1
|
Wu H, Jiang R, Huang X, Hu X. The Causal Relationship Between Neurotrophic Factors and Delirium: A Mendelian Randomization Study. Brain Behav 2025; 15:e70494. [PMID: 40321107 PMCID: PMC12050958 DOI: 10.1002/brb3.70494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 03/10/2025] [Accepted: 03/31/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Several observational studies have revealed that different neurotrophic factors (NTFs) are associated with delirium, yet the direction and magnitude of the causal association remain poorly understood. Herein, we performed a two-sample Mendelian randomization (MR) analysis to investigate the causal relationship between these factors and delirium. METHODS GWAS data for delirium were sourced from the FINN10 database; GWAS data for risk factors (protein kinase C-binding protein NELL1, neurotrophin-3, neurotrophin-4, brain-derived neurotrophic factor levels, nerve growth factor, ciliary neurotrophic factor, and glial cell-derived neurotrophic factor levels) were from the IEU Open GWAS. Inverse-variance weighted (IVW) was used as a primary analysis. MR-Egger, weighted median (WM), and weighted model were applied to validate the robustness of the results. The MR-Egger regression method was used to explore the presence of horizontal pleiotropy, and the MR pleiotropy residual sum, and outlier (MR-PRESSO) method was applied to detect potential outliers. Cochran's Q test assessed heterogeneity among instrumental variables (IVs). The leave-one-out (LOO) method was used to enhance the precision and veracity of our findings. RESULTS IVW analyses revealed no association between risk factors and delirium. MR Egger, WM, and the weighted mode approach further confirmed these data. MR-Egger regression analysis confirmed the absence of directional pleiotropy in our analysis. Heterogeneity and sensitivity analyses showed reliable results. CONCLUSION No association between other factors and delirium was identified; however, further research is needed to determine if these results apply to other races. Also, advances in molecular biology and epigenetics may shed light on this topic.
Collapse
Affiliation(s)
- Han Wu
- Department of Geriatricsthe Second People's Hospital of LishuiLishuiChina
| | - Ruilai Jiang
- Zhejiang Chinese Medical UniversityHangzhouChina
| | - Xiaocheng Huang
- Department of Geriatricsthe Second People's Hospital of LishuiLishuiChina
| | - Xiaogang Hu
- Department of Geriatricsthe Second People's Hospital of LishuiLishuiChina
| |
Collapse
|
2
|
Gomez-Pinilla F, Myers SK. Traumatic brain injury from a peripheral axis perspective: Uncovering the roles of liver and adipose tissue in temperature regulation. Prog Neurobiol 2025; 247:102733. [PMID: 40032155 DOI: 10.1016/j.pneurobio.2025.102733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/21/2025] [Accepted: 02/18/2025] [Indexed: 03/05/2025]
Abstract
Traumatic brain injury (TBI) is a major cause of death and disability worldwide. Most current treatments for TBI and other neurological disorders focus on the brain, often overlooking the significant contributions of peripheral organs to disease progression. Emerging evidence suggests that organs such as the liver and adipose tissue play crucial roles in TBI pathogenesis. The liver synthesizes lipids and proteins vital for brain function, while adipose tissue provides hormones and metabolites that influence brain activity. New research indicates that the liver and adipose tissue work in concert with the hypothalamus to regulate essential processes, such as body temperature, which become disrupted in TBI. Additionally, the brain-peripheral axis-a complex network of visceral nerve pathways, hormones, and metabolites-plays a bidirectional role in regulating brain plasticity and function. Understanding how TBI leads to dysregulation of the liver, adipose tissue, and other organs could unlock new therapeutic opportunities for treating TBI and related neurological disorders. The intricate autonomic network involving hypothalamic and enteric neurons, along with visceral nerve pathways and hormones, presents both pathological targets and therapeutic potential. We examine scientific evidence suggesting that correcting disturbances in systemic physiology could enhance the brain's capacity for healing. However, the interdependence of this autonomic network implies that treating dysfunction in one area may affect others. Therefore, we also explore the mechanisms by which diet and exercise can comprehensively impact the brain-peripheral axis, supporting the healing process. CHEMICAL COMPOUNDS: D-Fructose (PubChem CID 2723872); docosahexaenoic acid (PubChem CID 45934466); eicosapentaenoic acid (PubChem 5282847).
Collapse
Affiliation(s)
- F Gomez-Pinilla
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095, USA; Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| | - Sydney K Myers
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
3
|
Landucci E, Mango D, Carloni S, Mazzantini C, Pellegrini-Giampietro DE, Saidi A, Balduini W, Schiavi E, Tigli L, Pioselli B, Imbimbo BP, Facchinetti F. Beneficial effects of CHF6467, a modified human nerve growth factor, in experimental neonatal hypoxic-ischaemic encephalopathy. Br J Pharmacol 2025; 182:510-529. [PMID: 39379341 DOI: 10.1111/bph.17353] [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: 03/08/2024] [Revised: 07/03/2024] [Accepted: 07/13/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND AND PURPOSE Therapeutic hypothermia (TH) has become the standard care to reduce morbidity and mortality in neonates affected by moderate-to-severe hypoxic-ischaemic encephalopathy (HIE). Despite the use of TH for HIE, the incidence of mortality and disabilities remains high. EXPERIMENTAL APPROACH Nerve growth factor (NGF) is a potent neurotrophin, but clinical use is limited by its pain eliciting effects. CHF6467 is a recombinant modified form of human NGF devoid of algogenic activity (painless NGF). KEY RESULTS In rodent hippocampal slices exposed to oxygen and glucose deprivation, CHF6467 protected neurons from death and reverted neurotransmission impairment when combined with hypothermia. In a model of rat neonatal HIE, intranasal CHF6467 (20 μg kg-1) significantly reduced brain infarct volume versus vehicle when delivered 10 min or 3 h after the insult. CHF6467 (20 and 40 μg kg-1, i.n.), significantly decreased brain infarct volume to a similar extent to TH and when combined, showed a synergistic neuroprotective effect. CHF6467 (20 μg kg-1, i.n.) per se and in combination with hypothermia reversed locomotor coordination impairment (Rotarod test) and memory deficits (Y-maze and novel object recognition test) in the neonatal HIE rat model. Intranasal administration of CHF6467 resulted in meaningful concentrations in the brain, blunted HIE-induced mRNA elevation of brain neuroinflammatory markers and, when combined to TH, significantly counteracted the increase in plasma levels of neurofilament light chain, a peripheral marker of neuroaxonal damage. CONCLUSION AND IMPLICATIONS CHF6467 administered intranasally is a promising therapy, in combination with TH, for the treatment of HIE.
Collapse
Affiliation(s)
- Elisa Landucci
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Dalila Mango
- Laboratory Pharmacology of Synaptic Plasticity, European Brain Research Institute, Rome, Italy
- School of Pharmacy, University of Rome "Tor Vergata", Rome, Italy
| | - Silvia Carloni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Costanza Mazzantini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | | | - Amira Saidi
- Laboratory Pharmacology of Synaptic Plasticity, European Brain Research Institute, Rome, Italy
| | - Walter Balduini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Elisa Schiavi
- Corporate Preclinical R&D, Chiesi Farmaceutici, Parma, Italy
| | - Laura Tigli
- Corporate Preclinical R&D, Chiesi Farmaceutici, Parma, Italy
| | | | - Bruno P Imbimbo
- Corporate Preclinical R&D, Chiesi Farmaceutici, Parma, Italy
| | | |
Collapse
|
4
|
Elder GA, Gama Sosa MA, De Gasperi R, Perez Garcia G, Perez GM, Abutarboush R, Kawoos U, Zhu CW, Janssen WGM, Stone JR, Hof PR, Cook DG, Ahlers ST. The Neurovascular Unit as a Locus of Injury in Low-Level Blast-Induced Neurotrauma. Int J Mol Sci 2024; 25:1150. [PMID: 38256223 PMCID: PMC10816929 DOI: 10.3390/ijms25021150] [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: 12/12/2023] [Revised: 01/11/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Blast-induced neurotrauma has received much attention over the past decade. Vascular injury occurs early following blast exposure. Indeed, in animal models that approximate human mild traumatic brain injury or subclinical blast exposure, vascular pathology can occur in the presence of a normal neuropil, suggesting that the vasculature is particularly vulnerable. Brain endothelial cells and their supporting glial and neuronal elements constitute a neurovascular unit (NVU). Blast injury disrupts gliovascular and neurovascular connections in addition to damaging endothelial cells, basal laminae, smooth muscle cells, and pericytes as well as causing extracellular matrix reorganization. Perivascular pathology becomes associated with phospho-tau accumulation and chronic perivascular inflammation. Disruption of the NVU should impact activity-dependent regulation of cerebral blood flow, blood-brain barrier permeability, and glymphatic flow. Here, we review work in an animal model of low-level blast injury that we have been studying for over a decade. We review work supporting the NVU as a locus of low-level blast injury. We integrate our findings with those from other laboratories studying similar models that collectively suggest that damage to astrocytes and other perivascular cells as well as chronic immune activation play a role in the persistent neurobehavioral changes that follow blast injury.
Collapse
Affiliation(s)
- Gregory A. Elder
- Neurology Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY 10468, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY 10029, USA;
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY 10029, USA; (M.A.G.S.); (R.D.G.)
- Mount Sinai Alzheimer’s Disease Research Center and the Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (C.W.Z.); (P.R.H.)
| | - Miguel A. Gama Sosa
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY 10029, USA; (M.A.G.S.); (R.D.G.)
- General Medical Research Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY 10468, USA
| | - Rita De Gasperi
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY 10029, USA; (M.A.G.S.); (R.D.G.)
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY 10468, USA;
| | - Georgina Perez Garcia
- Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY 10029, USA;
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY 10468, USA;
| | - Gissel M. Perez
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY 10468, USA;
| | - Rania Abutarboush
- Department of Neurotrauma, Operational and Undersea Medicine Directorate, Naval Medical ResearchCommand, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA; (R.A.); (U.K.); (S.T.A.)
- The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Usmah Kawoos
- Department of Neurotrauma, Operational and Undersea Medicine Directorate, Naval Medical ResearchCommand, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA; (R.A.); (U.K.); (S.T.A.)
- The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
| | - Carolyn W. Zhu
- Mount Sinai Alzheimer’s Disease Research Center and the Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (C.W.Z.); (P.R.H.)
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY 10468, USA;
- Department of Geriatrics and Palliative Care, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - William G. M. Janssen
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - James R. Stone
- Department of Radiology and Medical Imaging, University of Virginia, 480 Ray C Hunt Drive, Charlottesville, VA 22903, USA;
| | - Patrick R. Hof
- Mount Sinai Alzheimer’s Disease Research Center and the Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (C.W.Z.); (P.R.H.)
- Department of Geriatrics and Palliative Care, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - David G. Cook
- Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, 1660 S Columbian Way, Seattle, WA 98108, USA;
- Department of Medicine, University of Washington, 1959 NE Pacific St., Seattle, WA 98195, USA
| | - Stephen T. Ahlers
- Department of Neurotrauma, Operational and Undersea Medicine Directorate, Naval Medical ResearchCommand, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA; (R.A.); (U.K.); (S.T.A.)
| |
Collapse
|
5
|
Yap RS, Kumar J, Teoh SL. Potential Neuroprotective Role of Neurotrophin in Traumatic Brain Injury. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:1189-1202. [PMID: 38279761 DOI: 10.2174/0118715273289222231219094225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/04/2023] [Accepted: 12/12/2023] [Indexed: 01/28/2024]
Abstract
Traumatic brain injury (TBI) is a major global health issue that affects millions of people every year. It is caused by any form of external force, resulting in temporary or permanent impairments in the brain. The pathophysiological process following TBI usually involves excitotoxicity, mitochondrial dysfunction, oxidative stress, inflammation, ischemia, and apoptotic cell death. It is challenging to find treatment for TBI due to its heterogeneous nature, and no therapeutic interventions have been approved thus far. Neurotrophins may represent an alternative approach for TBI treatment because they influence various functional activities in the brain. The present review highlights recent studies on neurotrophins shown to possess neuroprotective roles in TBI. Neurotrophins, specifically brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF) have demonstrated reduced neuronal death, alleviated neuroinflammatory responses and improved neurological functions following TBI via their immunomodulatory, anti-inflammatory and antioxidant properties. Further studies are required to ensure the efficacy and safety of neurotrophins to be used as TBI treatment in clinical settings.
Collapse
Affiliation(s)
- Rei Shian Yap
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Seong Lin Teoh
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
6
|
Mirzahosseini G, Adam JM, Nasoohi S, El-Remessy AB, Ishrat T. Lost in Translation: Neurotrophins Biology and Function in the Neurovascular Unit. Neuroscientist 2023; 29:694-714. [PMID: 35769016 DOI: 10.1177/10738584221104982] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The neurovascular unit (NVU) refers to the functional building unit of the brain and the retina, where neurons, glia, and microvasculature orchestrate to meet the demand of the retina's and brain's function. Neurotrophins (NTs) are structural families of secreted proteins and are known for exerting neurotrophic effects on neuronal differentiation, survival, neurite outgrowth, synaptic formation, and plasticity. NTs include several molecules, such as nerve growth factor, brain-derived neurotrophic factor, NT-3, NT-4, and their precursors. Furthermore, NTs are involved in signaling pathways such as inflammation, apoptosis, and angiogenesis in a nonneuronal cell type. Interestingly, NTs and the precursors can bind and activate the p75 neurotrophin receptor (p75NTR) at low and high affinity. Mature NTs bind their cognate tropomyosin/tyrosine-regulated kinase receptors, crucial for maintenance and neuronal development in the brain and retina axis. Activation of p75NTR results in neuronal apoptosis and cell death, while tropomysin receptor kinase upregulation contributes to differentiation and cell growth. Recent findings indicate that modulation of NTs and their receptors contribute to neurovascular dysfunction in the NVU. Several chronic metabolic and acute ischemic diseases affect the NVU, including diabetic and ischemic retinopathy for the retina, as well as stroke, acute encephalitis, and traumatic brain injury for the brain. This work aims to review the current evidence through published literature studying the impact of NTs and their receptors, including the p75NTR receptor, on the injured and healthy brain-retina axis.
