1
|
Sutherland DM, Strebl M, Koehler M, Welsh OL, Yu X, Hu L, dos Santos Natividade R, Knowlton JJ, Taylor GM, Moreno RA, Wörz P, Lonergan ZR, Aravamudhan P, Guzman-Cardozo C, Kour S, Pandey UB, Alsteens D, Wang Z, Prasad BVV, Stehle T, Dermody TS. NgR1 binding to reovirus reveals an unusual bivalent interaction and a new viral attachment protein. Proc Natl Acad Sci U S A 2023; 120:e2219404120. [PMID: 37276413 PMCID: PMC10268256 DOI: 10.1073/pnas.2219404120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 04/19/2023] [Indexed: 06/07/2023] Open
Abstract
Nogo-66 receptor 1 (NgR1) binds a variety of structurally dissimilar ligands in the adult central nervous system to inhibit axon extension. Disruption of ligand binding to NgR1 and subsequent signaling can improve neuron outgrowth, making NgR1 an important therapeutic target for diverse neurological conditions such as spinal crush injuries and Alzheimer's disease. Human NgR1 serves as a receptor for mammalian orthoreovirus (reovirus), but the mechanism of virus-receptor engagement is unknown. To elucidate how NgR1 mediates cell binding and entry of reovirus, we defined the affinity of interaction between virus and receptor, determined the structure of the virus-receptor complex, and identified residues in the receptor required for virus binding and infection. These studies revealed that central NgR1 surfaces form a bridge between two copies of viral capsid protein σ3, establishing that σ3 serves as a receptor ligand for reovirus. This unusual binding interface produces high-avidity interactions between virus and receptor to prime early entry steps. These studies refine models of reovirus cell-attachment and highlight the evolution of viruses to engage multiple receptors using distinct capsid components.
Collapse
Affiliation(s)
- Danica M. Sutherland
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA15224
- Institute of Infection, Inflammation, and Immunity, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, PA15224
| | - Michael Strebl
- Interfaculty Institute of Biochemistry, University of Tübingen, D-72076Tübingen, Germany
| | - Melanie Koehler
- Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain, 1348Louvain-la-Neuve, Belgium
| | - Olivia L. Welsh
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA15224
- Institute of Infection, Inflammation, and Immunity, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, PA15224
| | - Xinzhe Yu
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX77030
| | - Liya Hu
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX77030
| | - Rita dos Santos Natividade
- Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain, 1348Louvain-la-Neuve, Belgium
| | - Jonathan J. Knowlton
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA15224
- Cryo-Electron Microscopy and Tomography Core, Baylor College of Medicine, Houston, TX77030
| | - Gwen M. Taylor
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA15224
- Institute of Infection, Inflammation, and Immunity, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, PA15224
| | - Rodolfo A. Moreno
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX77030
| | - Patrick Wörz
- Interfaculty Institute of Biochemistry, University of Tübingen, D-72076Tübingen, Germany
| | - Zachery R. Lonergan
- Cryo-Electron Microscopy and Tomography Core, Baylor College of Medicine, Houston, TX77030
| | - Pavithra Aravamudhan
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA15224
- Institute of Infection, Inflammation, and Immunity, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, PA15224
| | - Camila Guzman-Cardozo
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA15224
- Institute of Infection, Inflammation, and Immunity, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, PA15224
| | - Sukhleen Kour
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA15224
| | - Udai Bhan Pandey
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA15224
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN37232
- Department of Human Genetics, University of Pittsburgh School of Public Health, Pittsburgh, PA15261
| | - David Alsteens
- Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain, 1348Louvain-la-Neuve, Belgium
- Children’s Neuroscience Institute, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, PA15224
| | - Zhao Wang
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX77030
- Walloon Excellence in Life Sciences and Biotechnology, 1300Wavre, Belgium
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX77030
| | - B. V. Venkataram Prasad
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX77030
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX77030
| | - Thilo Stehle
- Interfaculty Institute of Biochemistry, University of Tübingen, D-72076Tübingen, Germany
| | - Terence S. Dermody
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA15224
- Institute of Infection, Inflammation, and Immunity, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, PA15224
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA15219
| |
Collapse
|
2
|
Soto A, Nieto-Díaz M, Reigada D, Barreda-Manso MA, Muñoz-Galdeano T, Maza RM. miR-182-5p Regulates Nogo-A Expression and Promotes Neurite Outgrowth of Hippocampal Neurons In Vitro. Pharmaceuticals (Basel) 2022; 15:ph15050529. [PMID: 35631355 PMCID: PMC9146179 DOI: 10.3390/ph15050529] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/21/2022] [Accepted: 04/22/2022] [Indexed: 11/25/2022] Open
Abstract
Nogo-A protein is a key myelin-associated inhibitor of axonal growth, regeneration, and plasticity in the central nervous system (CNS). Regulation of the Nogo-A/NgR1 pathway facilitates functional recovery and neural repair after spinal cord trauma and ischemic stroke. MicroRNAs are described as effective tools for the regulation of important processes in the CNS, such as neuronal differentiation, neuritogenesis, and plasticity. Our results show that miR-182-5p mimic specifically downregulates the expression of the luciferase reporter gene fused to the mouse Nogo-A 3′UTR, and Nogo-A protein expression in Neuro-2a and C6 cells. Finally, we observed that when rat primary hippocampal neurons are co-cultured with C6 cells transfected with miR-182-5p mimic, there is a promotion of the outgrowth of neuronal neurites in length. From all these data, we suggest that miR-182-5p may be a potential therapeutic tool for the promotion of axonal regeneration in different diseases of the CNS.
Collapse
Affiliation(s)
| | | | | | | | | | - Rodrigo M. Maza
- Correspondence: (M.N.-D.); (R.M.M.); Tel.: +34-92539-6834 (R.M.M.)
| |
Collapse
|
3
|
Golub VM, Reddy DS. Post-Traumatic Epilepsy and Comorbidities: Advanced Models, Molecular Mechanisms, Biomarkers, and Novel Therapeutic Interventions. Pharmacol Rev 2022; 74:387-438. [PMID: 35302046 PMCID: PMC8973512 DOI: 10.1124/pharmrev.121.000375] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Post-traumatic epilepsy (PTE) is one of the most devastating long-term, network consequences of traumatic brain injury (TBI). There is currently no approved treatment that can prevent onset of spontaneous seizures associated with brain injury, and many cases of PTE are refractory to antiseizure medications. Post-traumatic epileptogenesis is an enduring process by which a normal brain exhibits hypersynchronous excitability after a head injury incident. Understanding the neural networks and molecular pathologies involved in epileptogenesis are key to preventing its development or modifying disease progression. In this article, we describe a critical appraisal of the current state of PTE research with an emphasis on experimental models, molecular mechanisms of post-traumatic epileptogenesis, potential biomarkers, and the burden of PTE-associated comorbidities. The goal of epilepsy research is to identify new therapeutic strategies that can prevent PTE development or interrupt the epileptogenic process and relieve associated neuropsychiatric comorbidities. Therefore, we also describe current preclinical and clinical data on the treatment of PTE sequelae. Differences in injury patterns, latency period, and biomarkers are outlined in the context of animal model validation, pathophysiology, seizure frequency, and behavior. Improving TBI recovery and preventing seizure onset are complex and challenging tasks; however, much progress has been made within this decade demonstrating disease modifying, anti-inflammatory, and neuroprotective strategies, suggesting this goal is pragmatic. Our understanding of PTE is continuously evolving, and improved preclinical models allow for accelerated testing of critically needed novel therapeutic interventions in military and civilian persons at high risk for PTE and its devastating comorbidities.
