critical factors. Specifically, we propose that during peptide endocytosis neuronal cell surface structures such as ion channels and transporters are internalised, thereby reducing calcium influx associated with excitotoxicity and other receptor-mediated neurodamaging signalling pathways. We also hypothesise that a peptide cargo can act synergistically with TAT and other arginine-rich CPPs due to potentiation of the CPPs endocytic traits rather than by the cargo-peptide acting directly on its supposedly intended intracellular target. In this review, we systematically consider a number of studies that have used CPPs to deliver neuroprotective peptides to the central nervous system (CNS) following stroke and other neurological disorders. Consequently, we critically review evidence that supports our hypothesis that neuroprotection is mediated by carrier peptide endocytosis. In conclusion, we believe that there are strong grounds to regard arginine-rich peptides as a new class of neuroprotective molecules for the treatment of a range of neurological disorders.
Collapse
Affiliation(s)
- Bruno P Meloni
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, Australia; Department of Neurosurgery, Sir Charles Gairdner Hospital, QEII Medical Centre, Nedlands, Western Australia, Australia; Western Australian Neuroscience Research Institute, Nedlands, Australia.
| | - Diego Milani
- Western Australian Neuroscience Research Institute, Nedlands, Australia; School of Heath Sciences, The University Notre Dame, Fremantle, Western Australia, Australia
| | - Adam B Edwards
- Western Australian Neuroscience Research Institute, Nedlands, Australia; School of Heath Sciences, The University Notre Dame, Fremantle, Western Australia, Australia
| | - Ryan S Anderton
- Western Australian Neuroscience Research Institute, Nedlands, Australia; School of Heath Sciences, The University Notre Dame, Fremantle, Western Australia, Australia
| | - Ryan L O'Hare Doig
- Experimental and Regenerative Neurosciences, Western Australia, Australia; School of Anatomy, Physiology and Human Biology, The University of Western Australia, Nedlands, Australia; School of Animal Biology, The University of Western Australia, Nedlands, Australia
| | - Melinda Fitzgerald
- Experimental and Regenerative Neurosciences, Western Australia, Australia; School of Anatomy, Physiology and Human Biology, The University of Western Australia, Nedlands, Australia; School of Animal Biology, The University of Western Australia, Nedlands, Australia
| | - T Norman Palmer
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, Australia; Western Australian Neuroscience Research Institute, Nedlands, Australia
| | - Neville W Knuckey
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, Australia; Department of Neurosurgery, Sir Charles Gairdner Hospital, QEII Medical Centre, Nedlands, Western Australia, Australia; Western Australian Neuroscience Research Institute, Nedlands, Australia
| |
Collapse
|
24
|
Peterson TC, Hoane MR, McConomy KS, Farin FM, Bammler TK, MacDonald JW, Kantor ED, Anderson GD. A Combination Therapy of Nicotinamide and Progesterone Improves Functional Recovery following Traumatic Brain Injury. J Neurotrauma 2015; 32:765-79. [PMID: 25313690 DOI: 10.1089/neu.2014.3530] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Neuroprotection, recovery of function, and gene expression were evaluated in an animal model of traumatic brain injury (TBI) after a combination treatment of nicotinamide (NAM) and progesterone (Prog). Animals received a cortical contusion injury over the sensorimotor cortex, and were treated with either Vehicle, NAM, Prog, or a NAM/Prog combination for 72 h and compared with a craniotomy only (Sham) group. Animals were assessed in a battery of behavioral, sensory, and both fine and gross motor tasks, and given histological assessments at 24 h post-injury to determine lesion cavity size, degenerating neurons, and reactive astrocytes. Microarray-based transcriptional profiling was used to determine treatment-specific changes on gene expression. Our results confirm the beneficial effects of treatment with either NAM or Prog, demonstrating significant improvements in recovery of function and a reduction in lesion cavitation, degenerating neurons, and reactive astrocytes 24 h post-injury. The combination treatment of NAM and Prog led to a significant improvement in both neuroprotection at 24 h post-injury and recovery of function in sensorimotor related tasks when compared with individual treatments. The NAM/Prog-treated group was the only treatment group to show a significant reduction of cortical loss 24 h post-injury. The combination appears to affect inflammatory and immune processes, reducing expression of a significant number of genes in both pathways. Further preclinical trials using NAM and Prog as a combination treatment should be conducted to identify the window of opportunity, determine the optimal duration of treatment, and evaluate the combination in other pre-clinical models of TBI.
Collapse
Affiliation(s)
- Todd C Peterson
- 1Department of Psychology, Southern Illinois University, Carbondale, Illinois
| | - Michael R Hoane
- 1Department of Psychology, Southern Illinois University, Carbondale, Illinois
| | - Keith S McConomy
- 1Department of Psychology, Southern Illinois University, Carbondale, Illinois
| | - Fred M Farin
- 2Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - Theo K Bammler
- 2Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - James W MacDonald
- 2Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington
| | - Eric D Kantor
- 3Department of Pharmacy, University of Washington, Seattle, Washington
| | - Gail D Anderson
- 3Department of Pharmacy, University of Washington, Seattle, Washington
| |
Collapse
|
25
|
Tewari A, Mahendru V, Sinha A, Bilotta F. Antioxidants: The new frontier for translational research in cerebroprotection. J Anaesthesiol Clin Pharmacol 2014; 30:160-71. [PMID: 24803750 PMCID: PMC4009632 DOI: 10.4103/0970-9185.130001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
It is important for the anesthesiologist to understand the etiology of free radical damage and how free-radical scavengers attenuate this, so that this knowledge can be applied to diverse neuro-pathological conditions. This review will concentrate on the role of reactive species of oxygen in the pathophysiology of organ dysfunction, specifically sub arachnoid hemorrhage (SAH), traumatic brain injury (TBI) as well as global central nervous system (CNS) hypoxic, ischemic and reperfusion states. We enumerate potential therapeutic modalities that are been currently investigated and of interest for future trials. Antioxidants are perhaps the next frontier of translational research, especially in neuro-anesthesiology.