Collapse
Affiliation(s)
- Golnoush Mirzahosseini
- Department of Anatomy and Neurobiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Justin Mark Adam
- Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Sanaz Nasoohi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Tauheed Ishrat
- Department of Anatomy and Neurobiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
- Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
7
|
Gatto A, Capossela L, Conti G, Eftimiadi G, Ferretti S, Manni L, Curatola A, Graglia B, Di Sarno L, Calcagni ML, Di Giuda D, Cecere S, Romeo DM, Soligo M, Picconi E, Piastra M, Della Marca G, Staccioli S, Ruggiero A, Cocciolillo F, Pulitanò S, Chiaretti A. Intranasal human-recombinant NGF administration improves outcome in children with post-traumatic unresponsive wakefulness syndrome. Biol Direct 2023; 18:61. [PMID: 37789391 PMCID: PMC10546699 DOI: 10.1186/s13062-023-00418-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 09/26/2023] [Indexed: 10/05/2023] Open
Abstract
BACKGROUND Severe traumatic brain injury (TBI) is one of the most dramatic events in pediatric age and, despite advanced neuro-intensive care, the survival rate of these patients remains low. Children suffering from severe TBI show long-term sequelae, more pronounced in behavioral, neurological and neuropsychological functions leading to, in the most severe cases, an unresponsive wakefulness syndrome (UWS). Currently, no effective treatments can restore neuronal loss or produce significant improvement in these patients. In experimental animal models, human- recombinant Nerve Growth Factor (hr-NGF) promotes neural recovery supporting neuronal growth, differentiation and survival of brain cells and up-regulating the neurogenesis-associated processes. Only a few studies reported the efficacy of intranasal hr-NGF administration in children with post- traumatic UWS. METHODS Children with the diagnosis of post-traumatic UWS were enrolled. These patients underwent a treatment with intranasal hr-NGF administration, at a total dose of 50 gamma/kg, three times a day for 7 consecutive days. The treatment schedule was performed for 4 cycles, at one month distance each. Neuroradiogical evaluation by Positron Emission Tomography scan (PET), Single Photon Emission Computed Tomography (SPECT), Electroencephalography (EEG), and Power Spectral Density (PSD) was determined before the treatment and one month after the end. Neurological assessment was also deepened by using modified Ashworth Scale, Gross Motor Function Measure, and Disability Rating Scale. RESULTS Three children with post-traumatic UWS were treated. hr-NGF administration improved functional (PET and SPECT) and electrophysiological (EEG and PSD) assessment. Also clinical conditions improved, mainly for the reduction of spasticity and with the acquisition of voluntary movements, facial mimicry, attention and verbal comprehension, ability to cry, cough reflex, oral motility, and feeding capacity, with a significant improvement of their neurological scores. No side effects were reported. CONCLUSION These promising results and the ease of administration of this treatment make it worthwhile to be investigated further, mainly in the early stages from severe TBI and in patients with better baseline neurological conditions, to explore more thoroughly the benefits of this new approach on neuronal function recovery after traumatic brain damage.
Collapse
Affiliation(s)
- Antonio Gatto
- Dipartimento di Pediatria, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Lavinia Capossela
- Dipartimento di Pediatria, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giorgio Conti
- Terapia Intensiva Pediatrica, Dipartimento di Scienze dell'Emergenza, Anestesiologiche e Rianimazione, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Gemma Eftimiadi
- Dipartimento di Pediatria, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Serena Ferretti
- Dipartimento di Pediatria, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Luigi Manni
- Istituto di Farmacologia Traslazionale, Consiglio Nazionale delle Ricerche (CNR), Rome, Italy
| | - Antonietta Curatola
- Dipartimento di Pediatria, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Benedetta Graglia
- Dipartimento di Pediatria, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Lorenzo Di Sarno
- Dipartimento di Pediatria, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Maria Lucia Calcagni
- UOC di Medicina Nucleare, Fondazione Policlinico Universitario "A. Gemelli" IRCCS - Università Cattolica del Sacro Cuore, Rome, Italy
| | - Daniela Di Giuda
- UOC di Medicina Nucleare, Fondazione Policlinico Universitario "A. Gemelli" IRCCS - Università Cattolica del Sacro Cuore, Rome, Italy
| | - Stefano Cecere
- Dipartimento di Pediatria, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Domenico Marco Romeo
- Unità di Neurologia Pediatrica, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Marzia Soligo
- Istituto di Farmacologia Traslazionale, Consiglio Nazionale delle Ricerche (CNR), Rome, Italy
| | - Enzo Picconi
- Terapia Intensiva Pediatrica, Dipartimento di Scienze dell'Emergenza, Anestesiologiche e Rianimazione, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Marco Piastra
- Terapia Intensiva Pediatrica, Dipartimento di Scienze dell'Emergenza, Anestesiologiche e Rianimazione, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Giacomo Della Marca
- Dipartimento di Scienze dell'Invecchiamento, Neurologiche, Ortopediche e della Testa-Collo, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
| | - Susanna Staccioli
- Dipartimento di Neuroriabilitazione Intensiva, Ospedale Pediatrico "Bambino Gesù", Rome, Italy
| | - Antonio Ruggiero
- Oncologia Pediatrica, Fondazione Policlinico Universitario A.Gemelli IRCCS - Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Fabrizio Cocciolillo
- UOC di Medicina Nucleare, Fondazione Policlinico Universitario "A. Gemelli" IRCCS - Università Cattolica del Sacro Cuore, Rome, Italy
| | - Silvia Pulitanò
- Terapia Intensiva Pediatrica, Dipartimento di Scienze dell'Emergenza, Anestesiologiche e Rianimazione, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Antonio Chiaretti
- Dipartimento di Pediatria, Università Cattolica del Sacro Cuore, Rome, Italy.
- Department of Women's Health Sciences, Fondazione Policlinico Universitario A. Gemelli - IRCCS, Largo Agostino Gemelli 8, 00168, Rome, Italy.
| |
Collapse
|
8
|
Manni L, Leotta E, Mollica I, Serafino A, Pignataro A, Salvatori I, Conti G, Chiaretti A, Soligo M. Acute intranasal treatment with nerve growth factor limits the onset of traumatic brain injury in young rats. Br J Pharmacol 2023; 180:1949-1964. [PMID: 36780920 DOI: 10.1111/bph.16056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/15/2023] Open
Abstract
BACKGROUND AND PURPOSE Traumatic brain injury (TBI) comprises a primary injury directly induced by impact, which progresses into a secondary injury leading to neuroinflammation, reactive astrogliosis, and cognitive and motor damage. To date, treatment of TBI consists solely of palliative therapies that do not prevent and/or limit the outcomes of secondary damage and only stabilize the deficits. The neurotrophin, nerve growth factor (NGF), delivered to the brain parenchyma following intranasal application, could be a useful means of limiting or improving the outcomes of the secondary injury, as suggested by pre-clinical and clinical data. EXPERIMENTAL APPROACH We evaluated the effect of acute intranasal treatment of young (20-postnatal day) rats, with NGF in a TBI model (weight drop/close head), aggravated by hypoxic complications. Immediately after the trauma, rats were intranasally treated with human recombinant NGF (50 μg·kg-1 ), and motor behavioural test, morphometric and biochemical assays were carried out 24 h later. KEY RESULTS Acute intranasal NGF prevented the onset of TBI-induced motor disabilities, and decreased reactive astrogliosis, microglial activation and IL-1β content, which after TBI develops to the same extent in the impact zone and the hypothalamus. CONCLUSION AND IMPLICATIONS Intranasal application of NGF was effective in decreasing the motor dysfunction and neuroinflammation in the brain of young rats in our model of TBI. This work forms an initial pre-clinical evaluation of the potential of early intranasal NGF treatment in preventing and limiting the disabling outcomes of TBI, a clinical condition that remains one of the unsolved problems of paediatric neurology.
Collapse
Affiliation(s)
- Luigi Manni
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), Rome, Italy
| | - Eleonora Leotta
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), Rome, Italy
| | - Ilia Mollica
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), Rome, Italy
| | - Annalucia Serafino
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), Rome, Italy
| | - Annabella Pignataro
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), Rome, Italy
- IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Illari Salvatori
- IRCCS Fondazione Santa Lucia, Rome, Italy
- Department of Experimental Medicine, Faculty of Medicine, University of Rome 'La Sapienza', Rome, Italy
| | - Giorgio Conti
- Intensive Pediatric Therapy and Pediatric Trauma Center, Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Antonio Chiaretti
- Institute of Pediatrics, Department of Woman and Child Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Marzia Soligo
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), Rome, Italy
| |
Collapse
|
9
|
Curatola A, Graglia B, Granata G, Conti G, Capossela L, Manni L, Ferretti S, Di Giuda D, Romeo DM, Calcagni ML, Soligo M, Castelli E, Piastra M, Mantelli F, Marca GD, Staccioli S, Romeo T, Pani M, Cocciolillo F, Mancino A, Gatto A, Chiaretti A. Combined treatment of nerve growth factor and transcranical direct current stimulations to improve outcome in children with vegetative state after out-of-hospital cardiac arrest. Biol Direct 2023; 18:24. [PMID: 37165387 PMCID: PMC10170696 DOI: 10.1186/s13062-023-00379-5] [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: 03/22/2023] [Accepted: 05/01/2023] [Indexed: 05/12/2023] Open
Abstract
BACKGROUND Out-of-hospital cardiac arrest (OHCA) is one of the most dramatic events in pediatric age and, despite advanced neurointensive care, the survival rate remains low. Currently, no effective treatments can restore neuronal loss or produce significant improvement in these patients. Nerve Growth Factor (NGF) is a neurotrophin potentially able to counteract many of the deleterious effects triggered by OHCA. Transcranial Direct Current Stimulation (tDCS) has been reported to be neuroprotective in many neurological diseases, such as motor deficit and cognitive impairment. Children with the diagnosis of chronic vegetative state after OHCA were enrolled. These patients underwent a combined treatment of intranasal administration of human recombinant NGF (hr-NGF), at a total dose of 50 gamma/kg, and tDCS, in which current intensity was increased from zero to 2 mA from the first 5 s of stimulation and maintained constant for 20 min. The treatment schedule was performed twice, at one month distance each. Neuroradiogical evaluation with Positron Emission Tomography scan (PET), Single Photon Emission Computed Tomography (SPECT), Electroencephalography (EEG) and Power Spectral Density of the brain (PSD) was determined before the treatment and one month after the end. Neurological assessment was deepened by using modified Ashworth Scale, Gross Motor Function Measure, and Disability Rating Scale. RESULTS Three children with a chronic vegetative state secondary to OHCA were treated. The combined treatment with hr-NGF and tDCS improved functional (PET and SPECT) and electrophysiological (EEG and PSD) assessment. Also clinical conditions improved, mainly for the reduction of spasticity and with the acquisition of voluntary finger movements, improved facial mimicry and reaction to painful stimuli. No side effects were reported. CONCLUSIONS These promising preliminary results and the ease of administration of this treatment make it worthwhile to be investigated further, mainly in the early stages from OHCA and in patients with better baseline neurological conditions, in order to explore more thoroughly the benefits of this new approach on neuronal function recovery after OHCA.
Collapse
Affiliation(s)
- Antonietta Curatola
- Dipartimento di Pediatria, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Benedetta Graglia
- Dipartimento di Pediatria, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giuseppe Granata
- Istituto di Neurologia, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Giorgio Conti
- Dipartimento di Scienze dell'Emergenza, Anestesiologiche e Rianimazione, Terapia Intensiva Pediatrica, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Lavinia Capossela
- Dipartimento di Pediatria, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Luigi Manni
- Istituto di Farmacologia Traslazionale, Consiglio Nazionale delle Ricerche (CNR), Rome, Italy
| | - Serena Ferretti
- Dipartimento di Pediatria, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Daniela Di Giuda
- UOC di Medicina Nucleare, Fondazione Policlinico Universitario "A. Gemelli" IRCCS - Università Cattolica del Sacro Cuore, Rome, Italy
| | - Domenico Marco Romeo
- Unità di Neurologia Pediatrica, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Maria Lucia Calcagni
- UOC di Medicina Nucleare, Fondazione Policlinico Universitario "A. Gemelli" IRCCS - Università Cattolica del Sacro Cuore, Rome, Italy
| | - Marzia Soligo
- Istituto di Farmacologia Traslazionale, Consiglio Nazionale delle Ricerche (CNR), Rome, Italy
| | - Enrico Castelli
- Dipartimento di Neuroriabilitazione Intensiva, Ospedale Pediatrico "Bambino Gesù", Rome, Italy
| | - Marco Piastra
- Dipartimento di Scienze dell'Emergenza, Anestesiologiche e Rianimazione, Terapia Intensiva Pediatrica, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Flavio Mantelli
- Dompé Farmaceutici Spa, Via Campo di Pile, snc, L'Aquila, 67100, Italy
| | - Giacomo Della Marca
- Dipartimento di Scienze dell'Invecchiamento, Neurologiche, Ortopediche e della Testa-Collo, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
| | - Susanna Staccioli
- Dipartimento di Neuroriabilitazione Intensiva, Ospedale Pediatrico "Bambino Gesù", Rome, Italy
| | - Tiziana Romeo
- Dompé Farmaceutici Spa, Via Campo di Pile, snc, L'Aquila, 67100, Italy
| | - Marcello Pani
- Direttore Farmacia Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy
| | - Fabrizio Cocciolillo
- UOC di Medicina Nucleare, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Aldo Mancino
- Dipartimento di Scienze dell'Emergenza, Anestesiologiche e Rianimazione, Terapia Intensiva Pediatrica, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Antonio Gatto
- Dipartimento di Pediatria, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Antonio Chiaretti
- Dipartimento di Pediatria, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy.