Collapse
Affiliation(s)
- Victoria M Golub
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| | - Doodipala Samba Reddy
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas
| |
Collapse
|
4
|
Inampudi C, Ciccotosto GD, Cappai R, Crack PJ. Genetic Modulators of Traumatic Brain Injury in Animal Models and the Impact of Sex-Dependent Effects. J Neurotrauma 2021; 37:706-723. [PMID: 32027210 DOI: 10.1089/neu.2019.6955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Traumatic brain injury (TBI) is a major health problem causing disability and death worldwide. There is no effective treatment, due in part to the complexity of the injury pathology and factors affecting its outcome. The extent of brain injury depends on the type of insult, age, sex, lifestyle, genetic risk factors, socioeconomic status, other co-injuries, and underlying health problems. This review discusses the genes that have been directly tested in TBI models, and whether their effects are known to be sex-dependent. Sex differences can affect the incidence, symptom onset, pathology, and clinical outcomes following injury. Adult males are more susceptible at the acute phase and females show greater injury in the chronic phase. TBI is not restricted to a single sex; despite variations in the degree of symptom onset and severity, it is important to consider both female and male animals in TBI pre-clinical research studies.
Collapse
Affiliation(s)
- Chaitanya Inampudi
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia
| | - Giuseppe D Ciccotosto
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia
| | - Roberto Cappai
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia
| | - Peter J Crack
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
5
|
Förstner P, Knöll B. Interference of neuronal activity-mediated gene expression through serum response factor deletion enhances mortality and hyperactivity after traumatic brain injury. FASEB J 2020; 34:3855-3873. [PMID: 31930559 DOI: 10.1096/fj.201902257rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 12/19/2019] [Accepted: 12/23/2019] [Indexed: 12/31/2022]
Abstract
Traumatic brain injury (TBI) is one of the most frequent causes of brain injury and mortality in young adults with detrimental sequelae such as cognitive impairments, epilepsy, and attention-deficit hyperactivity disorder. TBI modulates the neuronal excitability resulting in propagation of a neuronal activity-driven gene expression program. However, the impact of such neuronal activity mediated gene expression in TBI has been poorly studied. In this study we analyzed mouse mutants of the prototypical neuronal activity-dependent transcription factor SRF (serum response factor) in a weight-drop TBI model. Neuron-restricted SRF deletion elevated TBI inflicted mortality suggesting a neuroprotective SRF function during TBI. Behavioral inspection uncovered elevated locomotor activity in Srf mutant mice after TBI in contrast to hypoactivity observed in wild-type littermates. This indicates an SRF role in modulation of TBI-associated alterations in locomotor activity. Finally, induction of a neuronal activity induced gene expression program composed of immediate early genes (IEGs) such as Egr1, Egr2, Egr3, Npas4, Atf3, Arc, Ptgs2, and neuronal pentraxins (Nptx2) was compromised upon SRF depletion. Overall, our data show a role of neuronal activity-mediated gene transcription during TBI and suggest a molecular link between TBI and such post-TBI neurological comorbidities involving hyperactivity phenotypes.
Collapse
Affiliation(s)
- Philip Förstner
- Institute of Physiological Chemistry, Ulm University, Ulm, Germany
| | - Bernd Knöll
- Institute of Physiological Chemistry, Ulm University, Ulm, Germany
| |
Collapse
|
6
|
Lai JH, Karlsson TE, Wu JCC, Huang CZ, Chen YH, Kang SJ, Brodin ATS, Hoffer BJ, Olson L, Chiang YH, Chen KY. Role of Nogo Receptor-1 for Recovery of Balance, Cognition, and Emotion after Mild Traumatic Brain Injury in Mice. J Neurotrauma 2018; 36:1054-1059. [PMID: 30226403 DOI: 10.1089/neu.2018.5949] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Mild traumatic brain injury (mTBI) constitutes 75 ∼ 90% of all TBI cases and causes various physical, cognitive, emotional, and other psychological symptoms. Nogo receptor 1 (NgR1) is a regulator of structural brain plasticity during development and in adulthood. Here, we used mice that, in the absence of doxycycline, overexpress NgR1 in forebrain neurons (MemoFlex) to determine the role of NgR1 in recovery from mTBI with respect to balance, cognition, memory, and emotion. We compared wild-type (WT), MemoFlex, and MemoFlex + doxycycline mice to the same three groups subjected to mTBI. mTBI was induced by a controlled 30-g weight drop. We found that inability to downregulate NgR1 significantly impairs recovery from mTBI-induced impairments. When the NgR1 transgene was turned off, recovery was similar to that of WT mice. The results suggest that the ability to regulate NgR1 signaling is needed for optimal recovery of motor coordination and balance, spatial memory, cognition, and emotional functions after mTBI.
Collapse
Affiliation(s)
- Jing-Huei Lai
- 1 Core Laboratory of Neuroscience, Office of R&D, Taipei Medical University, Taipei, Taiwan.,2 Translational Laboratory, Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan.,3 Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei, Taiwan
| | - Tobias E Karlsson
- 4 Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - John Chung-Che Wu
- 2 Translational Laboratory, Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan.,3 Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei, Taiwan.,5 Department of Surgery, College of Medicine, Taipei Medical University, Taipei, Taiwan.,6 Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Chi-Zong Huang
- 2 Translational Laboratory, Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan.,3 Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei, Taiwan.,6 Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Yen-Hua Chen
- 2 Translational Laboratory, Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan.,3 Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei, Taiwan.,5 Department of Surgery, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shuo-Jhen Kang
- 2 Translational Laboratory, Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan.,3 Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei, Taiwan.,5 Department of Surgery, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Alvin T S Brodin
- 4 Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Barry J Hoffer
- 7 Department of Neurosurgery, Case Western Reserve University, School of Medicine, Cleveland, Ohio
| | - Lars Olson
- 4 Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Yung-Hsiao Chiang
- 1 Core Laboratory of Neuroscience, Office of R&D, Taipei Medical University, Taipei, Taiwan.,2 Translational Laboratory, Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan.,3 Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei, Taiwan.,5 Department of Surgery, College of Medicine, Taipei Medical University, Taipei, Taiwan.,6 Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Kai-Yun Chen
- 2 Translational Laboratory, Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan.,3 Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei, Taiwan.,6 Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
7
|
Genetic variation is associated with RTN4R expression and working memory processing in healthy humans. Brain Res Bull 2017; 134:162-167. [PMID: 28755979 DOI: 10.1016/j.brainresbull.2017.07.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 07/19/2017] [Accepted: 07/23/2017] [Indexed: 02/01/2023]
Abstract
The Nogo receptor (NgR) is implicated in neurodevelopmental processes and it participates in inhibiting axonal growth. Consistent with its high levels of expression in the prefrontal cortex, animal studies indicate that NgR is relevant for prefrontal-related cognitive processing. Given that genetic variation may alter mechanisms of gene expression impacting molecular and systems-level phenotypes, we investigated the association of genetic variation with the expression of the NgR coding gene (RTN4R), as well as with prefrontal correlates at progressively greater biological distance from gene effects. First, we studied the association of single nucleotide polymorphisms (SNPs) with RTN4R mRNA expression in postmortem prefrontal cortex of humans without psychiatric illnesses. Then, we probed in peripheral blood mononuclear cells (PBMCs) the association that we found in prefrontal tissue. Thus, we investigated whether functional genetic variation affecting RTN4R expression is also associated with prefrontal activity during working memory. We found that rs696884 (A/G) predicted these phenotypes. Specifically, the AA genotype was associated with lower RTN4R mRNA expression levels in the prefrontal cortex and PBMCs and inefficient prefrontal activity during working memory compared to the GG genotype. These results suggest that genetic variation associated with RTN4R mRNA expression influences prefrontal physiology in healthy individuals. Furthermore, they highlight the need for further investigations of the role of NgR in the pathophysiology of brain disorders associated with prefrontal dysfunction.