Collapse
Affiliation(s)
- Anurag Tewari
- Department of Anesthesiology, Dayanand Medical College, Ludhiana, Punjab, India
| | - Vidhi Mahendru
- Department of Anesthesiology, All India Institute of Medical Sciences, New Delhi, India
| | - Ashish Sinha
- Department of Anesthesiology and Perioperative Medicine, Drexel University College of Medicine, Philadelphia, USA
| | - Federico Bilotta
- Department of Anesthesiology, Critical Care and Pain Medicine, “Sapienza” University of Rome, Rome, Italy
| |
Collapse
|
26
|
Wang R, Hong J, Lu M, Neil JE, Vitek MP, Liu X, Warner DS, Li F, Sheng H. ApoE mimetic ameliorates motor deficit and tissue damage in rat spinal cord injury. J Neurosci Res 2014; 92:884-92. [DOI: 10.1002/jnr.23371] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 10/13/2013] [Accepted: 01/17/2014] [Indexed: 12/12/2022]
Affiliation(s)
- Ruihua Wang
- The First Affiliated Hospital of Zhengzhou University; Zhengzhou China
- Department of Anesthesiology, Multidisciplinary Neuroprotection Laboratories; Duke University Medical Center; Durham North Carolina
| | - Jun Hong
- Department of Anesthesiology, Multidisciplinary Neuroprotection Laboratories; Duke University Medical Center; Durham North Carolina
- Tangshan Gongren Hospital; Hebei China
| | - Miaomiao Lu
- Department of Anesthesiology, Multidisciplinary Neuroprotection Laboratories; Duke University Medical Center; Durham North Carolina
- The Second Affiliated Hospital of Zhengzhou University; Zhengzhou China
| | | | | | - Xiaozhi Liu
- Department of Anesthesiology, Multidisciplinary Neuroprotection Laboratories; Duke University Medical Center; Durham North Carolina
- The Fifth Central Hospital of Tianjin; Tianjin China
| | - David S. Warner
- Department of Anesthesiology, Multidisciplinary Neuroprotection Laboratories; Duke University Medical Center; Durham North Carolina
- Department of Surgery (Neurosurgery); Duke University Medical Center; Durham North Carolina
- Department of Neurobiology; Duke University Medical Center; Durham North Carolina
| | - Fengqiao Li
- Cognosci Inc., Research Triangle Park; North Carolina
| | - Huaxin Sheng
- Department of Anesthesiology, Multidisciplinary Neuroprotection Laboratories; Duke University Medical Center; Durham North Carolina
| |
Collapse
|
27
|
Wang H, Anderson LG, Lascola CD, James ML, Venkatraman TN, Bennett ER, Acheson SK, Vitek MP, Laskowitz DT. ApolipoproteinE mimetic peptides improve outcome after focal ischemia. Exp Neurol 2012; 241:67-74. [PMID: 23219883 DOI: 10.1016/j.expneurol.2012.11.027] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 11/24/2012] [Accepted: 11/29/2012] [Indexed: 10/27/2022]
Abstract
Growing clinical evidence implicates isoform-specific effects of apolipoprotein E (apoE) in reducing neuroinflammation and mediating adaptive responses following ischemic and traumatic brain injury. However, the intact apoE holoprotein does not cross the blood-brain barrier and thus has limited therapeutic potential. We have created a small peptide, COG1410 (acetyl-AS-Aib-LRKL-Aib-KRLL-amide), derived from the apoE receptor-binding region. COG1410 retains the anti-inflammatory and neuroprotective biological properties of the intact holoprotein and penetrates the blood-brain barrier. In the current study, we utilized a murine model of transient focal cerebral ischemia and reperfusion to demonstrate that intravenous (IV) administration of COG1410 reduces infarct volume and radiographic progression of infarct, and improves functional outcome as assessed by rotarod when delivered up to 4h after ischemia onset.