- Dipartimento di Pediatria, Università Cattolica del Sacro Cuore, Rome, Italy.
| |
Collapse
|
10
|
Neurotrophic Factors as Regenerative Therapy for Neurodegenerative Diseases: Current Status, Challenges and Future Perspectives. Int J Mol Sci 2023; 24:ijms24043866. [PMID: 36835277 PMCID: PMC9968045 DOI: 10.3390/ijms24043866] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/25/2023] [Accepted: 02/06/2023] [Indexed: 02/17/2023] Open
Abstract
Neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), multiple sclerosis (MS), spinal cord injury (SCI), and amyotrophic lateral sclerosis (ALS), are characterized by acute or chronic progressive loss of one or several neuronal subtypes. However, despite their increasing prevalence, little progress has been made in successfully treating these diseases. Research has recently focused on neurotrophic factors (NTFs) as potential regenerative therapy for neurodegenerative diseases. Here, we discuss the current state of knowledge, challenges, and future perspectives of NTFs with a direct regenerative effect in chronic inflammatory and degenerative disorders. Various systems for delivery of NTFs, such as stem and immune cells, viral vectors, and biomaterials, have been applied to deliver exogenous NTFs to the central nervous system, with promising results. The challenges that currently need to be overcome include the amount of NTFs delivered, the invasiveness of the delivery route, the blood-brain barrier permeability, and the occurrence of side effects. Nevertheless, it is important to continue research and develop standards for clinical applications. In addition to the use of single NTFs, the complexity of chronic inflammatory and degenerative diseases may require combination therapies targeting multiple pathways or other possibilities using smaller molecules, such as NTF mimetics, for effective treatment.
Collapse
|
11
|
D'Mello V, Subramaniam M, Bhalla AP, Saavedra S, Leiba O, Levison SW. Intranasal Leukemia Inhibitory Factor Attenuates Gliosis and Axonal Injury and Improves Sensorimotor Function After a Mild Pediatric Traumatic Brain Injury. Neurotrauma Rep 2023; 4:236-250. [PMID: 37095853 PMCID: PMC10122240 DOI: 10.1089/neur.2021.0075] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023] Open
Abstract
Leukemia inhibitory factor (LIF) is a neuroprotective cytokine that is essential for appropriate glial responses, remyelination, and preservation of neuronal conductance after injury. The intranasal route for delivery of therapeutics to the central nervous system is of particular interest given that it bypasses the blood-brain barrier and peripheral clearance systems. We explored the possibility that LIF might improve neurological function when administered intranasally during the acute phase in a pediatric model of mild traumatic brain injury (mTBI). We tested two doses of LIF and evaluated behavioral outcomes. Here, we show that acute 40-ng intranasal LIF treatment twice a day for 3 days attenuates astrogliosis and microgliosis, protects against axonal damage, significantly improves sensorimotor function, and is well tolerated without detrimental effects on growth. Altogether, our studies provide pre-clinical evidence for the use of acute intranasal LIF treatment as a viable therapeutic for pediatric cases of mTBIs.
Collapse
Affiliation(s)
- Veera D'Mello
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Malini Subramaniam
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Aditya Paul Bhalla
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Sherlyn Saavedra
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Ofri Leiba
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Steven W. Levison
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
- Address correspondence to: Steven W. Levison, PhD, Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers University, 205 South Orange Avenue, Newark, NJ 07103, USA.
| |
Collapse
|
12
|
Soligo M, Manni L, Conti G, Chiaretti A. Intranasal nerve growth factor for prevention and recovery of the outcomes of traumatic brain injury. Neural Regen Res 2023; 18:773-778. [DOI: 10.4103/1673-5374.354513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
13
|
Amyloidogenesis and Neurotrophic Dysfunction in Alzheimer’s Disease: Do They have a Common Regulating Pathway? Cells 2022; 11:cells11203201. [PMID: 36291068 PMCID: PMC9600014 DOI: 10.3390/cells11203201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/07/2022] [Accepted: 10/09/2022] [Indexed: 11/17/2022] Open
Abstract
The amyloid cascade hypothesis has predominately been used to describe the pathogenesis of Alzheimer’s disease (AD) for decades, as Aβ oligomers are thought to be the prime cause of AD. Meanwhile, the neurotrophic factor hypothesis has also been proposed for decades. Accumulating evidence states that the amyloidogenic process and neurotrophic dysfunction are mutually influenced and may coincidently cause the onset and progress of AD. Meanwhile, there are intracellular regulators participating both in the amyloidogenic process and neurotrophic pathways, which might be the common original causes of amyloidogenesis and neurotrophic dysfunction. In this review, the current understanding regarding the role of neurotrophic dysfunction and the amyloidogenic process in AD pathology is briefly summarized. The mutual influence of these two pathogenesis pathways and their potential common causal pathway are further discussed. Therapeutic strategies targeting the common pathways to simultaneously prevent amyloidogenesis and neurotrophic dysfunction might be anticipated for the disease-modifying treatment of AD.
Collapse
|
14
|
Humpel C. Intranasal neprilysin rapidly eliminates amyloid-beta plaques, but causes plaque compensations: the explanation why the amyloid-beta cascade may fail? Neural Regen Res 2022; 17:1881-1884. [PMID: 35142662 PMCID: PMC8848595 DOI: 10.4103/1673-5374.335138] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/03/2021] [Accepted: 08/17/2021] [Indexed: 11/04/2022] Open
Abstract
Neurodegenerative brain disorders are a major burden in our society, such as Alzheimer´s disease. In order to repair or prevent such diseases, drugs are designed which enter the brain, but the blood-brain barrier limits their entry and the search for alternative pathways is important. Recently, we reported that intranasal delivery of the amyloid-beta degrading enzyme neprilysin eliminated amyloid-beta plaques in transgenic Alzheimer´s disease mice. This review describes the anatomical structure of the intranasal pathway, explains the intranasal delivery of pure neprilysin, cell-loaded neprilysin (platelets) and collagen-embedded neprilysin to destruct amyloid-beta plaques in Alzheimer´s disease in transgenic APP_SweDI mice and hypothesizes why this may cause compensation and why the amyloid-beta cascade hypothesis may fail.
Collapse
Affiliation(s)
- Christian Humpel
- Laboratory of Psychiatry and Experimental Alzheimer's Research, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
15
|
Mahajan C, Kapoor I, Prabhakar H. A Narrative Review on Translational Research in Acute Brain Injury. JOURNAL OF NEUROANAESTHESIOLOGY AND CRITICAL CARE 2022. [DOI: 10.1055/s-0042-1744399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
AbstractThere has been a constant endeavor to reduce the mortality and morbidity associated with acute brain injury. The associated complex mechanisms involving biomechanics, markers, and neuroprotective drugs/measures have been extensively studied in preclinical studies with an ultimate aim to improve the patients' outcomes. Despite such efforts, only few have been successfully translated into clinical practice. In this review, we shall be discussing the major hurdles in the translation of preclinical results into clinical practice. The need is to choose an appropriate animal model, keeping in mind the species, age, and gender of the animal, choosing suitable outcome measures, ensuring quality of animal trials, and carrying out systematic review and meta-analysis of experimental studies before proceeding to human trials. The interdisciplinary collaboration between the preclinical and clinical scientists will help to design better, meaningful trials which might help a long way in successful translation. Although challenging at this stage, the advent of translational precision medicine will help the integration of mechanism-centric translational medicine and patient-centric precision medicine.
Collapse
Affiliation(s)
- Charu Mahajan
- Department of Neuroanaesthesiology and Critical Care, All India Institute of Medical Sciences, New Delhi, India
| | - Indu Kapoor
- Department of Neuroanaesthesiology and Critical Care, All India Institute of Medical Sciences, New Delhi, India
| | - Hemanshu Prabhakar
- Department of Neuroanaesthesiology and Critical Care, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
16
|
Li C, Huang S, Zhou W, Xie Z, Xie S, Li M. Effects of the Notch Signaling Pathway on Secondary Brain Changes Caused by Spinal Cord Injury in Mice. Neurochem Res 2022; 47:1651-1663. [PMID: 35211828 DOI: 10.1007/s11064-022-03558-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 01/22/2022] [Accepted: 02/14/2022] [Indexed: 12/15/2022]
Abstract
Spinal cord injury (SCI) can cause secondary brain changes, leading to hypomyelination in the dorsolateral prefrontal cortex (dlPFC). Some studies have shown that notch signaling pathway activation can regulate oligodendrocyte maturation and myelination. The aim of this study was to investigate whether inhibition of the Notch signaling pathway can alleviate hypomyelination in the dlPFC caused by SCI. Moreover, we further investigated whether the changes in myelination in the dlPFC are associated with neuropathic pain following SCI. We established a mouse model of SCI and observed the changes in mechanical and thermal hyperalgesia. Western blotting and immunofluorescence were used to analyze the changes in myelination in the dlPFC. The results indicated the existence of a relationship between activation of the Notch signaling pathway and hypomyelination in the dlPFC and confirmed the existence of a relationship between hypomyelination in the dlPFC and decreases in mechanical and thermal hyperalgesia thresholds. In conclusion, these results suggested that the Notch signaling pathway is activated after SCI, leading to hypomyelination in the dlPFC, and that DAPT can inhibit the Notch signaling pathway and improve mechanical and thermal hyperalgesia thresholds. Our findings provide a new target for the treatment of neuropathic pain caused by SCI.
Collapse
Affiliation(s)
- Chengcai Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, NO17 Yong Wai Zheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Shaoxin Huang
- School of Basic Medicine, Jiujiang University, Jiujiang, 332005, Jiangxi, People's Republic of China
| | - Wu Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, NO17 Yong Wai Zheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Zhiping Xie
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, NO17 Yong Wai Zheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Shenke Xie
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, NO17 Yong Wai Zheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Meihua Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, NO17 Yong Wai Zheng Street, Nanchang, 330006, Jiangxi, People's Republic of China.
| |
Collapse
|
17
|
Manni L, Conti G, Chiaretti A, Soligo M. Intranasal Delivery of Nerve Growth Factor in Neurodegenerative Diseases and Neurotrauma. Front Pharmacol 2021; 12:754502. [PMID: 34867367 PMCID: PMC8635100 DOI: 10.3389/fphar.2021.754502] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/01/2021] [Indexed: 01/01/2023] Open
Abstract
Since the 1980s, the development of a pharmacology based on nerve growth factor (NGF) has been postulated for the therapy of Alzheimer’s disease (AD). This hypothesis was based on the rescuing effect of the neurotrophin on the cholinergic phenotype of the basal forebrain neurons, primarily compromised during the development of AD. Subsequently, the use of NGF was put forward to treat a broader spectrum of neurological conditions affecting the central nervous system, such as Parkinson’s disease, degenerative retinopathies, severe brain traumas and neurodevelopmental dysfunctions. While supported by solid rational assumptions, the progress of a pharmacology founded on these hypotheses has been hampered by the difficulty of conveying NGF towards the brain parenchyma without resorting to invasive and risky delivery methods. At the end of the last century, it was shown that NGF administered intranasally to the olfactory epithelium was able to spread into the brain parenchyma. Notably, after such delivery, pharmacologically relevant concentration of exogenous NGF was found in brain areas located at considerable distances from the injection site along the rostral-caudal axis. These observations paved the way for preclinical characterization and clinical trials on the efficacy of intranasal NGF for the treatment of neurodegenerative diseases and of the consequences of brain trauma. In this review, a summary of the preclinical and clinical studies published to date will be attempted, as well as a discussion about the mechanisms underlying the efficacy and the possible development of the pharmacology based on intranasal conveyance of NGF to the brain.
Collapse
Affiliation(s)
- Luigi Manni
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), Rome, Italy
| | - Giorgio Conti
- Department of Emergency, Intensive Pediatric Therapy and Pediatric Trauma Center, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Antonio Chiaretti
- Department of Woman and Child Health, Institute of Pediatrics, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Marzia Soligo
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), Rome, Italy
| |
Collapse
|
18
|
Bahlakeh G, Rahbarghazi R, Mohammadnejad D, Abedelahi A, Karimipour M. Current knowledge and challenges associated with targeted delivery of neurotrophic factors into the central nervous system: focus on available approaches. Cell Biosci 2021; 11:181. [PMID: 34641969 PMCID: PMC8507154 DOI: 10.1186/s13578-021-00694-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 09/28/2021] [Indexed: 12/23/2022] Open
Abstract
During the last decades, numerous basic and clinical studies have been conducted to assess the delivery efficiency of therapeutic agents into the brain and spinal cord parenchyma using several administration routes. Among conventional and in-progress administrative routes, the eligibility of stem cells, viral vectors, and biomaterial systems have been shown in the delivery of NTFs. Despite these manifold advances, the close association between the delivery system and regeneration outcome remains unclear. Herein, we aimed to discuss recent progress in the delivery of these factors and the pros and cons related to each modality.