Collapse
|
8
|
Ziebell JM, Ray-Jones H, Lifshitz J. Nogo presence is inversely associated with shifts in cortical microglial morphology following experimental diffuse brain injury. Neuroscience 2017; 359:209-223. [PMID: 28736137 DOI: 10.1016/j.neuroscience.2017.07.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 06/28/2017] [Accepted: 07/12/2017] [Indexed: 01/08/2023]
Abstract
Diffuse traumatic brain injury (TBI) initiates secondary pathology, including inflammation and reduced myelination. Considering these injury-related pathologies, the many states of activated microglia as demonstrated by differing morphologies would form, migrate, and function in and through fields of growth-inhibitory myelin byproduct, specifically Nogo. Here we evaluate the relationship between inflammation and reduced myelin antigenicity in the wake of diffuse TBI and present the hypothesis that the Nogo-66 receptor antagonist peptide NEP(1-40) would reverse the injury-induced shift in distribution of microglia morphologies by limiting myelin-based inhibition. Adult male rats were subjected to midline fluid percussion sham or brain injury. At 2h, 6h, 1d, 2d, 7d, and 21d post-injury, immunohistochemical staining was analyzed in sensory cortex (S1BF) for myelin antigens (myelin basic protein; MBP and CNPase), microglia morphology (ionized calcium-binding adapter protein; Iba1), Nogo receptor and Nogo. Pronounced reduction in myelin antigenicity was evident transiently at 1d post-injury, as evidenced by decreased MBP and CNPase staining, as well as loss of white matter organization, compared to sham and later injury time points. Concomitant with reduced myelin antigenicity, injury shifted microglia morphology from the predominantly ramified morphology observed in sham-injured cortex to hyper-ramified, activated, fully activated, or rod. Changes in microglial morphology were evident as early as 2h post-injury, and remained at least until day 21. Additional cohorts of uninjured and brain-injured animals received vehicle or drug (NEP(1-40), i.p., 15min and 19h post-injury) and brains were collected at 2h, 6h, 1d, 2d, or 7d post-injury. NEP(1-40) administration further shifted distributions of microglia away from an injury-induced activated morphology toward greater proportions of rod and macrophage-like morphologies compared to vehicle-treated. By 7d post-injury, no differences in the distributions of microglia were noted between vehicle and NEP(1-40). This study begins to link secondary pathologies of white matter damage and inflammation after diffuse TBI. In the injured brain, secondary pathologies co-occur and likely interact, with consequences for neuronal circuit disruption leading to neurological symptoms.
Collapse
Affiliation(s)
- Jenna M Ziebell
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA; Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA.
| | - Helen Ray-Jones
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA; Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA; Department of Biology and Biochemistry, University of Bath, Bath, England, UK
| | - Jonathan Lifshitz
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA; Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA; VA Healthcare System, Phoenix, AZ, USA; Psychology, Arizona State University, Tempe, AZ, USA
| |
Collapse
|
9
|
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
|
10
|
Osier ND, Dixon CE. The Controlled Cortical Impact Model: Applications, Considerations for Researchers, and Future Directions. Front Neurol 2016; 7:134. [PMID: 27582726 PMCID: PMC4987613 DOI: 10.3389/fneur.2016.00134] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 08/02/2016] [Indexed: 12/26/2022] Open
Abstract
Controlled cortical impact (CCI) is a mechanical model of traumatic brain injury (TBI) that was developed nearly 30 years ago with the goal of creating a testing platform to determine the biomechanical properties of brain tissue exposed to direct mechanical deformation. Initially used to model TBIs produced by automotive crashes, the CCI model rapidly transformed into a standardized technique to study TBI mechanisms and evaluate therapies. CCI is most commonly produced using a device that rapidly accelerates a rod to impact the surgically exposed cortical dural surface. The tip of the rod can be varied in size and geometry to accommodate scalability to difference species. Typically, the rod is actuated by a pneumatic piston or electromagnetic actuator. With some limits, CCI devices can control the velocity, depth, duration, and site of impact. The CCI model produces morphologic and cerebrovascular injury responses that resemble certain aspects of human TBI. Commonly observed are graded histologic and axonal derangements, disruption of the blood-brain barrier, subdural and intra-parenchymal hematoma, edema, inflammation, and alterations in cerebral blood flow. The CCI model also produces neurobehavioral and cognitive impairments similar to those observed clinically. In contrast to other TBI models, the CCI device induces a significantly pronounced cortical contusion, but is limited in the extent to which it models the diffuse effects of TBI; a related limitation is that not all clinical TBI cases are characterized by a contusion. Another perceived limitation is that a non-clinically relevant craniotomy is performed. Biomechanically, this is irrelevant at the tissue level. However, craniotomies are not atraumatic and the effects of surgery should be controlled by including surgical sham control groups. CCI devices have also been successfully used to impact closed skulls to study mild and repetitive TBI. Future directions for CCI research surround continued refinements to the model through technical improvements in the devices (e.g., minimizing mechanical sources of variation). Like all TBI models, publications should report key injury parameters as outlined in the NIH common data elements (CDEs) for pre-clinical TBI.
Collapse
Affiliation(s)
- Nicole D. Osier
- Department of Acute and Tertiary Care, University of Pittsburgh School of Nursing, Pittsburgh, PA, USA
- Safar Center for Resuscitation Research, Pittsburgh, PA, USA
| | - C. Edward Dixon
- Safar Center for Resuscitation Research, Pittsburgh, PA, USA
- Department of Neurosurgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- VA Pittsburgh Healthcare System, Pittsburgh, PA, USA
| |
Collapse
|
11
|
Osier ND, Carlson SW, DeSana A, Dixon CE. Chronic Histopathological and Behavioral Outcomes of Experimental Traumatic Brain Injury in Adult Male Animals. J Neurotrauma 2015; 32:1861-82. [PMID: 25490251 PMCID: PMC4677114 DOI: 10.1089/neu.2014.3680] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The purpose of this review is to survey the use of experimental animal models for studying the chronic histopathological and behavioral consequences of traumatic brain injury (TBI). The strategies employed to study the long-term consequences of TBI are described, along with a summary of the evidence available to date from common experimental TBI models: fluid percussion injury; controlled cortical impact; blast TBI; and closed-head injury. For each model, evidence is organized according to outcome. Histopathological outcomes included are gross changes in morphology/histology, ventricular enlargement, gray/white matter shrinkage, axonal injury, cerebrovascular histopathology, inflammation, and neurogenesis. Behavioral outcomes included are overall neurological function, motor function, cognitive function, frontal lobe function, and stress-related outcomes. A brief discussion is provided comparing the most common experimental models of TBI and highlighting the utility of each model in understanding specific aspects of TBI pathology. The majority of experimental TBI studies collect data in the acute postinjury period, but few continue into the chronic period. Available evidence from long-term studies suggests that many of the experimental TBI models can lead to progressive changes in histopathology and behavior. The studies described in this review contribute to our understanding of chronic TBI pathology.