Collapse
Affiliation(s)
- Haichen Wang
- Department of Medicine (Neurology), Duke University School of Medicine, Durham, NC 27710, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Jones TA, Liput DJ, Maresh EL, Donlan N, Parikh TJ, Marlowe D, Kozlowski DA. Use-dependent dendritic regrowth is limited after unilateral controlled cortical impact to the forelimb sensorimotor cortex. J Neurotrauma 2012; 29:1455-68. [PMID: 22352953 PMCID: PMC5749646 DOI: 10.1089/neu.2011.2207] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Compensatory neural plasticity occurs in both hemispheres following unilateral cortical damage incurred by seizures, stroke, and focal lesions. Plasticity is thought to play a role in recovery of function, and is important for the utility of rehabilitation strategies. Such effects have not been well described in models of traumatic brain injury (TBI). We examined changes in immunoreactivity for neural structural and plasticity-relevant proteins in the area surrounding a controlled cortical impact (CCI) to the forelimb sensorimotor cortex (FL-SMC), and in the contralateral homotopic cortex over time (3-28 days). CCI resulted in considerable motor deficits in the forelimb contralateral to injury, and increased reliance on the ipsilateral forelimb. The density of dendritic processes, visualized with immunostaining for microtubule-associated protein-2 (MAP-2), were bilaterally decreased at all time points. Synaptophysin (SYN) immunoreactivity increased transiently in the injured hemisphere, but this reflected an atypical labeling pattern, and it was unchanged in the contralateral hemisphere compared to uninjured controls. The lack of compensatory neuronal structural plasticity in the contralateral homotopic cortex, despite behavioral asymmetries, is in contrast to previous findings in stroke models. In the cortex surrounding the injury (but not the contralateral cortex), decreases in dendrites were accompanied by neurodegeneration, as indicated by Fluoro-Jade B (FJB) staining, and increased expression of the growth-inhibitory protein Nogo-A. These studies indicate that, following unilateral CCI, the cortex undergoes neuronal structural degradation in both hemispheres out to 28 days post-injury, which may be indicative of compromised compensatory plasticity. This is likely to be an important consideration in designing therapeutic strategies aimed at enhancing plasticity following TBI.
Collapse
Affiliation(s)
- Theresa A. Jones
- University of Texas at Austin, Department of Psychology and Institute for Neuroscience, Austin, Texas
| | - Daniel J. Liput
- DePaul University, Department of Biological Sciences, Chicago, Illinois
| | - Erin L. Maresh
- University of Texas at Austin, Department of Psychology and Institute for Neuroscience, Austin, Texas
| | - Nicole Donlan
- University of Texas at Austin, Department of Psychology and Institute for Neuroscience, Austin, Texas
| | - Toral J. Parikh
- University of Texas at Austin, Department of Psychology and Institute for Neuroscience, Austin, Texas
| | - Dana Marlowe
- DePaul University, Department of Biological Sciences, Chicago, Illinois
| | | |
Collapse
|
29
|
Sharifov OF, Nayyar G, Garber DW, Handattu SP, Mishra VK, Goldberg D, Anantharamaiah GM, Gupta H. Apolipoprotein E mimetics and cholesterol-lowering properties. Am J Cardiovasc Drugs 2012; 11:371-81. [PMID: 22149316 DOI: 10.2165/11594190-000000000-00000] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Apolipoprotein E (apoE) is a ligand for clearance of lipoprotein remnants such as chylomicrons and very low-density lipoproteins. It has anti-atherogenic and anti-inflammatory properties. Therefore, there is extensive ongoing research to create peptides that can mimic properties of apoE. A number of synthetic peptides that encompass different regions of apoE have been studied for inhibiting inflammatory states, including Alzheimer disease. However, peptides that clear atherogenic lipoproteins, analogous to apoE, via enhanced hepatic uptake have not been previously reviewed. Toward this end, we describe the design and studies of a dual-domain apoE mimetic peptide, Ac-hE18A-NH(2). This peptide consists of residues 141-150, the putative receptor-binding region of human apoE, covalently linked to a well characterized class A amphipathic helix, 18A, which has no sequence homology to any other exchangeable apolipoprotein sequences. It demonstrates dramatic effects in reducing plasma cholesterol levels in dyslipidemic mouse and rabbit models. We discuss the scientific rationale and review the literature for the design and efficacy of the peptide. Analogous to apoE, this peptide bypasses the low-density lipoprotein receptor for the hepatic uptake of atherogenic lipoproteins via heparan sulfate proteoglycan (HSPG). ApoE mimetics such as Ac-hE18A-NH(2) may therefore restore or replace ligands in genetically induced hyperlipidemias to enable reduction in atherogenic lipoproteins via HSPG even in the absence of functional low-density lipoprotein receptors. Therefore, this and similar peptides may be useful in the treatment of dyslipidemic disorders such as familial hyperlipidemia and atherosclerosis.
Collapse
Affiliation(s)
- Oleg F Sharifov
- Departments of Medicine, Biochemistry and Molecular Genetics and the Atherosclerosis Research Unit, University of Alabama at Birmingham, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
An increase in oxidative stress and overproduction of oxidizing reactive species plays an important role in the pathophysiology of several conditions encountered in the neurocritical care setting including: ischemic and hemorrhagic strokes, traumatic brain injury, acute respiratory distress syndrome, sepsis, and organ failure. The presence of oxidative stress in these conditions is supported by a large body of pre-clinical and clinical studies, and provides a rationale to support a potential therapeutic role for antioxidants. The purpose of this article is to briefly review the basic mechanisms and molecular biology of oxidative stress, summarize its role in critically ill neurological patients, and review available data regarding the potential role of antioxidant strategies in neurocritical care and future directions.