Collapse
Affiliation(s)
- Gozal Bahlakeh
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Daruosh Mohammadnejad
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Abedelahi
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad Karimipour
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran. .,Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
19
|
Perez Garcia G, De Gasperi R, Tschiffely AE, Gama Sosa MA, Abutarboush R, Kawoos U, Statz JK, Ciarlone S, Reed EM, Jeyarajah T, Perez G, Otero Pagan A, Pryor D, Hof P, Cook D, Gandy S, Elder G, Ahlers S. Repetitive low-level blast exposure improves behavioral deficits and chronically lowers Aβ42 in an Alzheimer's disease transgenic mouse model. J Neurotrauma 2021; 38:3146-3173. [PMID: 34353119 DOI: 10.1089/neu.2021.0184] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Public awareness of traumatic brain injury (TBI) in the military increased recently because of the conflicts in Iraq and Afghanistan where blast injury was the most common mechanism of injury. Besides overt injuries, concerns also exist over the potential adverse consequences of subclinical blast exposures, which are common for many service members. TBI is a risk factor for the later development of neurodegenerative diseases, including Alzheimer's disease (AD)-like disorders. Studies of acute TBI in humans and animals have suggested that increased processing of the amyloid precursor protein (APP) towards the amyloid beta protein (Aβ) may explain the epidemiological associations with AD. However, in a prior study we found in both rat and mouse models of blast overpressure exposure (BOP), that rather than increasing, rodent brain Aβ42 levels were decreased following acute blast exposure. Here we subjected APP/presenilin 1 transgenic mice (APP/PS1 Tg) to an extended sequence of repetitive low-level blast exposures (34.5 kPa) administered three times per week over 8 weeks. If initiated at 20 weeks of age, these repetitive exposures, which were designed to mimic human subclinical blast exposures, reduced anxiety and improved cognition as well as social interactions in APP/PS1 Tg mice, returning many behavioral parameters in APP/PS1 Tg mice to levels of non-transgenic wild type mice. Repetitive low-level blast exposure was less effective at improving behavioral deficits in APP/PS1 Tg mice when begun at 36 weeks of age. While amyloid plaque loads were unchanged, Aβ42 levels and Aβ oligomers were reduced in brain of mice exposed to repetitive low-level blast exposures initiated at 20 weeks of age, although levels did not directly correlate with behavioral parameters in individual animals. These results have implications for understanding the nature of blast effects on the brain and their relationship to human neurodegenerative diseases.
Collapse
Affiliation(s)
- Georgina Perez Garcia
- Icahn School of Medicine at Mount Sinai, 5925, Neurology, 1468 Madison Avenue Annenberg Building Floor 14 Room 60, New York, New York, New York, United States, 10029-6574.,James J Peters VA Medical Center, 20071, Research, 130 W Kingsbridge Rd, The Bronx, NY 10468, Bronx, United States, 10468-3904;
| | - Rita De Gasperi
- James J. Peters VA Medical Center, Research and Development, 130 west kingsbridge road, RD 3F-20, Bronx, New York, United States, 10468;
| | - Anna E Tschiffely
- Naval Medical Research Center, 19930, Silver Spring, Maryland, United States;
| | - Miguel A Gama Sosa
- James J. Peters VA Medical Center, Research and Development, 130 W Kingsbridge Rd, Bronx, New York, United States, 10468;
| | - Rania Abutarboush
- Naval Medical Research Center, 19930, Neurotrauma, 503 Robert Grant Ave, Silver Spring, Maryland, United States, 20910;
| | - Usmah Kawoos
- Naval Medical Research Center, 19930, Neurotrauma, 503 Robert Grant Ave, Silver Spring, Maryland, United States, 20910.,Henry M Jackson Foundation for the Advancement of Military Medicine Inc, 44069, Bethesda, Maryland, United States;
| | | | - Stephanie Ciarlone
- Naval Medical Research Center, 19930, Silver Spring, Maryland, United States;
| | - Eileen M Reed
- Naval Medical Research Center, 19930, Silver Spring, Maryland, United States;
| | - Theepica Jeyarajah
- Naval Medical Research Center, 19930, Silver Spring, Maryland, United States;
| | - Gissel Perez
- James J Peters VA Medical Center, 20071, Research and Development, Bronx, New York, United States;
| | - Alena Otero Pagan
- James J Peters VA Medical Center, 20071, Research and Development, Bronx, New York, United States;
| | - Dylan Pryor
- James J Peters VA Medical Center, 20071, Research, 130 W. Kingsbridge Rd., Bronx, New York, United States, 10468;
| | - Patrick Hof
- Icahn School of Medicine at Mount Sinai, 5925, New York, New York, United States;
| | - David Cook
- VA Puget Sound Health Care System, 20128, Geriatric Research, Education, and Clinical Center, 1660 S Columbian Way, Seattle, Washington, United States, 98108.,University of Washington, 7284, Division of Gerontology and Geriatric Medicine, Seattle, Washington, United States;
| | - Samuel Gandy
- 88 Mercer AvenueHartsdaleHartsdale, New York, United States, 10530.,Sam Gandy, 88 Mercer Avenue, United States;
| | - Gregory Elder
- James J. Peters VAMC, Research and Development 3F22, 130 West Kingsbridge Road, Bronx, New York, United States, 10468;
| | - Stephen Ahlers
- Naval Medical Research Center, OUMD, 503 Robert Grant Ave, Silver Spring, Maryland, United States, 20910;
| |
Collapse
|
20
|
Humpel C. Intranasal Delivery of Collagen-Loaded Neprilysin Clears Beta-Amyloid Plaques in a Transgenic Alzheimer Mouse Model. Front Aging Neurosci 2021; 13:649646. [PMID: 33967739 PMCID: PMC8100061 DOI: 10.3389/fnagi.2021.649646] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/29/2021] [Indexed: 11/13/2022] Open
Abstract
Alzheimer's disease (AD) is pathologically characterized by extracellular beta-amyloid (Aβ) plaques and intraneuronal tau tangles in the brain. A therapeutic strategy aims to prevent or clear these Aβ plaques and the Aβ-degrading enzyme neprilysin is a potent drug to degrade plaques. The major challenge is to deliver bioactive neprilysin into the brain via the blood-brain barrier. The aim of the present study is to explore if intranasal delivery of neprilysin can eliminate plaques in a transgenic AD mouse model (APP_SweDI). We will test if collagen or platelets are useful vehicles to deliver neprilysin into the brain. Using organotypic brain slices from adult transgenic APP_SweDI mice, we show that neprilysin alone or loaded in collagen hydrogels or in platelets cleared cortical plaques. Intransasal delivery of neprilysin alone increased small Aβ depositions in the middle and caudal cortex in transgenic mice. Platelets loaded with neprilysin cleared plaques in the frontal cortex after intranasal application. Intranasal delivery of collagen-loaded neprilysin was very potent to clear plaques especially in the middle and caudal parts of the cortex. Our data support that the Aβ degrading enzyme neprilysin delivered to the mouse brain can clear Aβ plaques and intranasal delivery (especially with collagen as a vehicle) is a fast and easy application. However, it must be considered that intranasal neprilysin may also activate more plaque production in the transgenic mouse brain as a side effect.
Collapse
Affiliation(s)
- Christian Humpel
- Laboratory of Psychiatry and Experimental Alzheimer’s Research, Department of Psychiatry and Psychotherapy, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
21
|
Eftimiadi G, Soligo M, Manni L, Di Giuda D, Calcagni ML, Chiaretti A. Topical delivery of nerve growth factor for treatment of ocular and brain disorders. Neural Regen Res 2021; 16:1740-1750. [PMID: 33510063 PMCID: PMC8328750 DOI: 10.4103/1673-5374.306062] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Neurotrophins are a family of proteins that support neuronal proliferation, survival, and differentiation in the central and peripheral nervous systems, and are regulators of neuronal plasticity. Nerve growth factor is one of the best-described neurotrophins and has advanced to clinical trials for treatment of ocular and brain diseases due to its trophic and regenerative properties. Prior trials over the past few decades have produced conflicting results, which have principally been ascribed to adverse effects of systemic nerve growth factor administration, together with poor penetrance of the blood-brain barrier that impairs drug delivery. Contrastingly, recent studies have revealed that topical ocular and intranasal nerve growth factor administration are safe and effective, suggesting that topical nerve growth factor delivery is a potential alternative to both systemic and invasive intracerebral delivery. The therapeutic effects of local nerve growth factor delivery have been extensively investigated for different ophthalmic diseases, including neurotrophic keratitis, glaucoma, retinitis pigmentosa, and dry eye disease. Further, promising pharmacologic effects were reported in an optic glioma model, which indicated that topically administered nerve growth factor diffused far beyond where it was topically applied. These findings support the therapeutic potential of delivering topical nerve growth factor preparations intranasally for acquired and degenerative brain disorders. Preliminary clinical findings in both traumatic and non-traumatic acquired brain injuries are encouraging, especially in pediatric patients, and clinical trials are ongoing. The present review will focus on the therapeutic effects of both ocular and intranasal nerve growth factor delivery for diseases of the brain and eye.
Collapse
Affiliation(s)
- Gemma Eftimiadi
- Institute of Pediatrics, Fondazione Policlinico Universitario Agostino Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Marzia Soligo
- Institute of Translational Pharmacology, Consiglio Nazionale delle Ricerche (CNR), Rome, Italy
| | - Luigi Manni
- Institute of Translational Pharmacology, Consiglio Nazionale delle Ricerche (CNR), Rome, Italy
| | - Daniela Di Giuda
- Institute of Nuclear Medicine, Fondazione Policlinico Universitario Agostino Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Maria Lucia Calcagni
- Institute of Nuclear Medicine, Fondazione Policlinico Universitario Agostino Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Antonio Chiaretti
- Institute of Pediatrics, Fondazione Policlinico Universitario Agostino Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| |
Collapse
|
22
|
Crupi R, Cordaro M, Cuzzocrea S, Impellizzeri D. Management of Traumatic Brain Injury: From Present to Future. Antioxidants (Basel) 2020; 9:antiox9040297. [PMID: 32252390 PMCID: PMC7222188 DOI: 10.3390/antiox9040297] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 03/29/2020] [Accepted: 03/31/2020] [Indexed: 12/12/2022] Open
Abstract
TBI (traumatic brain injury) is a major cause of death among youth in industrialized societies. Brain damage following traumatic injury is a result of direct and indirect mechanisms; indirect or secondary injury involves the initiation of an acute inflammatory response, including the breakdown of the blood–brain barrier (BBB), brain edema, infiltration of peripheral blood cells, and activation of resident immunocompetent cells, as well as the release of numerous immune mediators such as interleukins and chemotactic factors. TBI can cause changes in molecular signaling and cellular functions and structures, in addition to tissue damage, such as hemorrhage, diffuse axonal damages, and contusions. TBI typically disturbs brain functions such as executive actions, cognitive grade, attention, memory data processing, and language abilities. Animal models have been developed to reproduce the different features of human TBI, better understand its pathophysiology, and discover potential new treatments. For many years, the first approach to manage TBI has been treatment of the injured tissue with interventions designed to reduce the complex secondary-injury cascade. Several studies in the literature have stressed the importance of more closely examining injuries, including endothelial, microglia, astroglia, oligodendroglia, and precursor cells. Significant effort has been invested in developing neuroprotective agents. The aim of this work is to review TBI pathophysiology and existing and potential new therapeutic strategies in the management of inflammatory events and behavioral deficits associated with TBI.
Collapse
Affiliation(s)
- Rosalia Crupi
- Department of Veterinary Science, University of Messina, 98168 Messina, Italy;
| | - Marika Cordaro
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Via Consolare Valeria 1, 98100 Messina, Italy;
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmacological and Environmental Sciences, Messina University, Viale F. Stagno D’Alcontres 31, 98166 Messina, Italy;
- Department of Pharmacological and Physiological Science, Saint Louis University, Saint Louis, MO 63104, USA
- Correspondence: ; Tel.: +390-906-765-208
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmacological and Environmental Sciences, Messina University, Viale F. Stagno D’Alcontres 31, 98166 Messina, Italy;
| |
Collapse
|
23
|
Montroull LE, Rothbard DE, Kanal HD, D’Mello V, Dodson V, Troy CM, Zanin JP, Levison SW, Friedman WJ. Proneurotrophins Induce Apoptotic Neuronal Death After Controlled Cortical Impact Injury in Adult Mice. ASN Neuro 2020; 12:1759091420930865. [PMID: 32493127 PMCID: PMC7273561 DOI: 10.1177/1759091420930865] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 05/05/2020] [Indexed: 11/29/2022] Open
Abstract
The p75 neurotrophin receptor (p75NTR) can regulate multiple cellular functions including proliferation, survival, and apoptotic cell death. The p75NTR is widely expressed in the developing brain and is downregulated as the nervous system matures, with only a few neuronal subpopulations retaining expression into adulthood. However, p75NTR expression is induced following damage to the adult brain, including after traumatic brain injury, which is a leading cause of mortality and disability worldwide. A major consequence of traumatic brain injury is the progressive neuronal loss that continues secondary to the initial trauma, which ultimately contributes to cognitive decline. Understanding mechanisms governing this progressive neuronal death is key to developing targeted therapeutic strategies to provide neuroprotection and salvage cognitive function. In this study, we demonstrate that a cortical impact injury to the sensorimotor cortex elicits p75NTR expression in apoptotic neurons in the injury penumbra, confirming previous studies. To establish whether preventing p75NTR induction or blocking the ligands would reduce the extent of secondary neuronal cell death, we used a noninvasive intranasal strategy to deliver either siRNA to block the induction of p75NTR, or function-blocking antibodies to the ligands pro-nerve growth factor and pro-brain-derived neurotrophic factor. We demonstrate that either preventing the induction of p75NTR or blocking the proneurotrophin ligands provides neuroprotection and preserves sensorimotor function.