Collapse
Affiliation(s)
- Nicole D. Osier
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania
- School of Nursing, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Shaun W. Carlson
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Neurological Surgery, Brain Trauma Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Anthony DeSana
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania
- Seton Hill University, Greensburg, Pennsylvania
| | - C. Edward Dixon
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Neurological Surgery, Brain Trauma Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania
- V.A. Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
| |
Collapse
|
12
|
Wang JW, Yang JF, Ma Y, Hua Z, Guo Y, Gu XL, Zhang YF. Nogo-A expression dynamically varies after spinal cord injury. Neural Regen Res 2015; 10:225-9. [PMID: 25883620 PMCID: PMC4392669 DOI: 10.4103/1673-5374.152375] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2014] [Indexed: 01/22/2023] Open
Abstract
The mechanism involved in neural regeneration after spinal cord injury is unclear. The myelin-derived protein Nogo-A, which is specific to the central nervous system, has been identified to negatively affect the cytoskeleton and growth program of axotomized neurons. Studies have shown that Nogo-A exerts immediate and chronic inhibitory effects on neurite outgrowth. In vivo, inhibitors of Nogo-A have been shown to lead to a marked enhancement of regenerative axon extension. We established a spinal cord injury model in rats using a free-falling weight drop device to subsequently investigate Nogo-A expression. Nogo-A mRNA and protein expression and immunoreactivity were detected in spinal cord tissue using real-time quantitative PCR, immunohistochemistry and western blot analysis. At 24 hours after spinal cord injury, Nogo-A protein and mRNA expression was low in the injured group compared with control and sham-operated groups. The levels then continued to drop further and were at their lowest at 3 days, rapidly rose to a peak after 7 days, and then gradually declined again after 14 days. These changes were observed at both the mRNA and protein level. The transient decrease observed early after injury followed by high levels for a few days indicates Nogo-A expression is time dependent. This may contribute to the lack of regeneration in the central nervous system after spinal cord injury. The dynamic variation of Nogo-A should be taken into account in the treatment of spinal cord injury.
Collapse
Affiliation(s)
- Jian-wei Wang
- Wuxi Hospital of Traditional Chinese Medicine, Institute of Orthopedics and Traumatology of Nanjing University of Chinese Medicine, Wuxi, Jiangsu Province, China
| | - Jun-feng Yang
- Wuxi Hospital of Traditional Chinese Medicine, Institute of Orthopedics and Traumatology of Nanjing University of Chinese Medicine, Wuxi, Jiangsu Province, China
| | - Yong Ma
- Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Zhen Hua
- Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Yang Guo
- Wuxi Hospital of Traditional Chinese Medicine, Institute of Orthopedics and Traumatology of Nanjing University of Chinese Medicine, Wuxi, Jiangsu Province, China
| | - Xiao-lin Gu
- Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Ya-feng Zhang
- Wuxi Hospital of Traditional Chinese Medicine, Institute of Orthopedics and Traumatology of Nanjing University of Chinese Medicine, Wuxi, Jiangsu Province, China
| |
Collapse
|
13
|
Abstract
Three theories of regeneration dominate neuroscience today, all purporting to explain why the adult central nervous system (CNS) cannot regenerate. One theory proposes that Nogo, a molecule expressed by myelin, prevents axonal growth. The second theory emphasizes the role of glial scars. The third theory proposes that chondroitin sulfate proteoglycans (CSPGs) prevent axon growth. Blockade of Nogo, CSPG, and their receptors indeed can stop axon growth in vitro and improve functional recovery in animal spinal cord injury (SCI) models. These therapies also increase sprouting of surviving axons and plasticity. However, many investigators have reported regenerating spinal tracts without eliminating Nogo, glial scar, or CSPG. For example, many motor and sensory axons grow spontaneously in contused spinal cords, crossing gliotic tissue and white matter surrounding the injury site. Sensory axons grow long distances in injured dorsal columns after peripheral nerve lesions. Cell transplants and treatments that increase cAMP and neurotrophins stimulate motor and sensory axons to cross glial scars and to grow long distances in white matter. Genetic studies deleting all members of the Nogo family and even the Nogo receptor do not always improve regeneration in mice. A recent study reported that suppressing the phosphatase and tensin homolog (PTEN) gene promotes prolific corticospinal tract regeneration. These findings cannot be explained by the current theories proposing that Nogo and glial scars prevent regeneration. Spinal axons clearly can and will grow through glial scars and Nogo-expressing tissue under some circumstances. The observation that deleting PTEN allows corticospinal tract regeneration indicates that the PTEN/AKT/mTOR pathway regulates axonal growth. Finally, many other factors stimulate spinal axonal growth, including conditioning lesions, cAMP, glycogen synthetase kinase inhibition, and neurotrophins. To explain these disparate regenerative phenomena, I propose that the spinal cord has evolved regenerative mechanisms that are normally suppressed by multiple extrinsic and intrinsic factors but can be activated by injury, mediated by the PTEN/AKT/mTOR, cAMP, and GSK3b pathways, to stimulate neural growth and proliferation.
Collapse
Affiliation(s)
- Wise Young
- W. M. Keck Center for Collaborative Neuroscience, Rutgers, State University of New Jersey, Piscataway, NJ, USA
| |
Collapse
|
14
|
Abstract
BACKGROUND The handling of experimental animals prior to experimental interventions is often poorly described, even though it may affect the final functional outcome. This study explores how the use of repeated handling of C57BL/6 mice prior to Morris water maze (MWM) tests can affect the performance. METHODS AND MATERIALS The handled animals were subjected to the escalating handling protocol, with the investigator spending 5 min per day per cage for 8 days prior to the MWM test. On the last days of handling, the mice were introduced to water and the concept of a hidden platform. The MWM test consisted of four daily trials for 90 s per day for 4 days with a hidden platform. A probe test was performed 4 days after the last learning trial. Control animals were not handled prior to MWM. RESULTS Handling reduced the latency to find the platform on the first 2 days of the MWM tests and reduced thigmotaxis. The mice increased their swim speed and elicited more explorative behavior in the learning trials and to some lesser extent in the probe trials. CONCLUSIONS The improvement in MWM navigation was most likely due to reduced stress and anxiety regarding the investigator and the test. Handled mice displayed less variability than non-handled mice, suggesting that by using a controlled handling protocol prior to the experiments fewer C57BL/6 mice would be needed to achieve statistically significant differences in studies of learning and spatial memory using MWM.