Collapse
Affiliation(s)
- Khalid A. Hanafy
- Department of Neurology, Divisions of Neurocritical Care, Beth Israel Deaconess Medical Center, Boston, MA 02215 USA
| | - Magdy H. Selim
- Department of Neurology, Stroke Division, Beth Israel Deaconess Medical Center, 330 Brookline Avenue – Palmer 127, Boston, MA 02215 USA
| |
Collapse
|
31
|
Vonder Haar C, Anderson GD, Hoane MR. Continuous nicotinamide administration improves behavioral recovery and reduces lesion size following bilateral frontal controlled cortical impact injury. Behav Brain Res 2011; 224:311-7. [PMID: 21704653 PMCID: PMC3159802 DOI: 10.1016/j.bbr.2011.06.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Revised: 05/16/2011] [Accepted: 06/09/2011] [Indexed: 10/18/2022]
Abstract
Previous research has demonstrated considerable preclinical efficacy of nicotinamide (NAM; vitamin B(3)) in animal models of TBI with systemic dosing at 50 and 500 mg/kg yielding improvements on sensory, motor, cognitive and histological measures. The current study aimed to utilize a more specific dosing paradigm in a clinically relevant delivery mechanism: continuously secreting subcutaneous pumps. A bilateral frontal controlled cortical impact (CCI) or sham surgery was performed and rats were treated with NAM (150 mg/kg day) or saline (1 ml/kg) pumps 30 min after CCI, continuing until seven days post-CCI. Rats were given a loading dose of NAM (50mg/kg) or saline (1 ml/kg) following pump implant. Rats received behavioral testing (bilateral tactile adhesive removal, locomotor placing task and Morris water maze) starting on day two post-CCI and were sacrificed at 31 days post-CCI and brains were stained to examine lesion size. NAM-treated rats had reductions in sensory, motor and cognitive behavioral deficits compared to vehicle-treated rats. Specifically, NAM-treated rats significantly improved on the bilateral tactile adhesive removal task, locomotor placing task and the reference memory paradigm of the Morris water maze. Lesion size was also significantly reduced in the NAM-treated group. The results from this study indicate that at the current dose, NAM produces beneficial effects on recovery from a bilateral frontal brain injury and that it may be a relevant compound to be explored in human studies.
Collapse
Affiliation(s)
- Cole Vonder Haar
- Restorative Neuroscience Laboratory, Center for Integrative Research in Cognitive and Neural Sciences, Department of Psychology, Life Science II, MC 6502, Southern Illinois University, Carbondale, IL 62901, USA
| | | | | |
Collapse
|
32
|
SET oncoprotein overexpression in B-cell chronic lymphocytic leukemia and non-Hodgkin lymphoma: a predictor of aggressive disease and a new treatment target. Blood 2011; 118:4150-8. [PMID: 21844565 DOI: 10.1182/blood-2011-04-351072] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
B-cell chronic lymphocytic leukemia (CLL), an incurable leukemia, is characterized by defective apoptosis. We found that the SET oncoprotein, a potent inhibitor of the protein phosphatase 2A (PP2A) tumor suppressor, is overexpressed in primary CLL cells and B-cell non-Hodgkin lymphoma (NHL) cell line cells. In CLL, increased levels of SET correlated significantly with disease severity (shorter time to treatment and overall survival). We developed SET antagonist peptides that bound SET, increased cellular PP2A activity, decreased Mcl-1 expression, and displayed selective cytotoxicity for CLL and NHL cells in vitro. In addition, shRNA for SET was cytotoxic for NHL cells in vitro. The SET antagonist peptide COG449 inhibited growth of NHL tumor xenografts in mice. These data demonstrate that SET is a new treatment target in B-cell malignancies and that SET antagonists represent novel agents for treatment of CLL and NHL.
Collapse
|
33
|
Swan AA, Chandrashekar R, Beare J, Hoane MR. Preclinical efficacy testing in middle-aged rats: nicotinamide, a novel neuroprotectant, demonstrates diminished preclinical efficacy after controlled cortical impact. J Neurotrauma 2011; 28:431-40. [PMID: 21083416 PMCID: PMC3057203 DOI: 10.1089/neu.2010.1519] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Age is a consistent predictor of poor outcome following traumatic brain injury (TBI). Although the elderly population has one of the highest rates of TBI-related hospitalization and death, few preclinical studies have attempted to model and treat TBI in the aged population. Recent studies have indicated that nicotinamide (NAM), a soluble B-group vitamin, improved functional recovery in experimental models of TBI in young animals. The purpose of the present study was to examine the preclinical efficacy of NAM in middle-aged rats. Groups of middle-aged (14-month-old) rats were assigned to NAM (500 mg/kg or 50 mg/kg) or saline alone (1 mL/kg) treatment conditions, and received unilateral cortical contusion injuries (CCI) and injections at 1 h and 24 h following injury. The animals were tested on a variety of tasks to assess vestibulomotor (tapered beam) and cognitive performance (reference and working memory in the Morris water maze), and were evaluated for lesion size, blood-brain barrier compromise, astrocytic activation, and edema formation. In summary, the preclinical efficacy of NAM as a treatment following CCI in middle-aged rats differs from that previously documented in younger rats; while treatment with 50 mg/kg NAM appeared to have no effect, the 500-mg/kg dose worsened performance in middle-aged animals. Histological indicators demonstrated more nuanced group differences, indicating that NAM may positively impact some of the cellular cascades following injury, but were not substantial enough to improve functional recovery. These findings emphasize the need to examine potential treatments for TBI utilizing non-standard populations, and may explain why so many treatments have failed in clinical trials.