Collapse
Affiliation(s)
- Laura E. Montroull
- Department of Biological
Sciences, Rutgers University, Newark, New Jersey, United States
| | - Deborah E. Rothbard
- Department of Pharmacology,
Physiology and Neuroscience, Rutgers University, Newark, New Jersey,
United States
| | - Hur D. Kanal
- Department of Pharmacology,
Physiology and Neuroscience, Rutgers University, Newark, New Jersey,
United States
| | - Veera D’Mello
- Department of Pharmacology,
Physiology and Neuroscience, Rutgers University, Newark, New Jersey,
United States
| | - Vincent Dodson
- Department of Pharmacology,
Physiology and Neuroscience, Rutgers University, Newark, New Jersey,
United States
| | - Carol M. Troy
- Department of Pathology and
Cell Biology, Columbia University Medical Center, New York, NY, United
States
| | - Juan P. Zanin
- Department of Biological
Sciences, Rutgers University, Newark, New Jersey, United States
| | - Steven W. Levison
- Department of Pharmacology,
Physiology and Neuroscience, Rutgers University, Newark, New Jersey,
United States
| | - Wilma J. Friedman
- Department of Biological
Sciences, Rutgers University, Newark, New Jersey, United States
| |
Collapse
|
24
|
Ojo JO, Leary P, Lungmus C, Algamal M, Mouzon B, Bachmeier C, Mullan M, Stewart W, Crawford F. Subchronic Pathobiological Response Following Chronic Repetitive Mild Traumatic Brain Injury in an Aged Preclinical Model of Amyloid Pathogenesis. J Neuropathol Exp Neurol 2019; 77:1144-1162. [PMID: 30395237 DOI: 10.1093/jnen/nly101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 10/04/2018] [Indexed: 12/14/2022] Open
Abstract
Repetitive mild traumatic brain injury (r-mTBI) is a risk factor for Alzheimer disease (AD). The precise nature of how r-mTBI leads to, or precipitates, AD pathogenesis remains unclear. In this study, we explore subchronic effects of chronic r-mTBI (12-impacts) administered over 1-month in aged-PS1/APP mice and littermate controls. We investigate specific mechanisms that may elucidate the molecular link between AD and r-mTBI, focusing primarily on amyloid and tau pathology, amyloid processing, glial activation states, and associated clearance mechanisms. Herein, we demonstrate r-mTBI in aged PS1/APP mice does not augment, glial activation, amyloid burden, or tau pathology (with exception of pS202-positive Tau) 1 month after exposure to the last-injury. However, we observed a decrease in brain soluble Aβ42 levels without any appreciable change in peripheral soluble Aβ42 levels. This was accompanied by an increase in brain insoluble to soluble Aβ42 ratio in injured PS1/APP mice compared with sham injury. A parallel reduction in phagocytic receptor, triggering receptor expressed on myeloid cells 2, was also observed. This study demonstrates very subtle subchronic effects of r-mTBI on a preexisting amyloid pathology background, which may be on a continuum toward a slow and worsening neurodegenerative outcome compared with sham injury, and therefore, have many implications, especially in the elderly population exposed to TBI.
Collapse
Affiliation(s)
- Joseph O Ojo
- Experimental Neuropathology and TBI Research Division, Roskamp Institute, Sarasota, Florida.,James A. Haley Veterans' Hospital, Tampa, Florida.,Open University, Milton Keynes, UK
| | - Paige Leary
- Experimental Neuropathology and TBI Research Division, Roskamp Institute, Sarasota, Florida
| | - Caryln Lungmus
- Experimental Neuropathology and TBI Research Division, Roskamp Institute, Sarasota, Florida
| | - Moustafa Algamal
- Experimental Neuropathology and TBI Research Division, Roskamp Institute, Sarasota, Florida.,Open University, Milton Keynes, UK
| | - Benoit Mouzon
- Experimental Neuropathology and TBI Research Division, Roskamp Institute, Sarasota, Florida.,James A. Haley Veterans' Hospital, Tampa, Florida.,Open University, Milton Keynes, UK
| | - Corbin Bachmeier
- Experimental Neuropathology and TBI Research Division, Roskamp Institute, Sarasota, Florida.,Open University, Milton Keynes, UK.,Bay Pines VA Healthcare System, Bay Pines, Florida
| | - Michael Mullan
- Experimental Neuropathology and TBI Research Division, Roskamp Institute, Sarasota, Florida.,Open University, Milton Keynes, UK
| | - William Stewart
- Queen Elizabeth University Hospital and University of Glasgow, Glasgow, UK.,University of Pennsylvania, Philadelphia, Pennsylvania
| | - Fiona Crawford
- Experimental Neuropathology and TBI Research Division, Roskamp Institute, Sarasota, Florida.,James A. Haley Veterans' Hospital, Tampa, Florida.,Open University, Milton Keynes, UK
| |
Collapse
|
25
|
Zhang L, Chen C, Mak MSH, Lu J, Wu Z, Chen Q, Han Y, Li Y, Pi R. Advance of sporadic Alzheimer's disease animal models. Med Res Rev 2019; 40:431-458. [DOI: 10.1002/med.21624] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 05/21/2019] [Accepted: 06/27/2019] [Indexed: 01/06/2023]
Affiliation(s)
- Lili Zhang
- School of Pharmaceutical SciencesSun Yat‐Sen UniversityGuangzhou China
| | - Chen Chen
- School of Pharmaceutical SciencesSun Yat‐Sen UniversityGuangzhou China
| | - Marvin SH Mak
- Department of Applied Biology and Chemical Technology, Institute of Modern Chinese MedicineThe Hong Kong Polytechnic University, Hung Hom Hong Kong
| | - Junfeng Lu
- School of Pharmaceutical SciencesSun Yat‐Sen UniversityGuangzhou China
| | - Zeqing Wu
- School of Pharmaceutical SciencesSun Yat‐Sen UniversityGuangzhou China
| | - Qiuhe Chen
- School of Pharmaceutical SciencesSun Yat‐Sen UniversityGuangzhou China
| | - Yifan Han
- Department of Applied Biology and Chemical Technology, Institute of Modern Chinese MedicineThe Hong Kong Polytechnic University, Hung Hom Hong Kong
- International Joint Laboratory<SYSU‐PolyU HK>of Novel Anti‐Dementia Drugs of GuangzhouGuangzhou China
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation)The Hong Kong Polytechnic University Shenzhen Research InstituteShenzhen China
| | - Yuefeng Li
- Guangdong Landau Biotechnology Co LtdGuangzhou China
| | - Rongbiao Pi
- School of Pharmaceutical SciencesSun Yat‐Sen UniversityGuangzhou China
- International Joint Laboratory<SYSU‐PolyU HK>of Novel Anti‐Dementia Drugs of GuangzhouGuangzhou China
- National and Local United Engineering Lab of Druggability and New Drugs EvaluationSun Yat‐Sen UniversityGuangzhou China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of MedicineSun Yat‐Sen UniversityGuangzhou China
| |
Collapse
|
26
|
Traumatic Brain Injury by Weight-Drop Method Causes Transient Amyloid- β Deposition and Acute Cognitive Deficits in Mice. Behav Neurol 2019; 2019:3248519. [PMID: 30944661 PMCID: PMC6421814 DOI: 10.1155/2019/3248519] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 12/11/2018] [Accepted: 01/09/2019] [Indexed: 11/17/2022] Open
Abstract
There has been growing awareness of the correlation between an episode of traumatic brain injury (TBI) and the development of Alzheimer's disease (AD) later in life. It has been reported that TBI accelerated amyloid-β (Aβ) pathology and cognitive decline in the several lines of AD model mice. However, the short-term and long-term effects of TBI by the weight-drop method on amyloid-β pathology and cognitive performance are unclear in wild-type (WT) mice. Hence, we examined AD-related histopathological changes and cognitive impairment after TBI in wild-type C57BL6J mice. Five- to seven-month-old WT mice were subjected to either TBI by the weight-drop method or a sham treatment. Seven days after TBI, the WT mice exhibited significantly lower spatial learning than the sham-treated WT mice. However, 28 days after TBI, the cognitive impairment in the TBI-treated WT mice recovered. Correspondingly, while significant amyloid-β (Aβ) plaques and amyloid precursor protein (APP) accumulation were observed in the TBI-treated mouse hippocampus 7 days after TBI, the Aβ deposition was no longer apparent 28 days after TBI. Thus, TBI induced transient amyloid-β deposition and acute cognitive impairments in the WT mice. The present study suggests that the TBI could be a risk factor for acute cognitive impairment even when genetic and hereditary predispositions are not involved. The system might be useful for evaluating and developing a pharmacological treatment for the acute cognitive deficits.
Collapse
|
27
|
The delayed protective effect of GK-2, а dipeptide mimetic of Nerve Growth Factor, in a model of rat traumatic brain injury. Brain Res Bull 2018; 140:148-153. [PMID: 29730416 DOI: 10.1016/j.brainresbull.2018.05.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 04/29/2018] [Accepted: 05/02/2018] [Indexed: 11/24/2022]
Abstract
The delayed protective effect of GK-2, a dipeptide mimetic of Nerve Growth Factor, was investigated on the model of focal one-sided traumatic brain injury (TBI) of the sensorimotor cortex region on the 180th day after the injury. TBI caused a reliably disruption of the functions of the limbs contralateral to injury focus. The intraperitoneal administration of GK-2 (1 mg/kg) from 1st to 4th and from 7th to 10th days after TBI reduced the impairment of the motor functions of the limbs. This therapeutic effect significant manifested itself from the 7th day and continued until the end of the experiment - on the 180th day after TBI. Morphological studies of the animal brains on the 180th day after TBI demonstrated a decrease in the number of neurons in the V layer of the cerebral cortex and a decrease in the thickness of the corpus callosum. The treatment of animals with GK-2 after TBI statistically significant prevented a decrease in the density of neurons in the ipsilateral hemisphere and a decrease in the thickness of the corpus callosum in the contralateral hemisphere in comparison with untreated animals. Additionally, we showed in vitro that GK-2 exhibits neuroprotective properties under oxidative stress in primary hippocampal cultures. Our results demonstrate that the use of GK-2 at the early stages of development of traumatic brain damage can prevent such delayed damage as neuronal and axonal degeneration as well as reduce TBI-related disruptions of brain functions.
Collapse
|
28
|
Rocco ML, Soligo M, Manni L, Aloe L. Nerve Growth Factor: Early Studies and Recent Clinical Trials. Curr Neuropharmacol 2018; 16:1455-1465. [PMID: 29651949 PMCID: PMC6295934 DOI: 10.2174/1570159x16666180412092859] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 03/23/2018] [Accepted: 04/04/2018] [Indexed: 12/02/2022] Open
Abstract
Since its discovery, nerve growth factor (NGF) has long occupied a critical role in developmental and adult neurobiology for its many important regulatory functions on the survival, growth and differentiation of nerve cells in the peripheral and central nervous system. NGF is the first discovered member of a family of neurotrophic factors, collectively indicated as neurotrophins, (which include brain-derived neurotrophic factor, neurotrophin-3 and neurotrophin 4/5). NGF was discovered for its action on the survival and differentiation of selected populations of peripheral neurons. Since then, an enormous number of basic and human studies were undertaken to explore the role of purified NGF to prevent the death of NGF-receptive cells. These studies revealed that NGF possesses important therapeutic properties, after topical administration, on human cutaneous pressure ulcer, corneal ulcers, glaucoma, retinal maculopathy, Retinitis Pigmentosa and in pediatric optic gliomas and brain traumas. The aim of this review is to present our previous, recent and ongoing clinical studies on the therapeutic properties of NGF.
Collapse
Affiliation(s)
| | | | | | - Luigi Aloe
- Address correspondence to this author at the Fondazione IRET ONLUS, Via Tolara di Sopra 41/E, 40064 Ozzano Emilia (BO), Italy; Tel: +39-051-798776; Fax: +39-051-799673; E-mail:
| |
Collapse
|
29
|
Johnstone MR, Sun M, Taylor CJ, Brady RD, Grills BL, Church JE, Shultz SR, McDonald SJ. Gambogic amide, a selective TrkA agonist, does not improve outcomes from traumatic brain injury in mice. Brain Inj 2017; 32:257-268. [PMID: 29227174 DOI: 10.1080/02699052.2017.1394492] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVES There is evidence that treatment with nerve growth factor (NGF) may reduce neuroinflammation and apoptosis after a traumatic brain injury (TBI). NGF is thought to exert its effects via binding to either TrkA or p75 neurotrophin receptors. This study aimed to investigate the effects of a selective TrkA agonist, gambogic amide (GA), on TBI pathology and outcomes in mice following lateral fluid percussion injury. METHODS Male C57BL/6 mice were given either a TBI or sham injury, and then received subcutaneous injections of either 2 mg/kg of GA or vehicle at 1, 24, and 48 h post-injury. Following behavioural studies, mice were euthanized at 72 h post-injury for analysis of neuroinflammatory, apoptotic, and neurite outgrowth markers. RESULTS Behavioural testing revealed that GA did not mitigate motor deficits after TBI. TBI caused an increase in cortical and hippocampal expression of several markers of neuroinflammation and apoptosis compared to sham groups. GA treatment did not attenuate these increases in expression, possibly contributed to by our finding of TrkA receptor down-regulation post-TBI. CONCLUSIONS These findings suggest that GA treatment may not be suitable for attenuating TBI pathology and improving outcomes.