Collapse
Affiliation(s)
| | - Lars Hillered
- Department of Neuroscience, Section of Neurosurgery, Uppsala University, Uppsala, Sweden
| | - Fredrik Clausen
- Department of Neuroscience, Section of Neurosurgery, Uppsala University, Uppsala, Sweden
| |
Collapse
|
15
|
Gold EM, Su D, López-Velázquez L, Haus DL, Perez H, Lacuesta GA, Anderson AJ, Cummings BJ. Functional assessment of long-term deficits in rodent models of traumatic brain injury. Regen Med 2014; 8:483-516. [PMID: 23826701 DOI: 10.2217/rme.13.41] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Traumatic brain injury (TBI) ranks as the leading cause of mortality and disability in the young population worldwide. The annual US incidence of TBI in the general population is estimated at 1.7 million per year, with an estimated financial burden in excess of US$75 billion a year in the USA alone. Despite the prevalence and cost of TBI to individuals and society, no treatments have passed clinical trial to clinical implementation. The rapid expansion of stem cell research and technology offers an alternative to traditional pharmacological approaches targeting acute neuroprotection. However, preclinical testing of these approaches depends on the selection and characterization of appropriate animal models. In this article we consider the underlying pathophysiology for the focal and diffuse TBI subtypes, discuss the existing preclinical TBI models and functional outcome tasks used for assessment of injury and recovery, identify criteria particular to preclinical animal models of TBI in which stem cell therapies can be tested for safety and efficacy, and review these criteria in the context of the existing TBI literature. We suggest that 2 months post-TBI is the minimum period needed to evaluate human cell transplant efficacy and safety. Comprehensive review of the published TBI literature revealed that only 32% of rodent TBI papers evaluated functional outcome ≥1 month post-TBI, and only 10% evaluated functional outcomes ≥2 months post-TBI. Not all published papers that evaluated functional deficits at a minimum of 2 months post-TBI reported deficits; hence, only 8.6% of overall TBI papers captured in this review demonstrated functional deficits at 2 months or more postinjury. A 2-month survival and assessment period would allow sufficient time for differentiation and integration of human neural stem cells with the host. Critically, while trophic effects might be observed at earlier time points, it will also be important to demonstrate the sustainability of such an effect, supporting the importance of an extended period of in vivo observation. Furthermore, regulatory bodies will likely require at least 6 months survival post-transplantation for assessment of toxicology/safety, particularly in the context of assessing cell abnormalities.
Collapse
Affiliation(s)
- Eric M Gold
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine 2030 Gross Hall, CA 92697-1705, USA
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Zhang YP, Cai J, Shields LBE, Liu N, Xu XM, Shields CB. Traumatic brain injury using mouse models. Transl Stroke Res 2014; 5:454-71. [PMID: 24493632 DOI: 10.1007/s12975-014-0327-0] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Revised: 12/09/2013] [Accepted: 01/05/2014] [Indexed: 12/14/2022]
Abstract
The use of mouse models in traumatic brain injury (TBI) has several advantages compared to other animal models including low cost of breeding, easy maintenance, and innovative technology to create genetically modified strains. Studies using knockout and transgenic mice demonstrating functional gain or loss of molecules provide insight into basic mechanisms of TBI. Mouse models provide powerful tools to screen for putative therapeutic targets in TBI. This article reviews currently available mouse models that replicate several clinical features of TBI such as closed head injuries (CHI), penetrating head injuries, and a combination of both. CHI may be caused by direct trauma creating cerebral concussion or contusion. Sudden acceleration-deceleration injuries of the head without direct trauma may also cause intracranial injury by the transmission of shock waves to the brain. Recapitulation of temporary cavities that are induced by high-velocity penetrating objects in the mouse brain are difficult to produce, but slow brain penetration injuries in mice are reviewed. Synergistic damaging effects on the brain following systemic complications are also described. Advantages and disadvantages of CHI mouse models induced by weight drop, fluid percussion, and controlled cortical impact injuries are compared. Differences in the anatomy, biomechanics, and behavioral evaluations between mice and humans are discussed. Although the use of mouse models for TBI research is promising, further development of these techniques is warranted.
Collapse
Affiliation(s)
- Yi Ping Zhang
- Norton Neuroscience Institute, Norton Healthcare, 210 East Gray Street, Suite 1102, Louisville, KY, 40202, USA,
| | | | | | | | | | | |
Collapse
|
17
|
Omega-3 fatty acids and traumatic neurological injury: from neuroprotection to neuroplasticity? Trends Neurosci 2014; 37:30-8. [DOI: 10.1016/j.tins.2013.10.005] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 10/22/2013] [Accepted: 10/24/2013] [Indexed: 01/21/2023]
|
18
|
Tong J, Liu W, Wang X, Han X, Hyrien O, Samadani U, Smith DH, Huang JH. Inhibition of Nogo-66 receptor 1 enhances recovery of cognitive function after traumatic brain injury in mice. J Neurotrauma 2013; 30:247-58. [PMID: 22967270 DOI: 10.1089/neu.2012.2493] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Central nervous system (CNS) axons recover poorly following injury because of the expression of myelin-derived inhibitors of axonal outgrowth such as Nogo, myelin-associated glycoprotein (MAG), and oligodendrocyte-myelin glycoprotein (OMgp), all of which bind to the Nogo-66 receptor 1 (NgR1). Herein we examine the role of NgR1 in the recovery of motor and cognitive function after traumatic brain injury (TBI) using a controlled cortical impact (CCI) model in NgR1 knockout (KO) and wild-type (WT) mice. Four weeks post-injury, scores on the Novel Object Recognition test were significantly increased in NgR1 KO mice compared with WT mice (p<0.05), but motor behavior test scores did not differ significantly between the two groups. Nissl staining showed that NgR1 KO mice had less brain injury volume 2 weeks after CCI (p<0.05). Histological analysis revealed more doublecortin (DCX+) cells (p<0.01) and more Ki-67+ cells in the contralateral dentate gyrus (DG) (p<0.05) 2 weeks after CCI in NgR1 KO mice than in WT. Furthermore, DCX+ cells still retained their longer processes in KO mice (p<0.01) 4 weeks following trauma. The number of bromodeoxyuridine (BrdU)+ cells did not differ between the two groups at 4 weeks post-trauma, but KO mice had higher numbers of cells that co-stained with NeuN, a marker of mature neurons. Increased transcription of growth-associated protein (GAP)-43 in both the injured and contralateral sides of the hippocampus (both p<0.05) was detected in NgR1 KO mice relative to WT. These data suggest that NgR1 negatively influences plasticity and cognitive recovery after TBI.