Collapse
Affiliation(s)
- Alicia A Swan
- Restorative Neuroscience Laboratory, Center for Integrative Research for Cognitive and Neural Sciences, Department of Psychology, Southern Illinois University , Carbondale, Illinois 62901, USA
| | | | | | | |
Collapse
|
34
|
Kaufman NA, Beare JE, Tan AA, Vitek MP, McKenna SE, Hoane MR. COG1410, an apolipoprotein E-based peptide, improves cognitive performance and reduces cortical loss following moderate fluid percussion injury in the rat. Behav Brain Res 2010; 214:395-401. [PMID: 20600347 PMCID: PMC2936242 DOI: 10.1016/j.bbr.2010.06.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2010] [Revised: 05/19/2010] [Accepted: 06/10/2010] [Indexed: 02/06/2023]
Abstract
COG1410, a small, novel ApoE-mimetic peptide derived from the receptor binding region of apolipoprotein E (ApoE), has been classified as anti-inflammatory in nature and improves motor, sensorimotor, and cognitive dysfunction following cortical contusion injury (CCI). In order to further examine COG1410's preclinical efficacy on cognitive recovery, the present study evaluated COG1410 following moderate fluid percussion injury (FPI). Animals were prepared with a moderate, unilateral FPI over the hippocampus. Following FPI, animals received a regimen of five doses of COG1410 or vehicle at 2 and 4h (1.0mg/kg, i.v.) followed by additional doses administered 24, 48, and 72 h (1.0mg/kg, i.p.). Prior to injury, animals were trained for 4 days (4 trials/day) in the Morris water maze (MWM) and then tested for retrograde amnesia on post-FPI day 11 and then on a working memory task on day 18. Testing for motor dysfunction on the tapered balanced beam began on day 2 post-FPI. Administration of this regimen of COG1410 significantly improved retention of memory in the retrograde amnesia test compared to vehicle post-FPI. However, COG1410 did not significantly improve acquisition of working memory in the MWM. Motor dysfunction on the tapered beam post-FPI was improved in the COG1410-treated group compared to vehicle treatment. Cortical lesion analysis revealed that the COG1410-treated animals demonstrated significantly less tissue loss compared to vehicle-treated animals. The results of this study suggest that COG1410 significantly limited the behavioral dysfunction and tissue loss associated with FPI and demonstrated continued preclinical efficacy for TBI.
Collapse
Affiliation(s)
- Nicholas A Kaufman
- Restorative Neuroscience Laboratory, Center for Integrative Research in Cognitive and Neural Sciences, Department of Psychology, Southern Illinois University, Carbondale, IL, USA
| | | | | | | | | | | |
Collapse
|
35
|
Laskowitz DT, Song P, Wang H, Mace B, Sullivan PM, Vitek MP, Dawson HN. Traumatic Brain Injury Exacerbates Neurodegenerative Pathology: Improvement with an Apolipoprotein E-Based Therapeutic. J Neurotrauma 2010; 27:1983-95. [DOI: 10.1089/neu.2010.1396] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Daniel T. Laskowitz
- Department of Medicine (Neurology), Duke University Medical Center, Durham, North Carolina
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina
| | - Pingping Song
- Department of Medicine (Neurology), Duke University Medical Center, Durham, North Carolina
| | - Haichen Wang
- Department of Medicine (Neurology), Duke University Medical Center, Durham, North Carolina
| | - Brian Mace
- Department of Medicine (Geriatrics), Duke University Medical Center, Durham, North Carolina
| | - Patrick M. Sullivan
- Department of Medicine (Geriatrics), Duke University Medical Center, Durham, North Carolina
| | - Michael P. Vitek
- Department of Medicine (Neurology), Duke University Medical Center, Durham, North Carolina
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina
- Cognosci, Inc., Research Triangle Park, North Carolina
| | - Hana N. Dawson
- Department of Medicine (Neurology), Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
36
|
Kuypers NJ, Hoane MR. Pyridoxine administration improves behavioral and anatomical outcome after unilateral contusion injury in the rat. J Neurotrauma 2010; 27:1275-82. [PMID: 20486803 PMCID: PMC2942865 DOI: 10.1089/neu.2010.1327] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The purpose of this project was to evaluate the preclinical efficacy of pyridoxine, or vitamin B(6). Rats received a 3.0 mm unilateral controlled cortical impact (CCI) injury of the sensorimotor cortex or sham surgery. Treatment with vitamin B(6) (600 or 300 mg/kg IP) or vehicle was administered at 30 min and 24 h post-CCI. Somatosensory dysfunction was evaluated with the vibrissae-forelimb placing and bilateral tactile adhesive removal tests. Sensorimotor dysfunction was evaluated with the locomotor placing and the forelimb asymmetry tests. On the forelimb asymmetry test both treatment groups displayed no asymmetry bias on any of the testing days post-CCI and were statistically no different than the shams. Both vitamin B(6) groups displayed a significant improvement in behavioral performance on the locomotor placing test compared to the vehicle-treated group. Administration of 600 mg/kg also significantly reduced tactile adhesive removal latencies on days 2, 4, 6, and 12 post-CCI. Both treatment groups were improved in their rate of recovery post-CCI on the vibrissae-forelimb placing test, but only the recovery seen in the 600-mg/kg group was significantly improved compared to vehicle. Finally, the 600-mg/kg dose resulted in significant cortical sparing compared to the vehicle-treated group. In general, the effects of vitamin B(6) on recovery of function were dose-dependent, with the 600-mg/kg dose consistently showing greater recovery than the 300-mg/kg dose. More experimental analyses are warranted to evaluate the potential preclinical efficacy and mechanistic action of vitamin B(6).