Collapse
Affiliation(s)
- Maddison R Johnstone
- a Department of Physiology, Anatomy and Microbiology , School of Life Sciences, La Trobe University , Melbourne , VIC , Australia
| | - Mujun Sun
- b Department of Medicine , The Royal Melbourne Hospital, The University of Melbourne , Parkville , VIC , Australia
| | - Caroline J Taylor
- a Department of Physiology, Anatomy and Microbiology , School of Life Sciences, La Trobe University , Melbourne , VIC , Australia
| | - Rhys D Brady
- a Department of Physiology, Anatomy and Microbiology , School of Life Sciences, La Trobe University , Melbourne , VIC , Australia.,b Department of Medicine , The Royal Melbourne Hospital, The University of Melbourne , Parkville , VIC , Australia
| | - Brian L Grills
- a Department of Physiology, Anatomy and Microbiology , School of Life Sciences, La Trobe University , Melbourne , VIC , Australia
| | - Jarrod E Church
- a Department of Physiology, Anatomy and Microbiology , School of Life Sciences, La Trobe University , Melbourne , VIC , Australia
| | - Sandy R Shultz
- b Department of Medicine , The Royal Melbourne Hospital, The University of Melbourne , Parkville , VIC , Australia.,c Department of Neuroscience , Central Clinical School, Monash University , Melbourne , VIC , Australia
| | - Stuart J McDonald
- a Department of Physiology, Anatomy and Microbiology , School of Life Sciences, La Trobe University , Melbourne , VIC , Australia
| |
Collapse
|
30
|
Samaridou E, Alonso MJ. Nose-to-brain peptide delivery - The potential of nanotechnology. Bioorg Med Chem 2017; 26:2888-2905. [PMID: 29170026 DOI: 10.1016/j.bmc.2017.11.001] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 10/26/2017] [Accepted: 11/02/2017] [Indexed: 12/11/2022]
Abstract
Nose-to-brain (N-to-B) delivery offers to protein and peptide drugs the possibility to reach the brain in a non-invasive way. This article is a comprehensive review of the state-of-the-art of this emerging peptide delivery route, as well as of the challenges associated to it. Emphasis is given on the potential of nanosized drug delivery carriers to enhance the direct N-to-B transport of protein or peptide drugs. In particular, polymer- and lipid- based nanocarriers are comparatively analyzed in terms of the influence of their physicochemical characteristics and composition on their in vivo fate and efficacy. The use of biorecognitive ligands and permeation enhancers in order to enhance their brain targeting efficiency is also discussed. The article concludes highlighting the early stage of this research field and its still unveiled potential. The final message is that more explicatory PK/PD studies are required in order to achieve the translation from preclinical to the clinical development phase.
Collapse
Affiliation(s)
- Eleni Samaridou
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Av. Barcelona s/n, Campus Vida, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Maria José Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Av. Barcelona s/n, Campus Vida, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| |
Collapse
|
31
|
Chiaretti A, Conti G, Falsini B, Buonsenso D, Crasti M, Manni L, Soligo M, Fantacci C, Genovese O, Calcagni ML, Di Giuda D, Mattoli MV, Cocciolillo F, Ferrara P, Ruggiero A, Staccioli S, Colafati GS, Riccardi R. Intranasal Nerve Growth Factor administration improves cerebral functions in a child with severe traumatic brain injury: A case report. Brain Inj 2017; 31:1538-1547. [PMID: 28972396 DOI: 10.1080/02699052.2017.1376760] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND Nerve growth factor (NGF) promotes neural recovery after experimental traumatic brain injury (TBI) supporting neuronal growth, differentiation and survival of brain cells and up-regulating the neurogenesis-associated protein Doublecortin (DCX). Only a few studies reported NGF administration in paediatric patients with severe TBI. METHODS A four-year-old boy in a persistent unresponsive wakefulness syndrome (UWS) was treated with intranasal murine NGF administration 6 months after severe TBI. The patient received four cycles of intranasal NGF (0.1 mg/kg, twice a day for 10 consecutive days). RESULTS NGF administration improved functional [Positron Emission Tomography/Computed Tomography (PET/CT); Single photon emission/Computed Tomography (SPECT/CT) and Magnetic Resonance Imaging (MRI)] assessment, electrophysiological [Electroencephalogram (EEG) and Visual Evoked Potential (VEP)] studies and clinical conditions. He showed improvements in voluntary movements, facial mimicry, phonation, attention and verbal comprehension, ability to cry, cough reflex, oral motility, feeding capacity, and bowel and urinary functions. After NGF administration, raised levels of both NGF and DCX were found in the cerebrospinal fluid of the patient. No side effects were reported. CONCLUSIONS Although further studies are needed for better understanding the neuroprotective role of this neurotrophin, intranasal NGF administration appears to be a promising and safe rescuing strategy treatment in children with neurological impairment after TBI.
Collapse
Affiliation(s)
- Antonio Chiaretti
- a Institute of Pediatrics , Università Cattolica del Sacro Cuore , Rome , Italy
| | - Giorgio Conti
- b Pediatric Intensive Care Unit , Università Cattolica del Sacro Cuore , Rome , Italy
| | - Benedetto Falsini
- c Institute of Ophthalmology , Università Cattolica del Sacro Cuore , Rome , Italy
| | - Danilo Buonsenso
- a Institute of Pediatrics , Università Cattolica del Sacro Cuore , Rome , Italy
| | - Matteo Crasti
- a Institute of Pediatrics , Università Cattolica del Sacro Cuore , Rome , Italy
| | - Luigi Manni
- d Institute of Translational Pharmacology , CNR , Rome , Italy
| | - Marzia Soligo
- d Institute of Translational Pharmacology , CNR , Rome , Italy
| | - Claudia Fantacci
- a Institute of Pediatrics , Università Cattolica del Sacro Cuore , Rome , Italy
| | - Orazio Genovese
- b Pediatric Intensive Care Unit , Università Cattolica del Sacro Cuore , Rome , Italy
| | - Maria Lucia Calcagni
- e Institute of Nuclear Medicine , Università Cattolica del Sacro Cuore , Rome , Italy
| | - Daniela Di Giuda
- e Institute of Nuclear Medicine , Università Cattolica del Sacro Cuore , Rome , Italy
| | | | - Fabrizio Cocciolillo
- e Institute of Nuclear Medicine , Università Cattolica del Sacro Cuore , Rome , Italy
| | - Pietro Ferrara
- a Institute of Pediatrics , Università Cattolica del Sacro Cuore , Rome , Italy
| | - Antonio Ruggiero
- f Pediatric Oncology , Università Cattolica del Sacro Cuore , Rome , Italy
| | - Susanna Staccioli
- g Department of Neuroscience and Neurorehabilitation , Bambino Gesù Children's Hospital , Rome , Italy
| | | | - Riccardo Riccardi
- f Pediatric Oncology , Università Cattolica del Sacro Cuore , Rome , Italy
| |
Collapse
|
32
|
Rehman SU, Ahmad A, Yoon GH, Khan M, Abid MN, Kim MO. Inhibition of c-Jun N-Terminal Kinase Protects Against Brain Damage and Improves Learning and Memory After Traumatic Brain Injury in Adult Mice. Cereb Cortex 2017; 28:2854-2872. [DOI: 10.1093/cercor/bhx164] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 06/12/2017] [Indexed: 12/26/2022] Open
Affiliation(s)
- Shafiq Ur Rehman
- Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Ashfaq Ahmad
- Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Gwang-Ho Yoon
- Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Mehtab Khan
- Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Muhammad Noman Abid
- Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| | - Myeong Ok Kim
- Department of Biology and Applied Life Science (BK21), College of Natural Sciences, Gyeongsang National University, Jinju, Republic of Korea
| |
Collapse
|
33
|
Neurotrauma: The Crosstalk between Neurotrophins and Inflammation in the Acutely Injured Brain. Int J Mol Sci 2017; 18:ijms18051082. [PMID: 28524074 PMCID: PMC5454991 DOI: 10.3390/ijms18051082] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 04/25/2017] [Accepted: 05/11/2017] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI) is a major cause of morbidity and mortality among young individuals worldwide. Understanding the pathophysiology of neurotrauma is crucial for the development of more effective therapeutic strategies. After the trauma occurs, immediate neurologic damage is produced by the traumatic forces; this primary injury triggers a secondary wave of biochemical cascades together with metabolic and cellular changes, called secondary neural injury. In the scenario of the acutely injured brain, the ongoing secondary injury results in ischemia and edema culminating in an uncontrollable increase in intracranial pressure. These areas of secondary injury progression, or areas of “traumatic penumbra”, represent crucial targets for therapeutic interventions. Neurotrophins are a class of signaling molecules that promote survival and/or maintenance of neurons. They also stimulate axonal growth, synaptic plasticity, and neurotransmitter synthesis and release. Therefore, this review focuses on the role of neurotrophins in the acute post-injury response. Here, we discuss possible endogenous neuroprotective mechanisms of neurotrophins in the prevailing environment surrounding the injured areas, and highlight the crosstalk between neurotrophins and inflammation with focus on neurovascular unit cells, particularly pericytes. The perspective is that neurotrophins may represent promising targets for research on neuroprotective and neurorestorative processes in the short-term following TBI.
Collapse
|
34
|
Pearn ML, Niesman IR, Egawa J, Sawada A, Almenar-Queralt A, Shah SB, Duckworth JL, Head BP. Pathophysiology Associated with Traumatic Brain Injury: Current Treatments and Potential Novel Therapeutics. Cell Mol Neurobiol 2017; 37:571-585. [PMID: 27383839 PMCID: PMC11482200 DOI: 10.1007/s10571-016-0400-1] [Citation(s) in RCA: 210] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 06/24/2016] [Indexed: 12/20/2022]
Abstract
Traumatic brain injury (TBI) is one of the leading causes of death of young people in the developed world. In the United States alone, 1.7 million traumatic events occur annually accounting for 50,000 deaths. The etiology of TBI includes traffic accidents, falls, gunshot wounds, sports, and combat-related events. TBI severity ranges from mild to severe. TBI can induce subtle changes in molecular signaling, alterations in cellular structure and function, and/or primary tissue injury, such as contusion, hemorrhage, and diffuse axonal injury. TBI results in blood-brain barrier (BBB) damage and leakage, which allows for increased extravasation of immune cells (i.e., increased neuroinflammation). BBB dysfunction and impaired homeostasis contribute to secondary injury that occurs from hours to days to months after the initial trauma. This delayed nature of the secondary injury suggests a potential therapeutic window. The focus of this article is on the (1) pathophysiology of TBI and (2) potential therapies that include biologics (stem cells, gene therapy, peptides), pharmacological (anti-inflammatory, antiepileptic, progrowth), and noninvasive (exercise, transcranial magnetic stimulation). In final, the review briefly discusses membrane/lipid rafts (MLR) and the MLR-associated protein caveolin (Cav). Interventions that increase Cav-1, MLR formation, and MLR recruitment of growth-promoting signaling components may augment the efficacy of pharmacologic agents or already existing endogenous neurotransmitters and neurotrophins that converge upon progrowth signaling cascades resulting in improved neuronal function after injury.
Collapse
Affiliation(s)
- Matthew L Pearn
- Department of Anesthesiology, Veterans Affairs San Diego Healthcare System, VA Medical Center 125, University of California, 3350 La Jolla Village Drive, San Diego, CA, 92161-5085, USA
- Department of Anesthesiology, School of Medicine, University of California, La Jolla, San Diego, CA, 92093, USA
| | - Ingrid R Niesman
- Department of Cellular and Molecular Medicine, University of California, La Jolla, San Diego, CA, 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, San Diego, CA, 92037, USA
| | - Junji Egawa
- Department of Anesthesiology, Veterans Affairs San Diego Healthcare System, VA Medical Center 125, University of California, 3350 La Jolla Village Drive, San Diego, CA, 92161-5085, USA
- Department of Anesthesiology, School of Medicine, University of California, La Jolla, San Diego, CA, 92093, USA
| | - Atsushi Sawada
- Department of Anesthesiology, Veterans Affairs San Diego Healthcare System, VA Medical Center 125, University of California, 3350 La Jolla Village Drive, San Diego, CA, 92161-5085, USA
- Department of Anesthesiology, School of Medicine, University of California, La Jolla, San Diego, CA, 92093, USA
| | - Angels Almenar-Queralt
- Department of Cellular and Molecular Medicine, University of California, La Jolla, San Diego, CA, 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, San Diego, CA, 92037, USA
| | - Sameer B Shah
- UCSD Departments of Orthopaedic Surgery and Bioengineering, University of California, La Jolla, San Diego, CA, 92093, USA
| | - Josh L Duckworth
- Department of Neurology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Brian P Head
- Department of Anesthesiology, Veterans Affairs San Diego Healthcare System, VA Medical Center 125, University of California, 3350 La Jolla Village Drive, San Diego, CA, 92161-5085, USA.