Collapse
Affiliation(s)
- Jing Tong
- Department of Neurosurgery, University of Rochester Medical Center, Rochester, New York 14642, USA
| | | | | | | | | | | | | | | |
Collapse
|
19
|
|
20
|
Hånell A, Clausen F, Djupsjö A, Vallstedt A, Patra K, Israelsson C, Larhammar M, Björk M, Paixão S, Kullander K, Marklund N. Functional and Histological Outcome after Focal Traumatic Brain Injury Is Not Improved in Conditional EphA4 Knockout Mice. J Neurotrauma 2012; 29:2660-71. [DOI: 10.1089/neu.2012.2376] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Anders Hånell
- Section for Neurosurgery, Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Fredrik Clausen
- Section for Neurosurgery, Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Anders Djupsjö
- Section for Neurosurgery, Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Anna Vallstedt
- Section for Developmental Genetics, Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Kalicharan Patra
- Section for Developmental Genetics, Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Charlotte Israelsson
- Section for Developmental Neuroscience, Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Martin Larhammar
- Section for Developmental Genetics, Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Maria Björk
- Section for Neurosurgery, Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Sónia Paixão
- Department of Molecular Neurobiology, Max-Planck Institute of Neurobiology, Martinsried, Germany
| | - Klas Kullander
- Section for Developmental Genetics, Department of Neuroscience, Uppsala University, Uppsala, Sweden
| | - Niklas Marklund
- Section for Neurosurgery, Department of Neuroscience, Uppsala University, Uppsala, Sweden
| |
Collapse
|
21
|
Hussain ZM, Fitting S, Watanabe H, Usynin I, Yakovleva T, Knapp PE, Scheff SW, Hauser KF, Bakalkin G. Lateralized response of dynorphin a peptide levels after traumatic brain injury. J Neurotrauma 2012; 29:1785-93. [PMID: 22468884 PMCID: PMC3360894 DOI: 10.1089/neu.2011.2286] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Traumatic brain injury (TBI) induces a cascade of primary and secondary events resulting in impairment of neuronal networks that eventually determines clinical outcome. The dynorphins, endogenous opioid peptides, have been implicated in secondary injury and neurodegeneration in rodent and human brain. To gain insight into the role of dynorphins in the brain's response to trauma, we analyzed short-term (1-day) and long-term (7-day) changes in dynorphin A (Dyn A) levels in the frontal cortex, hippocampus, and striatum, induced by unilateral left-side or right-side cortical TBI in mice. The effects of TBI were significantly different from those of sham surgery (Sham), while the sham surgery also produced noticeable effects. Both sham and TBI induced short-term changes and long-term changes in all three regions. Two types of responses were generally observed. In the hippocampus, Dyn A levels were predominantly altered ipsilateral to the injury. In the striatum and frontal cortex, injury to the right (R) hemisphere affected Dyn A levels to a greater extent than that seen in the left (L) hemisphere. The R-TBI but not L-TBI produced Dyn A changes in the striatum and frontal cortex at 7 days after injury. Effects of the R-side injury were similar in the two hemispheres. In naive animals, Dyn A was symmetrically distributed between the two hemispheres. Thus, trauma may reveal a lateralization in the mechanism mediating the response of Dyn A-expressing neuronal networks in the brain. These networks may differentially mediate effects of left and right brain injury on lateralized brain functions.
Collapse
Affiliation(s)
- Zubair Muhammad Hussain
- The Division of Biological Research on Drug Dependence, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Sylvia Fitting
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
| | - Hiroyuki Watanabe
- The Division of Biological Research on Drug Dependence, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Ivan Usynin
- Institute of Biochemistry, Siberian Division of the Russian Academy of Medical Sciences, Novosibirsk, Russia
| | - Tatjana Yakovleva
- The Division of Biological Research on Drug Dependence, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Pamela E. Knapp
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
- Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, Virginia
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, Virginia
| | - Stephen W. Scheff
- Spinal Cord and Brain Injury Research Center and Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky
| | - Kurt F. Hauser
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
- Institute for Drug and Alcohol Studies, Virginia Commonwealth University, Richmond, Virginia
| | - Georgy Bakalkin
- The Division of Biological Research on Drug Dependence, Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
22
|
Chao PK, Lu KT, Jhu JY, Wo YYP, Huang TC, Ro LS, Yang YL. Indomethacin protects rats from neuronal damage induced by traumatic brain injury and suppresses hippocampal IL-1β release through the inhibition of Nogo-A expression. J Neuroinflammation 2012; 9:121. [PMID: 22676811 PMCID: PMC3416695 DOI: 10.1186/1742-2094-9-121] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Accepted: 03/08/2012] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Nogo-A is a member of the reticulon family of membrane-associated proteins and plays an important role in axonal remodeling. The present study aimed to investigate alterations in Nogo-A expression following traumatic brain injury (TBI)-induced inflammation and neuronal damage. METHODS A weight-drop device was used to deliver a standard traumatic impact to rats. Western blot, RT-PCR and ELISA were used to analyze the expression of Nogo-A and IL-1β. Nogo-A antisense, and an irrelevant control oligonucleotide was intracerebroventricularly infused. We also performed H & E staining and luxol fast blue staining to evaluate the neuronal damage and demyelination resulting from TBI and various treatments. RESULTS Based on RT-PCR and western blot analyses, the expression of Nogo-A was found to be significantly upregulated in the hippocampus beginning eight hours after TBI. In addition, TBI caused an apparent elevation in IL-1β levels and severe neuronal damage and demyelination in the tested animals. All of the TBI-associated molecular and cellular consequences could be effectively reversed by treating the animals with the anti-inflammatory drug indomethacin. More importantly, the TBI-associated stimulation in the levels of both Nogo-A and IL-1β could be effectively inhibited by a specific Nogo-A antisense oligonucleotide. CONCLUSIONS Our findings suggest that the suppression of Nogo-A expression appears to be an early response conferred by indomethacin, which then leads to decreases in the levels of IL-1β and TBI-induced neuron damage.
Collapse
Affiliation(s)
- Po-Kuan Chao
- Department of Life Science, National Taiwan Normal University, 88 Section 4, Ting-Chou Road, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
23
|
Deak F, Sonntag WE. Aging, synaptic dysfunction, and insulin-like growth factor (IGF)-1. J Gerontol A Biol Sci Med Sci 2012; 67:611-25. [PMID: 22503992 PMCID: PMC3348499 DOI: 10.1093/gerona/gls118] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Accepted: 03/16/2012] [Indexed: 01/05/2023] Open
Abstract
Insulin-like growth factor (IGF)-1 is an important neurotrophic hormone. Deficiency of this hormone has been reported to influence the genesis of cognitive impairment and dementia in the elderly patients. Nevertheless, there are studies indicating that cognitive function can be maintained into old age even in the absence of circulating IGF-1 and studies that link IGF-1 to an acceleration of neurological diseases. Although IGF-1 has a complex role in brain function, synaptic effects appear to be central to the IGF-1-induced improvement in learning and memory. In this review, synaptic mechanisms of learning and memory and the effects of IGF-1 on synaptic communication are discussed. The emerging data indicate that synaptic function decreases with age and that IGF-1 contributes to information processing in the brain. Further studies that detail the specific actions of this important neurotrophic hormone will likely lead to therapies that result in improved cognitive function for the elderly patients.
Collapse
Affiliation(s)
- Ferenc Deak
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
| | - William E. Sonntag
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, Oklahoma City, Oklahoma
| |
Collapse
|
24
|
Schoch KM, Madathil SK, Saatman KE. Genetic manipulation of cell death and neuroplasticity pathways in traumatic brain injury. Neurotherapeutics 2012; 9:323-37. [PMID: 22362424 PMCID: PMC3337028 DOI: 10.1007/s13311-012-0107-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Traumatic brain injury (TBI) initiates a complex cascade of secondary neurodegenerative mechanisms contributing to cell dysfunction and necrotic and apoptotic cell death. The injured brain responds by activating endogenous reparative processes to counter the neurodegeneration or remodel the brain to enhance functional recovery. A vast array of genetically altered mice provide a unique opportunity to target single genes or proteins to better understand their role in cell death and endogenous repair after TBI. Among the earliest targets for transgenic and knockout studies in TBI have been programmed cell death mediators, such as the Bcl-2 family of proteins, caspases, and caspase-independent pathways. In addition, the role of cell cycle regulatory elements in the posttraumatic cell death pathway has been explored in mouse models. As interest grows in neuroplasticity in TBI, the use of transgenic and knockout mice in studies focused on gliogenesis, neurogenesis, and the balance of growth-promoting and growth-inhibiting molecules has increased in recent years. With proper consideration of potential effects of constitutive gene alteration, traditional transgenic and knockout models can provide valuable insights into TBI pathobiology. Through increasing sophistication of conditional and cell-type specific genetic manipulations, TBI studies in genetically altered mice will be increasingly useful for identification and validation of novel therapeutic targets.