Collapse
Affiliation(s)
- Nicholas J Kuypers
- Restorative Neuroscience Laboratory, Center for Integrative Research in Cognitive and Neural Sciences, Department of Psychology, Southern Illinois University, Carbondale, Illinois 62901, USA
| | | |
Collapse
|
37
|
Goffus AM, Anderson GD, Hoane M. Sustained delivery of nicotinamide limits cortical injury and improves functional recovery following traumatic brain injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2010; 3:145-52. [PMID: 20716938 PMCID: PMC2952098 DOI: 10.4161/oxim.3.2.11315] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Revised: 01/26/2010] [Accepted: 01/27/2010] [Indexed: 11/19/2022]
Abstract
Previously, we have demonstrated that nicotinamide (NAM), a neuroprotective soluble B-group vitamin, improves recovery of function following traumatic brain injury (TBI). However, no prior studies have examined whether NAM is beneficial following continuous infusions over 7 days post-TBI. The purpose of this study was to investigate the preclinical efficacy of NAM treatment as it might be delivered clinically; over several days by slow infusion. Rats were prepared with either unilateral controlled cortical impact (CCI) injuries or sham procedures and divided into three groups: CCI-NAM, CCI-vehicle, and sham. Thirty minutes following CCI, Alzet osmotic mini-pumps were implanted subcutaneously. NAM was delivered at a rate of 50 mg/kg/day for 7 days immediately post-CCI. On day 7 following injury, the pumps were removed and blood draws were collected for serum NAM and nicotinamide adenine dinucleotide (NAD+) analyses. Starting on day 2 post-CCI, animals were tested on a battery of sensorimotor tests (bilateral tactile adhesive removal, locomotor placing, and limb-use asymmetry). Continuous infusion of NAM resulted in a significant serum elevation in NAM, but not NAD+. Statistical analyses of the tactile removal and locomotor placing data revealed that continuous administration of NAM significantly reduced the initial magnitude of the injury deficit and improved overall recovery compared to the vehicle-treated animals. NAM treatment also significantly decreased limb-use asymmetries compared to vehicle-treated animals. The overall extent of the cortical damage was also reduced by NAM treatment. No detrimental effects were seen following continuous infusion. The present results suggest that NAM delivered via a clinically relevant therapeutic regimen may truncate behavioral damage following TBI. Thus our results offer strong support for translation into the clinical population.
Collapse
Affiliation(s)
- Andrea M Goffus
- Restorative Neuroscience Laboratory, Center for Integrative Research in Cognitive and Neural Sciences, Department of Psychology, Southern Illinois University, Carbondale, IL, USA
| | | | | |
Collapse
|
38
|
Crutcher KA, Lilley HN, Anthony SR, Zhou W, Narayanaswami V. Full-length apolipoprotein E protects against the neurotoxicity of an apoE-related peptide. Brain Res 2009; 1306:106-15. [PMID: 19836363 DOI: 10.1016/j.brainres.2009.10.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2009] [Revised: 10/06/2009] [Accepted: 10/08/2009] [Indexed: 11/26/2022]
Abstract
Apolipoprotein E was found to protect against the neurotoxic effects of a dimeric peptide derived from the receptor-binding region of this protein (residues 141-149). Both apoE3 and apoE4 conferred protection but the major N-terminal fragment of each isoform did not. Nor was significant protection provided by bovine serum albumin or apoA-I. Full-length apoE3 and apoE4 also inhibited the uptake of a fluorescent-labeled derivative of the peptide, suggesting that the mechanism of inhibition might involve competition for cell surface receptors/proteoglycans that mediate endocytosis and/or signaling pathways. These results might bear on the question of the role of apoE in neuronal degeneration, such as occurs in Alzheimer's disease where apoE4 confers a significantly greater risk of pathology.
Collapse
Affiliation(s)
- K A Crutcher
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | | | | | | | | |
Collapse
|
39
|
Tan AA, Quigley A, Smith DC, Hoane MR. Strain differences in response to traumatic brain injury in Long-Evans compared to Sprague-Dawley rats. J Neurotrauma 2009; 26:539-48. [PMID: 19216636 PMCID: PMC2748727 DOI: 10.1089/neu.2008.0611] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The selected strain of rodent used in experimental models of traumatic brain injury is typically dependent upon the experimental questions asked and the familiarity of the investigator with a specific rodent strain. This archival study compares the injury responsiveness and recovery profiles of two popular outbred strains, the Long-Evans (LE) and the Sprague-Dawley (SD), after brain injury induced by lateral fluid percussion injury (LFPI). General findings include a significantly longer duration of unconsciousness in LE rats, but similar durations of apnea. Both strains displayed the same level of initial FPI-induced behavioral deficits, followed by a more rapid rate of functional recovery in SD rats. Cortical volume loss was not significantly different, but close inspection of the data suggests the possibility that LE rats may be more susceptible to damage in the hemisphere contralateral to the injury site than are SD rats. It is hoped that the information provided here encourages greater attention to the subtle differences and similarities between strains in future pre-clinical efficacy studies of traumatic brain injury.