- Department of Anesthesiology, School of Medicine, University of California, La Jolla, San Diego, CA, 92093, USA.
| |
Collapse
|
35
|
Back MJ, Lee HK, Lee JH, Fu Z, Son MW, Choi SZ, Go HS, Yoo S, Hwang SW, Kim DK. P2X1 Receptor-Mediated Ca 2+ Influx Triggered by DA-9801 Potentiates Nerve Growth Factor-Induced Neurite Outgrowth. ACS Chem Neurosci 2016; 7:1488-1498. [PMID: 27442785 DOI: 10.1021/acschemneuro.6b00082] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Nerve growth factor (NGF)-induced neuronal regeneration has emerged as a strategy to treat neuronal degeneration-associated disorders. However, direct NGF administration is limited by the occurrence of adverse effects at high doses of NGF. Therefore, development of a therapeutic strategy to promote the NGF trophic effect is required. In view of the lack of understanding of the mechanism for potentiating the NGF effect, this study investigated molecular targets of DA-9801, a well-standardized Dioscorea rhizome extract, which has a promoting effect on NGF. An increase in intracellular calcium ion level was induced by DA-9801, and chelation of extracellular calcium ions with ethylene-bis(oxyethylenenitrilo)tetraacetic acid (EGTA) suppressed the potentiating effect of DA-9801 on NGF-induced neurite outgrowth. In addition, EGTA treatment reduced the DA-9801-induced phosphorylation of extracellular signal-regulated kinase1/2 (ERK1/2), the major mediators of neurite outgrowth. To find which calcium ion-permeable channel contributes to the calcium ion influx induced by DA-9801, we treated PC12 cells with various inhibitors of calcium ion-permeable channels. NF449, a P2X1 receptor selective antagonist, significantly abolished the potentiating effect of DA-9801 on NGF-induced neurite outgrowth and abrogated the DA-9801-induced ERK1/2 phosphorylation. In addition, transfection with siRNA of P2X1 receptor significantly reduced the DA-9801-enhanced neurite outgrowth. In conclusion, calcium ion influx through P2X1 receptor mediated the promoting effect of DA-9801 on NGF-induced neurite outgrowth via ERK1/2 phosphorylation.
Collapse
Affiliation(s)
- Moon Jung Back
- Department
of Environmental and Health Chemistry, College of Pharmacy, Chung-Ang University; 84 Heukseok-ro, Dongjak-Ku, Seoul 06974, Republic of Korea
| | - Hae Kyung Lee
- Department
of Environmental and Health Chemistry, College of Pharmacy, Chung-Ang University; 84 Heukseok-ro, Dongjak-Ku, Seoul 06974, Republic of Korea
| | - Joo Hyun Lee
- Department
of Environmental and Health Chemistry, College of Pharmacy, Chung-Ang University; 84 Heukseok-ro, Dongjak-Ku, Seoul 06974, Republic of Korea
| | - Zhicheng Fu
- Department
of Environmental and Health Chemistry, College of Pharmacy, Chung-Ang University; 84 Heukseok-ro, Dongjak-Ku, Seoul 06974, Republic of Korea
| | - Mi Won Son
- Department of Research Planning & Management, Research Center of Dong-A ST Co., Ltd.; 21 Geumhwa-ro, 105 beon-gil, Giheung-gu, Yongin-si, Gyeonggi-do 446-905, Republic of Korea
| | - Sang Zin Choi
- Department of Research Planning & Management, Research Center of Dong-A ST Co., Ltd.; 21 Geumhwa-ro, 105 beon-gil, Giheung-gu, Yongin-si, Gyeonggi-do 446-905, Republic of Korea
| | - Hyo Sang Go
- Department of Research Planning & Management, Research Center of Dong-A ST Co., Ltd.; 21 Geumhwa-ro, 105 beon-gil, Giheung-gu, Yongin-si, Gyeonggi-do 446-905, Republic of Korea
| | - Sungjae Yoo
- Department
of Biomedical Sciences, Korea University College of Medicine, Seoul 136-705, Republic of Korea
| | - Sun Wook Hwang
- Department
of Biomedical Sciences, Korea University College of Medicine, Seoul 136-705, Republic of Korea
| | - Dae Kyong Kim
- Department
of Environmental and Health Chemistry, College of Pharmacy, Chung-Ang University; 84 Heukseok-ro, Dongjak-Ku, Seoul 06974, Republic of Korea
| |
Collapse
|
36
|
Zhou W, Zhang J, Wang G, Ling L, Yan C. Permeability and distribution of nerve growth factor in the brain of neonatal rats by periphery venous injection in hypoxic-ischemic state. SPRINGERPLUS 2016; 5:1893. [PMID: 27843750 PMCID: PMC5084138 DOI: 10.1186/s40064-016-3594-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 10/21/2016] [Indexed: 12/23/2022]
Abstract
Objective To investigate the permeability of β-NGF through blood–brain-barrier (BBB) in neonatal and adult rats, and the spatial distribution of β-NGF in different brain regions in hypoxic-ischemic (HI) and normal neonatal rats. Methods To investigate the overall permeability of β-NGF through BBB, β-NGF labeled with I125 was injected into adult rats, neonatal rats and HI neonatal rats via tail vein. The radioactivity of brain tissue and blood was examined and analyzed 30 min after injection. Also, brain regions including the basal forebrain, frontal cortex, hippocampus, hypothalamus, cerebellum, bulbus olfactorius and hypophysis, of all the rats were dissected and radioactivity was examined to investigate the spatial specificity of NGF permeation through BBB. Results Statistically significant results were observed in I125-β-NGF contents in brain tissues of adult rats group, neonatal rats group and HI neonatal rats group (P < 0.05). Compared to the HI neonatal rats’ brain with the highest I125-β-NGF contents, normal neonatal rats ranks the second while the adult rats were the lowest. While for the spatial specificity examination part, I125-β-NGF in both HI group and control group were widely distributed in basal forebrain, frontal cortex, hippocampus, cerebellum and bulbus olfactorius. But the radioactivity in frontal cortex, hippocampus and cerebellum of HI groups are statistically higher than control groups (P < 0.05). Conclusion β-NGF can more easily penetrate the BBB of newborn rats than adult rats via peripheral venous administration and this effect can be enhanced by HI insult. Also, this HI-induced permeation of β-NGF through BBB is more obvious in frontal cortex, hippocampus and cerebellum.
Collapse
Affiliation(s)
- Wenli Zhou
- Department of Neonatology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021 Jilin China
| | - Jiantao Zhang
- Department of Colorectal and Anal Surgery, The First Hospital of Jilin University, Changchun, 130021 Jilin China
| | - Guangming Wang
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, 130021 Jilin China
| | - Limian Ling
- Department of Colorectal and Anal Surgery, The First Hospital of Jilin University, Changchun, 130021 Jilin China
| | - Chaoying Yan
- Department of Neonatology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021 Jilin China
| |
Collapse
|
37
|
Corrigan F, Arulsamy A, Teng J, Collins-Praino LE. Pumping the Brakes: Neurotrophic Factors for the Prevention of Cognitive Impairment and Dementia after Traumatic Brain Injury. J Neurotrauma 2016; 34:971-986. [PMID: 27630018 DOI: 10.1089/neu.2016.4589] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) is the leading cause of disability and death worldwide, affecting as many as 54,000,000-60,000,000 people annually. TBI is associated with significant impairments in brain function, impacting cognitive, emotional, behavioral, and physical functioning. Although much previous research has focused on the impairment immediately following injury, TBI may have much longer-lasting consequences, including neuropsychiatric disorders and cognitive impairment. TBI, even mild brain injury, has also been recognized as a significant risk factor for the later development of dementia and Alzheimer's disease. Although the link between TBI and dementia is currently unknown, several proposed mechanisms have been put forward, including alterations in glucose metabolism, excitotoxicity, calcium influx, mitochondrial dysfunction, oxidative stress, and neuroinflammation. A treatment for the devastating long-term consequences of TBI is desperately needed. Unfortunately, however, no such treatment is currently available, making this a major area of unmet medical need. Increasing the level of neurotrophic factor expression in key brain areas may be one potential therapeutic strategy. Of the neurotrophic factors, granulocyte-colony stimulating factor (G-CSF) may be particularly effective for preventing the emergence of long-term complications of TBI, including dementia, because of its ability to reduce apoptosis, stimulate neurogenesis, and increase neuroplasticity.
Collapse
Affiliation(s)
- Frances Corrigan
- Translational Neuropathology Lab, Discipline of Anatomy and Pathology, School of Medicine, University of Adelaide , Adelaide, Australia
| | - Alina Arulsamy
- Translational Neuropathology Lab, Discipline of Anatomy and Pathology, School of Medicine, University of Adelaide , Adelaide, Australia
| | - Jason Teng
- Translational Neuropathology Lab, Discipline of Anatomy and Pathology, School of Medicine, University of Adelaide , Adelaide, Australia
| | - Lyndsey E Collins-Praino
- Translational Neuropathology Lab, Discipline of Anatomy and Pathology, School of Medicine, University of Adelaide , Adelaide, Australia
| |
Collapse
|
38
|
Selective and differential interactions of BNN27, a novel C17-spiroepoxy steroid derivative, with TrkA receptors, regulating neuronal survival and differentiation. Neuropharmacology 2016; 111:266-282. [PMID: 27618740 DOI: 10.1016/j.neuropharm.2016.09.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 08/11/2016] [Accepted: 09/07/2016] [Indexed: 12/21/2022]
Abstract
Nerve growth factor (NGF) holds a pivotal role in brain development and maintenance, been also involved in the pathophysiology of neurodegenerative diseases. Here, we provide evidence that a novel C17-spiroepoxy steroid derivative, BNN27, specifically interacts with and activates the TrkA receptor of NGF, inducing phosphorylation of TrkA tyrosine residues and down-stream neuronal survival-related kinase signaling. Additionally, BNN27 potentiates the efficacy of low levels of NGF, by facilitating its binding to the TrkA receptors and differentially inducing fast return of internalized TrkA receptors into neuronal cell membranes. Furthermore, BNN27 synergizes with NGF in promoting axonal outgrowth, effectively rescues from apoptosis NGF-dependent and TrkA positive sympathetic and sensory neurons, in vitro, ex vivo and in vivo in NGF null mice. Interestingly, BNN27 does not possess the hyperalgesic properties of NGF. BNN27 represents a lead molecule for the development of neuroprotective TrkA receptor agonists, with potential therapeutic applications in neurodegenerative diseases and in brain trauma.
Collapse
|
39
|
Bird SM, Sohrabi HR, Sutton TA, Weinborn M, Rainey-Smith SR, Brown B, Patterson L, Taddei K, Gupta V, Carruthers M, Lenzo N, Knuckey N, Bucks RS, Verdile G, Martins RN. Cerebral amyloid-β accumulation and deposition following traumatic brain injury--A narrative review and meta-analysis of animal studies. Neurosci Biobehav Rev 2016; 64:215-28. [PMID: 26899257 DOI: 10.1016/j.neubiorev.2016.01.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Accepted: 01/15/2016] [Indexed: 10/22/2022]
Abstract
Traumatic brain injury (TBI) increases the risk of neurodegenerative disorders many years post-injury. However, molecular mechanisms underlying the relationship between TBI and neurodegenerative diseases, such as Alzheimer's disease (AD), remain to be elucidated. Nevertheless, previous studies have demonstrated a link between TBI and increased amyloid-β (Aβ), a protein involved in AD pathogenesis. Here, we review animal studies that measured Aβ levels following TBI. In addition, from a pool of initially identified 1209 published papers, we examined data from 19 eligible animal model studies using a meta-analytic approach. We found an acute increase in cerebral Aβ levels ranging from 24h to one month following TBI (overall log OR=2.97 ± 0.40, p<0.001). These findings may contribute to further understanding the relationship between TBI and future dementia risk. The methodological inconsistencies of the studies discussed in this review suggest the need for improved and more standardised data collection and study design, in order to properly elucidate the role of TBI in the expression and accumulation of Aβ.