Collapse
Affiliation(s)
- Kathleen M. Schoch
- Spinal Cord and Brain Injury Research Center and Department of Physiology, University of Kentucky College of Medicine, B473 Biomedical and Biological Sciences Research Building (BBSRB), 741 South Limestone Street, Lexington, KY 40536 USA
| | - Sindhu K. Madathil
- Spinal Cord and Brain Injury Research Center and Department of Physiology, University of Kentucky College of Medicine, B473 Biomedical and Biological Sciences Research Building (BBSRB), 741 South Limestone Street, Lexington, KY 40536 USA
| | - Kathryn E. Saatman
- Spinal Cord and Brain Injury Research Center and Department of Physiology, University of Kentucky College of Medicine, B473 Biomedical and Biological Sciences Research Building (BBSRB), 741 South Limestone Street, Lexington, KY 40536 USA
| |
Collapse
|
25
|
VanGuilder HD, Bixler GV, Sonntag WE, Freeman WM. Hippocampal expression of myelin-associated inhibitors is induced with age-related cognitive decline and correlates with deficits of spatial learning and memory. J Neurochem 2012; 121:77-98. [PMID: 22269040 PMCID: PMC3341628 DOI: 10.1111/j.1471-4159.2012.07671.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Impairment of cognitive functions including hippocampus-dependent spatial learning and memory affects nearly half of the aged population. Age-related cognitive decline is associated with synaptic dysfunction that occurs in the absence of neuronal cell loss, suggesting that impaired neuronal signaling and plasticity may underlie age-related deficits of cognitive function. Expression of myelin-associated inhibitors (MAIs) of synaptic plasticity, including the ligands myelin-associated glycoprotein, neurite outgrowth inhibitor A, and oligodendrocyte myelin glycoprotein, and their common receptor, Nogo-66 receptor, was examined in hippocampal synaptosomes and Cornu ammonis area (CA)1, CA3 and dentate gyrus subregions derived from adult (12-13 months) and aged (26-28 months) Fischer 344 × Brown Norway rats. Rats were behaviorally phenotyped by Morris water maze testing and classified as aged cognitively intact (n = 7-8) or aged cognitively impaired (n = 7-10) relative to adults (n = 5-7). MAI protein expression was induced in cognitively impaired, but not cognitively intact, aged rats and correlated with cognitive performance in individual rats. Immunohistochemical experiments demonstrated that up-regulation of MAIs occurs, in part, in hippocampal neuronal axons and somata. While a number of pathways and processes are altered with brain aging, we report a coordinated induction of myelin-associated inhibitors of functional and structural plasticity only in cognitively impaired aged rats. Induction of MAIs may decrease stimulus-induced synaptic strengthening and structural remodeling, ultimately impairing synaptic mechanisms of spatial learning and memory and resulting in cognitive decline.
Collapse
Affiliation(s)
- Heather D. VanGuilder
- Department of Pharmacology, R130, Hershey Center for Applied Research, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033 USA
| | - Georgina V. Bixler
- Department of Pharmacology, R130, Hershey Center for Applied Research, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033 USA
| | - William E. Sonntag
- Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Science Center, 975 NE 10th Street, BRC-1303, Oklahoma City OK 73104 USA
| | - Willard M. Freeman
- Department of Pharmacology, R130, Hershey Center for Applied Research, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033 USA
| |
Collapse
|
26
|
Hånell A, Hedin J, Clausen F, Marklund N. Facilitated assessment of tissue loss following traumatic brain injury. Front Neurol 2012; 3:29. [PMID: 22435063 PMCID: PMC3303156 DOI: 10.3389/fneur.2012.00029] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 02/16/2012] [Indexed: 11/17/2022] Open
Abstract
All experimental models of traumatic brain injury (TBI) result in a progressive loss of brain tissue. The extent of tissue loss reflects the injury severity and can be measured to evaluate the potential neuroprotective effect of experimental treatments. Quantitation of tissue volumes is commonly performed using evenly spaced brain sections stained using routine histochemical methods and digitally captured. The brain tissue areas are then measured and the corresponding volumes are calculated using the distance between the sections. Measurements of areas are usually performed using a general purpose image analysis software and the results are then transferred to another program for volume calculations. To facilitate the measurement of brain tissue loss we developed novel algorithms which automatically separate the areas of brain tissue from the surrounding image background and identify the ventricles. We implemented these new algorithms by creating a new computer program (SectionToVolume) which also has functions for image organization, image adjustments and volume calculations. We analyzed brain sections from mice subjected to severe focal TBI using both SectionToVolume and ImageJ, a commonly used image analysis program. The volume measurements made by the two programs were highly correlated and analysis using SectionToVolume required considerably less time. The inter-rater reliability was high. Given the extensive use of brain tissue loss measurements in TBI research, SectionToVolume will likely be a useful tool for TBI research. We therefore provide both the source code and the program as attachments to this article.
Collapse
Affiliation(s)
- Anders Hånell
- Neurosurgery, Department of Neuroscience, Uppsala University Uppsala, Sweden
| | | | | | | |
Collapse
|
27
|
Hunt RF, Haselhorst LA, Schoch KM, Bach EC, Rios-Pilier J, Scheff SW, Saatman KE, Smith BN. Posttraumatic mossy fiber sprouting is related to the degree of cortical damage in three mouse strains. Epilepsy Res 2012; 99:167-70. [PMID: 22047981 PMCID: PMC3290720 DOI: 10.1016/j.eplepsyres.2011.10.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Revised: 09/16/2011] [Accepted: 10/09/2011] [Indexed: 11/22/2022]
Abstract
Controlled cortical impact injury was used to examine relationships between focal posttraumatic cortical damage and mossy fiber sprouting (MFS) in the dentate gyrus in three mouse strains. Posttraumatic MFS was more robust when cortical injury impinged upon the hippocampus, versus contusions restricted to neocortex, and was qualitatively similar among CD-1, C57BL/6, and FVB/N background strains. Impact parameters influencing injury severity may be critical in reproducing epilepsy-related changes in neurotrauma models.
Collapse
Affiliation(s)
- Robert F. Hunt
- Department of Physiology, University of Kentucky, Lexington, KY 40536-0298, USA
| | - Laura A. Haselhorst
- Department of Physiology, University of Kentucky, Lexington, KY 40536-0298, USA
| | - Kathleen M. Schoch
- Department of Physiology, University of Kentucky, Lexington, KY 40536-0298, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536-0298, USA
| | - Eva C. Bach
- Department of Physiology, University of Kentucky, Lexington, KY 40536-0298, USA
| | | | - Stephen W. Scheff
- Department of Anatomy and Neurobiology, University of Kentucky, Lexington, KY 40536-0298, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536-0298, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536-0298, USA
| | - Kathryn E. Saatman
- Department of Physiology, University of Kentucky, Lexington, KY 40536-0298, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536-0298, USA
| | - Bret N. Smith
- Department of Physiology, University of Kentucky, Lexington, KY 40536-0298, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536-0298, USA
| |
Collapse
|
28
|
The Nogo-66 receptor family in the intact and diseased CNS. Cell Tissue Res 2012; 349:105-17. [PMID: 22311207 DOI: 10.1007/s00441-012-1332-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Accepted: 01/16/2012] [Indexed: 10/14/2022]
Abstract
The Nogo-66 receptor family (NgR) consists in three glycophosphatidylinositol (GPI)-anchored receptors (NgR1, NgR2 and NgR3), which are primarily expressed by neurons in the central and peripheral mammalian nervous system. NgR1 was identified as serving as a high affinity binding protein for the three classical myelin-associated inhibitors (MAIs) Nogo-A, myelin-associated glycoprotein (MAG) and oligodendrocyte myelin glycoprotein (OMgp), which limit axon regeneration and sprouting in the injured brain. Recent studies suggest that NgR signaling may also play an essential role in the intact adult CNS in restricting axonal and synaptic plasticity and are involved in neurodegenerative diseases, particularly in Alzheimer's disease pathology through modulation of β-secretase cleavage. Here, we outline the biochemical properties of NgRs and their functional roles in the intact and diseased CNS.