Collapse
Affiliation(s)
- Arlene A Tan
- Restorative Neuroscience Laboratory, Brain and Cognitive Sciences Program, Department of Psychology, Southern Illinois University, Carbondale, Illinois 62901, USA
| | | | | | | |
Collapse
|
40
|
Hoane MR, Kaufman N, Vitek MP, McKenna SE. COG1410 improves cognitive performance and reduces cortical neuronal loss in the traumatically injured brain. J Neurotrauma 2009; 26:121-9. [PMID: 19119914 PMCID: PMC2749004 DOI: 10.1089/neu.2008.0565] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We have previously shown that a single dose of COG1410, a small molecule ApoE-mimetic peptide derived from the apolipoprotein E (ApoE) receptor binding region, improves sensorimotor and motor outcome following cortical contusion injury (CCI). The present study evaluated a regimen of COG1410 following frontal CCI in order to examine its preclinical efficacy on cognitive recovery. Animals were prepared with a bilateral CCI of the frontal cortex. A regimen of COG1410 (0.8mg/kg intravenously [IV]) was administered twice, at 30min and again at 24h post-CCI. Starting on day 11, the animals were tested for their acquisition of a reference memory task in the Morris water maze (MWM), followed by a working memory task in the MWM on day 15. Following CCI, the animals were also tested on the bilateral tactile adhesive removal test to measure sensorimotor dysfunction. On all of the behavioral tests the COG1410 group was no different from the uninjured sham group. Administration of the regimen of COG1410 significantly improved recovery on the reference and working memory tests, as well as on the sensorimotor test. Lesion analysis revealed that COG1410 significantly reduced the size of the injury cavity. Administration of COG1410 also reduced the number of degenerating neurons, as measured by Fluoro-Jade C staining, in the frontal cortex at 48h post-CCI. These results suggest that a regimen of COG1410 appeared to block the development of significant behavioral deficits and reduced tissue loss. These combined findings suggest that COG1410 appears to have strong preclinical efficacy when administered following traumatic brain injury (TBI).
Collapse
Affiliation(s)
- Michael R Hoane
- Restorative Neuroscience Laboratory, Center for Integrative Research in Cognitive and Neural Sciences, Department of Psychology,Southern Illinois University, Carbondale, IL 62901, USA.
| | | | | | | |
Collapse
|
41
|
Hoane MR, Pierce JL, Kaufman NA, Beare JE. Variation in chronic nicotinamide treatment after traumatic brain injury can alter components of functional recovery independent of histological damage. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2008; 1:46-53. [PMID: 19794908 PMCID: PMC2715190 DOI: 10.4161/oxim.1.1.6694] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2008] [Revised: 07/02/2008] [Accepted: 07/10/2008] [Indexed: 11/19/2022]
Abstract
Previously, we have shown that the window of opportunity for nicotinamide (NAM) therapy (50 mg/kg) following cortical contusion injuries (CCI) extended to 4-8 hrs post-CCI when administered over a six day post-CCI interval. The purpose of the present study was to determine if a more chronic NAM treatment protocol administered following CCI would extend the current window of opportunity for effective treatment onset. Groups of rats received either unilateral CCI's or sham procedures. Initiation of NAM therapy (50 mg/kg, ip) began at either 15-min, 4-hrs, 8-hrs or 24-hrs post-injury. All groups received daily systemic treatments for 12 days post-CCI at 24 hr intervals. Behavioral assessments were conducted for 28 days post injury and included: vibrissae forelimb placing, bilateral tactile adhesive removal, forelimb asymmetry task and locomotor placing testing. Behavioral analysis on both the tactile removal and locomotor placing tests showed that all NAM-treated groups facilitated recovery of function compared to saline treatment. However, on the vibrissae-forelimb placing and forelimb asymmetry tests only the 4-hr and 8-hr NAM-treated groups were significantly different from the saline-treated group. The lesion analysis showed that treatment with NAM out to 8 hrs post-CCI significantly reduced the size of the injury cavity. The window of opportunity for NAM treatment is task-dependent and in some situations can extend to 24 hrs post-CCI. These results suggest that a long term treatment regimen of 50 mg/kg of NAM starting at the clinically relevant time points may prove efficacious in human TBI.
Collapse
Affiliation(s)
- Michael R Hoane
- Restorative Neuroscience Laboratory, Department of Psychology, Southern Illinois University, Center for Integrative Research in Cognitive and Neural Sciences, Carbondale, Illinois 62901, USA.