Collapse
|
29
|
Chinese Medicine's Intervention Effect on Nogo-A/NgR. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2012; 2012:528482. [PMID: 22216056 PMCID: PMC3247900 DOI: 10.1155/2012/528482] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Revised: 09/28/2011] [Accepted: 11/02/2011] [Indexed: 01/31/2023]
Abstract
Cerebral vascular disease is very common in the elderly and is one of the most dangerous diseases which is hazardous to the body's health, and it is the medical specialists' study hot spot not only in the clinical field but also in the medical basic research field. Neural regeneration has been paid more and more attention in recent years. Nogo's function in the process of neural regeneration has become the focal point since it was discovered in the year 2000. Many studies elucidate that Nogo negatively affects the neural regeneration and plasticity. Chinese medicine plays an important role in the prevention and treatment of neural diseases, and recently some researches about the Chinese medicine's intervention effect on Nogo-A/NgR sprang up, so it is necessary to make a review on this aspect.
Collapse
|
30
|
Bolkvadze T, Pitkänen A. Development of post-traumatic epilepsy after controlled cortical impact and lateral fluid-percussion-induced brain injury in the mouse. J Neurotrauma 2012; 29:789-812. [PMID: 22023672 DOI: 10.1089/neu.2011.1954] [Citation(s) in RCA: 135] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The present study investigated the development of hyperexcitability and epilepsy in mice with traumatic brain injury (TBI) induced by controlled cortical impact (CCI) or lateral fluid-percussion injury (FPI), which are the two most commonly used experimental models of human TBI in rodents. TBI was induced with CCI to 50 (14 controls) and with lateral FPI to 45 (15 controls) C57BL/6S adult male mice. The animals were followed-up for 9 months, including three 2-week periods of continuous video-electroencephalographic (EEG) monitoring, and a seizure susceptibility test with pentylenetetrazol (PTZ). In the end, the animals were perfusion-fixed for histology. The experiment included two independent cohorts of animals. Late post-traumatic spontaneous electrographic seizures were detected in 9% of mice after CCI and 3% after lateral FPI. Eighty-two percent of mice after CCI and 71% after lateral FPI had spontaneous epileptiform spiking on EEG. In addition, 58% of mice with lateral FPI showed spontaneous epileptiform discharges. A PTZ test demonstrated increased seizure susceptibility in the majority of mice in both models, compared to control mice. There was no further progression in the occurrence of epilepsy or epileptiform spiking when follow-up was extended from 6 to 9 months. The severity of cortical or hippocampal damage did not differentiate mice with or without epileptiform activity in either model. Finally, two independent series of experiments in both injury models provided comparable data demonstrating reproducibility of the modeling. These data show that different types of impact can trigger epileptogenesis in mice. Even though the frequency of spontaneous seizures in C57BL/6S mice is low, a large majority of animals develop hyperexcitability.
Collapse
Affiliation(s)
- Tamuna Bolkvadze
- Department of Neurobiology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | | |
Collapse
|
31
|
Marklund N, Hillered L. Animal modelling of traumatic brain injury in preclinical drug development: where do we go from here? Br J Pharmacol 2011; 164:1207-29. [PMID: 21175576 PMCID: PMC3229758 DOI: 10.1111/j.1476-5381.2010.01163.x] [Citation(s) in RCA: 179] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Revised: 12/02/2010] [Accepted: 12/06/2010] [Indexed: 11/26/2022] Open
Abstract
Traumatic brain injury (TBI) is the leading cause of death and disability in young adults. Survivors of TBI frequently suffer from long-term personality changes and deficits in cognitive and motor performance, urgently calling for novel pharmacological treatment options. To date, all clinical trials evaluating neuroprotective compounds have failed in demonstrating clinical efficacy in cohorts of severely injured TBI patients. The purpose of the present review is to describe the utility of animal models of TBI for preclinical evaluation of pharmacological compounds. No single animal model can adequately mimic all aspects of human TBI owing to the heterogeneity of clinical TBI. To successfully develop compounds for clinical TBI, a thorough evaluation in several TBI models and injury severities is crucial. Additionally, brain pharmacokinetics and the time window must be carefully evaluated. Although the search for a single-compound, 'silver bullet' therapy is ongoing, a combination of drugs targeting various aspects of neuroprotection, neuroinflammation and regeneration may be needed. In summary, finding drugs and prove clinical efficacy in TBI is a major challenge ahead for the research community and the drug industry. For a successful translation of basic science knowledge to the clinic to occur we believe that a further refinement of animal models and functional outcome methods is important. In the clinical setting, improved patient classification, more homogenous patient cohorts in clinical trials, standardized treatment strategies, improved central nervous system drug delivery systems and monitoring of target drug levels and drug effects is warranted.
Collapse
Affiliation(s)
- Niklas Marklund
- Department of Neuroscience, Neurosurgery, Uppsala University, Uppsala University Hospital, Uppsala, Sweden.
| | | |
Collapse
|
32
|
Zhou X, Hu X, He W, Tang X, Shi Q, Zhang Z, Yan R. Interaction between amyloid precursor protein and Nogo receptors regulates amyloid deposition. FASEB J 2011; 25:3146-56. [PMID: 21670066 DOI: 10.1096/fj.11-184325] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Excessive production or accumulation of β-amyloid (Aβ) peptides in human brains leads to increased amyloid deposition and cognitive dysfunction, which are invariable pathological features in patients with Alzheimer's disease (AD). Many cellular factors can regulate the production of Aβ. In this study, we show that a family of proteins named Nogo receptor proteins (NgR1 to NgR3) regulates Aβ production via interaction with amyloid precursor protein (APP). Further mapping of the interacting domain indicates that a small region adjacent to the BACE1 cleavage site of APP mediates interaction of APP with Nogo receptor proteins. Our results also indicate that increased interaction between Nogo receptor and APP reduces surface expression of APP and favors processing of APP by BACE1. When NgR2 was ablated in AD transgenic mice expressing Swedish APP and PS1ΔE9, amyloid deposition was clearly reduced (0.66% of total measured area in APP(swe)/PS1ΔE9/NgR2(-/-) mice vs. 0.76% of total measured area in APP(swe)/PS1ΔE9 mice). Our results demonstrate that down-regulation of NgR expression is a potential approach for inhibiting amyloid deposition in AD patients.
Collapse
Affiliation(s)
- Xiangdong Zhou
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| | | | | | | | | | | | | |
Collapse
|
33
|
|