| | | | | | | |
Collapse
|
42
|
Cartagena CM, Ahmed F, Burns MP, Pajoohesh-Ganji A, Pak DT, Faden AI, Rebeck GW. Cortical injury increases cholesterol 24S hydroxylase (Cyp46) levels in the rat brain. J Neurotrauma 2008; 25:1087-98. [PMID: 18729719 PMCID: PMC2745316 DOI: 10.1089/neu.2007.0444] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
In traumatic brain injury (TBI), cellular loss from initial impact as well as secondary neurodegeneration leads to increased cholesterol and lipid debris at the site of injury. Cholesterol accumulation in the periphery can trigger inflammatory mechanisms while cholesterol clearance may be anti-inflammatory. Here we investigated whether TBI altered the regulation of cholesterol 24S-hydroxylase (Cyp46), an enzyme that converts cholesterol to the more hydrophilic 24S-hydroxycholesterol. We examined by Western blot and immunohistochemistry changes in Cyp46 expression following fluid percussion injury. Under normal conditions, most Cyp46 was present in neurons, with very little measurable in glia. Cyp46 levels were significantly increased at 7 days post-injury, and cell type specific analysis at 3 days post-injury showed a significant increase in levels of Cyp46 (84%) in microglia. Since 24-hydroxycholesterol induces activation of genes through the liver X receptor (LXR), we examined protein levels of ATP-binding cassette transporter A1 and apolipoprotein E, two LXR regulated cholesterol homeostasis proteins. Apolipoprotein E and ATP-binding cassette transporter A1 were increased at 7 days post-injury, indicating that increased LXR activity coincided with increased Cyp46 levels. We found that activation of primary rat microglia by LPS in vitro caused increased Cyp46 levels. These data suggest that increased microglial Cyp46 activity is part of a system for removal of damaged cell membranes post-injury, by conversion of cholesterol to 24-hydroxycholesterol and by activation of LXR-regulated gene transcription.
Collapse
Affiliation(s)
| | - Farid Ahmed
- Department of Neuroscience, Georgetown University Medical Center, Washington, D.C
| | - Mark P. Burns
- Department of Neuroscience, Georgetown University Medical Center, Washington, D.C
| | | | - Daniel T. Pak
- Department of Pharmacology, Georgetown University Medical Center, Washington, D.C
| | - Alan I. Faden
- Department of Neuroscience, Georgetown University Medical Center, Washington, D.C
| | - G. William Rebeck
- Department of Neuroscience, Georgetown University Medical Center, Washington, D.C
| |
Collapse
|
43
|
Hoane MR, Pierce JL, Holland MA, Anderson GD. Nicotinamide treatment induces behavioral recovery when administered up to 4 hours following cortical contusion injury in the rat. Neuroscience 2008; 154:861-8. [PMID: 18514428 PMCID: PMC2495083 DOI: 10.1016/j.neuroscience.2008.04.044] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2008] [Revised: 04/21/2008] [Accepted: 04/22/2008] [Indexed: 10/22/2022]
Abstract
Recent studies have demonstrated nicotinamide (NAM), a soluble B-group vitamin, to be an effective treatment in experimental models of traumatic brain injury (TBI). However, research on this compound has been limited to administration regimens starting shortly after injury. This study was conducted to establish the window of opportunity for NAM administration following controlled cortical impact (CCI) injury to the frontal cortex. Groups of rats were assigned to NAM (50 mg/kg), saline (1 ml/kg), or sham conditions and received contusion injuries or sham procedures. Injections of NAM or saline were administered at 15 min, 4 h, or 8 h post-injury, followed by five boosters at 24 h intervals. Following the last injection, blood was taken for serum NAM analysis. Animals were tested on a variety of tasks to assess somatosensory performance (bilateral tactile adhesive removal and vibrissae-forelimb placement) and cognitive performance (reference and working memory) in the Morris water maze. The results of the serum NAM analysis showed that NAM levels were significantly elevated in treated animals. Behavioral analysis on the tactile removal test showed that all NAM-treated groups facilitated recovery of function compared with saline treatment. On the vibrissae-forelimb placing test all NAM-treated groups also were significantly different from the saline-treated group. However, the acquisition of reference memory was only significantly improved in the 15-min and 4-h groups. In the working memory task both the 15-min and 4-h groups also improved working memory compared with saline treatment. The window of opportunity for NAM treatment is task-dependent and extends to 8 h for the sensorimotor tests but only extends to 4 h post-injury in the cognitive tests. These results suggest that a 50 mg/kg treatment regimen starting at the clinically relevant time point of 4 h may result in attenuated injury severity in the human TBI population.
Collapse
Affiliation(s)
- M R Hoane
- Restorative Neuroscience Laboratory, Center for Integrative Research in Cognitive and Neural Sciences, Department of Psychology, Life Science II, MC 6502, Southern Illinois University, Carbondale, IL 62901, USA.
| | | | | | | |
Collapse
|
44
|
Laskowitz DT, Vitek MP. Apolipoprotein E and neurological disease: therapeutic potential and pharmacogenomic interactions. Pharmacogenomics 2008; 8:959-69. [PMID: 17716229 DOI: 10.2217/14622416.8.8.959] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The apolipoprotein E (apoE) polymorphism is emerging as a uniquely important genetic modifier that affects functional outcome from both acute and chronic neurological injuries. Recent attention has focused on common denominator mechanisms by which apoE might affect brain injury and/or brain repair responses in clinically diverse diseases. Although endogenous apoE likely serves several adaptive functions in the injured CNS, there is growing evidence that its effect on modifying brain inflammatory responses and providing protection from excitotoxic injury may be central to its protective properties. A more complete understanding of the role that apoE plays in the injured brain has led to novel therapeutic strategies for both acute and chronic neurological disease.
Collapse
Affiliation(s)
- Daniel T Laskowitz
- Duke University Medical Center, Department of Medicine (Neurology), Box 2900, Durham, NC 27710, USA.
| | | |
Collapse
|