Collapse
Affiliation(s)
- Sabine M Bird
- School of Psychiatry and Clinical Neurosciences, University of Western Australia, 35 Stirling Hwy, Crawley, 6009 WA, Australia; Sir James McCusker Alzheimer's Disease Research Unit (Hollywood Private Hospital), 115 Monash Avenue, Nedlands, 6009 WA, Australia
| | - Hamid R Sohrabi
- School of Psychiatry and Clinical Neurosciences, University of Western Australia, 35 Stirling Hwy, Crawley, 6009 WA, Australia; Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, 6027 WA, Australia; Sir James McCusker Alzheimer's Disease Research Unit (Hollywood Private Hospital), 115 Monash Avenue, Nedlands, 6009 WA, Australia
| | - Thomas A Sutton
- School of Psychiatry and Clinical Neurosciences, University of Western Australia, 35 Stirling Hwy, Crawley, 6009 WA, Australia
| | - Michael Weinborn
- Sir James McCusker Alzheimer's Disease Research Unit (Hollywood Private Hospital), 115 Monash Avenue, Nedlands, 6009 WA, Australia; School of Psychology, University of Western Australia, 35 Stirling Hwy, Crawley, 6009 WA, Australia
| | - Stephanie R Rainey-Smith
- Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, 6027 WA, Australia; Sir James McCusker Alzheimer's Disease Research Unit (Hollywood Private Hospital), 115 Monash Avenue, Nedlands, 6009 WA, Australia
| | - Belinda Brown
- Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, 6027 WA, Australia; Sir James McCusker Alzheimer's Disease Research Unit (Hollywood Private Hospital), 115 Monash Avenue, Nedlands, 6009 WA, Australia
| | - Leigh Patterson
- Sir James McCusker Alzheimer's Disease Research Unit (Hollywood Private Hospital), 115 Monash Avenue, Nedlands, 6009 WA, Australia
| | - Kevin Taddei
- Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, 6027 WA, Australia; Sir James McCusker Alzheimer's Disease Research Unit (Hollywood Private Hospital), 115 Monash Avenue, Nedlands, 6009 WA, Australia
| | - Veer Gupta
- Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, 6027 WA, Australia; Sir James McCusker Alzheimer's Disease Research Unit (Hollywood Private Hospital), 115 Monash Avenue, Nedlands, 6009 WA, Australia
| | - Malcolm Carruthers
- Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, 6027 WA, Australia; Centre for Men's Health, 96 Harley Street, London, W1G 7HY, United Kingdom
| | - Nat Lenzo
- Oceanic Medical Imaging, Hollywood Medical Centre, 85 Monash Avenue, Nedlands, 6009 WA, Australia
| | - Neville Knuckey
- Centre for Neuromuscular and Neurological Disorders (CNND), University of Western Australia, 35 Stirling Hwy, Crawley, 6009 WA, Australia
| | - Romola S Bucks
- School of Psychology, University of Western Australia, 35 Stirling Hwy, Crawley, 6009 WA, Australia
| | - Giuseppe Verdile
- School of Psychiatry and Clinical Neurosciences, University of Western Australia, 35 Stirling Hwy, Crawley, 6009 WA, Australia; Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, 6027 WA, Australia; School of Biomedical Sciences, CHIRI Biosciences, Curtin University, Kent Street, Bentley, 6102 WA, Australia
| | - Ralph N Martins
- School of Psychiatry and Clinical Neurosciences, University of Western Australia, 35 Stirling Hwy, Crawley, 6009 WA, Australia; Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, 6027 WA, Australia; Sir James McCusker Alzheimer's Disease Research Unit (Hollywood Private Hospital), 115 Monash Avenue, Nedlands, 6009 WA, Australia.
| |
Collapse
|
40
|
Xie H, Xiao Z, Huang J. C6 Glioma-Secreted NGF and FGF2 Regulate Neuronal APP Processing Through Up-Regulation of ADAM10 and Down-Regulation of BACE1, Respectively. J Mol Neurosci 2015; 59:334-42. [PMID: 26614345 DOI: 10.1007/s12031-015-0690-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 11/19/2015] [Indexed: 01/05/2023]
Abstract
Excessive accumulation of amyloid-β (Aβ) caused by cleavage of amyloid precursor protein (APP) is thought to be the primary cause of Alzheimer's disease (AD). Two key enzymes ADAM10 and BACE1 are involved in the initial cleavage of APP, resulting in the onset of two pathways, the amyloidogenic pathway and the non-amyloidogenic pathway, respectively. Altering APP metabolism towards the non-amyloidogenic pathway is thought to reduce Aβ production. It has been reported that, in vivo, exogenous neurotrophic factors make APP apt to entering the non-amyloidogenic pathway. Since astrocytes secrete a battery of neurotrophic factors, we investigated the role of astrocyte-derived factors in the dynamics of Aβ generation in neural cells. Results show that C6 glioma cell-conditioned medium (GCM), obtained from cultured astrocyte-derived C6 glioma cells, inhibit Aβ1-42 production and shift APP processing towards the non-amyloidogenic pathway in APPswe-HEK293 cells. Such effect is attributed to two key APP cleavage enzymes, ADAM10 and BACE1. Two neurotrophic factors in the GCM, nerve growth factor and fibroblast growth factor 2, are responsible for the up-regulation of ADAM10 and down-regulation of BACE1, respectively. Our findings enhance our understanding of the relationship between astrocytes and Aβ generation, indicating that stimulation of astrocytic neurotrophic factors could slow AD progression.
Collapse
Affiliation(s)
- Huiping Xie
- College of Life Sciences, Wuhan University, Room 5105, Wuhan, Hubei, 430072, People's Republic of China
| | - Zhimin Xiao
- College of Life Sciences, Wuhan University, Room 5105, Wuhan, Hubei, 430072, People's Republic of China.,Sanofi, Cambridge, MA, 02142, USA
| | - Jian Huang
- College of Life Sciences, Wuhan University, Room 5105, Wuhan, Hubei, 430072, People's Republic of China.
| |
Collapse
|
41
|
Li Y, Zhang L, Kallakuri S, Cohen A, Cavanaugh JM. Correlation of mechanical impact responses and biomarker levels: A new model for biomarker evaluation in TBI. J Neurol Sci 2015; 359:280-6. [PMID: 26671128 DOI: 10.1016/j.jns.2015.08.035] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 08/19/2015] [Accepted: 08/20/2015] [Indexed: 12/19/2022]
Abstract
A modified Marmarou impact acceleration model was used to help screen biomarkers to assess brain injury severity. Anesthetized male Sprague-Dawley rats were subjected to a closed head injury from 1.25, 1.75 and 2.25 m drop heights. Linear and angular responses of the head were measured in vivo. 24h after impact, cerebrospinal fluid (CSF) and serum were collected. CSF and serum levels of phosphorylated neurofilament heavy (pNF-H), glial fibrillary acidic protein (GFAP), interleukin 6 (IL-6), and amyloid beta (Aβ) 1-42 were assessed by enzyme-linked immunosorbent assay (ELISA). Compared to controls, significantly higher CSF and serum pNF-H levels were observed in all impact groups, except between 1.25 m and control in serum. Furthermore, CSF and serum pNF-H levels were significantly different between the impact groups. For GFAP, both CSF and serum levels were significantly higher at 2.25 m compared to 1.75 m, 1.25 m and controls. There was no significant difference in CSF and serum GFAP levels between 1.75 m and 1.25 m, although both groups were significantly higher than control. TBI rats also showed significantly higher levels of IL-6 versus control in both CSF and serum, but no significant difference was observed between each impact group. Levels of Aβ were not significantly different between groups. Pearson's correlation analysis showed pNF-H and GFAP levels in CSF and serum had positive correlation with power (rate of impact energy), followed by average linear acceleration and surface righting (p<0.01), which were good predictors for traumatic axonal injury according to histologic assessment in our previous study, suggesting that they are directly related to the injury mechanism. The model used in this study showed a unique ability in elucidating the relationship between biomarker levels and severity of the mechanical trauma to the brain.
Collapse
Affiliation(s)
- Yan Li
- Department of Biomedical Engineering, Wayne State University, 818 W. Hancock Street, Detroit, MI 48201, United States
| | - Liying Zhang
- Department of Biomedical Engineering, Wayne State University, 818 W. Hancock Street, Detroit, MI 48201, United States.
| | - Srinivasu Kallakuri
- Department of Biomedical Engineering, Wayne State University, 818 W. Hancock Street, Detroit, MI 48201, United States
| | - Abigail Cohen
- Department of Biomedical Engineering, Wayne State University, 818 W. Hancock Street, Detroit, MI 48201, United States
| | - John M Cavanaugh
- Department of Biomedical Engineering, Wayne State University, 818 W. Hancock Street, Detroit, MI 48201, United States
| |
Collapse
|
42
|
Zhao Q, Hu J, Xiang J, Gu Y, Jin P, Hua F, Zhang Z, Liu Y, Zan K, Zhang Z, Zu J, Yang X, Shi H, Zhu J, Xu Y, Cui G, Ye X. Intranasal administration of human umbilical cord mesenchymal stem cells-conditioned medium enhances vascular remodeling after stroke. Brain Res 2015; 1624:489-496. [PMID: 26279113 DOI: 10.1016/j.brainres.2015.08.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 08/04/2015] [Accepted: 08/05/2015] [Indexed: 01/24/2023]
Abstract
Stem cell-based treatments have been reported to be a potential strategy for stroke. However, tumorigenic potential and low survival rates of transplanted cells could attenuate the efficacy of the stem cell-based treatments. The application of stem cell-condition medium (CM) may be a practicable approach to conquer these limitations. In this study, we investigated whether intranasal administration of human umbilical cord mesenchymal stem cells (hUCMSCs)-CM has the therapeutic effects in rats after stroke. Adult male rats were subjected to middle cerebral artery occlusion (MCAo) and were treated by intranasal routine with or without hUCMSCs-CM (1 ml/kg/d), starting 24h after MCAo and daily for 14 days. Neurological functional tests, blood brain barrier (BBB) leakage, were measured. Angiogenesis and angiogenic factor expression were measured by immunohistochemistry, and Western blot, respectively. hUCMSCs-CM treatment of stroke by intranasal routine starting 24h after MCAo in rats significantly enhances BBB functional integrity and promotes functional outcome but does not decrease lesion volume compared to rats in DMEM/F12 medium control group and saline control group. Treatment of ischemic rats with hUCMSCs-CM by intranasal routine also significantly decreases the levels of Ang2 and increases the levels of both Ang1 and Tie2 in the ischemic brain. To take together, increased expression of Ang1 and Tie2 and decreased expression of Ang2, induced by hUCMSCs-CM treatment, contribute to vascular remodeling in the ischemic brain which plays an important role in functional outcome after stroke.
Collapse
Affiliation(s)
- Qiuchen Zhao
- Department of Neurology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Jinxia Hu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Jie Xiang
- Department of Rehabilitation Medicine, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Yuming Gu
- Department of Interventional Radiology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Peisheng Jin
- Department of Plastic Surgery, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Fang Hua
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Zunsheng Zhang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Yonghai Liu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Kun Zan
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Zuohui Zhang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Jie Zu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Xinxin Yang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Hongjuan Shi
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Jienan Zhu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Yun Xu
- Department of Neurology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Guiyun Cui
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Xinchun Ye
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, China.
| |
Collapse
|
43
|
Liu S, Liu Y, Deng S, Guo A, Wang X, Shen G. Beneficial effects of hyperbaric oxygen on edema in rat hippocampus following traumatic brain injury. Exp Brain Res 2015; 233:3359-65. [PMID: 26267487 DOI: 10.1007/s00221-015-4405-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Accepted: 08/04/2015] [Indexed: 12/11/2022]
Abstract
Hyperbaric oxygen (HBO) therapy helps alleviate secondary injury following brain trauma [traumatic brain injury (TBI)], although the mechanisms remain unclear. In this study, we assessed recovery of post-TBI spatial learning and memory in rats using the Morris water maze (MWM) and measured changes in apparent diffusion coefficient in the hippocampus by diffusion-weighted imaging (DWI) to evaluate possible therapeutic effects of HBO on TBI-associated brain edema. DWIs were obtained 8, 24, 48 h, 7 days, and 14 days post-TBI. Daily HBO therapy significantly improved post-TBI MWM performance and reduced edema in the ipsilateral hippocampus, suggesting that the therapeutic efficacy of HBO is mediated, at least in part, by a reduction in brain edema.
Collapse
Affiliation(s)
- Su Liu
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, China
| | - Ying Liu
- Department of Pathology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, China
| | - Shukun Deng
- Department of Rehabilitation Medicine, Wuxi People's Hospital, 299 Qingyang Road, Wuxi, 214023, China
| | - Aisong Guo
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, China
| | - Xiubing Wang
- Department of Imaging, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, China
| | - Guangyu Shen
- Department of Rehabilitation Medicine, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, China.
| |
Collapse
|
44
|
Aly AEE, Waszczak BL. Intranasal gene delivery for treating Parkinson's disease: overcoming the blood-brain barrier. Expert Opin Drug Deliv 2015; 12:1923-41. [PMID: 26289676 DOI: 10.1517/17425247.2015.1069815] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Developing a disease-modifying gene therapy for Parkinson's disease (PD) has been a high priority for over a decade. However, due to the inability of large biomolecules to cross the blood-brain barrier (BBB), the only means of delivery to the brain has been intracerebral infusion. Intranasal administration offers a non-surgical means of bypassing the BBB to deliver neurotrophic factors, and the genes encoding them, directly to the brain. AREAS COVERED This review summarizes: i) evidence demonstrating intranasal delivery to the brain of a number of biomolecules having therapeutic potential for various CNS disorders; and ii) evidence demonstrating neuroprotective efficacy of a subset of biomolecules specifically for PD. The intersection of these two spheres represents the area of opportunity for development of new intranasal gene therapies for PD. To that end, our laboratory showed that intranasal administration of glial cell line-derived neurotrophic factor (GDNF), or plasmid DNA nanoparticles encoding GDNF, provides neuroprotection in a rat model of PD, and that the cells transfected by the nanoparticle vector are likely to be pericytes. EXPERT OPINION A number of genes encoding neurotrophic factors have therapeutic potential for PD, but few have been tested by the intranasal route and shown to be neuroprotective in a model of PD. Intranasal delivery provides a largely unexplored, promising approach for development of a non-invasive gene therapy for PD.
Collapse
Affiliation(s)
- Amirah E-E Aly
- a 1 Northeastern University, School of Pharmacy, Bouvé College of Health Sciences, Department of Pharmaceutical Sciences , Boston, MA 02115, USA
| | - Barbara L Waszczak
- b 2 Northeastern University, School of Pharmacy, Bouvé College of Health Sciences, Department of Pharmaceutical Sciences , Boston, MA 02115, USA +1 617 373 3312 ; +1 617 373 8886 ;
| |
Collapse
|
45
|
Zhang ZC, Luan F, Xie CY, Geng DD, Wang YY, Ma J. Low-frequency transcranial magnetic stimulation is beneficial for enhancing synaptic plasticity in the aging brain. Neural Regen Res 2015. [PMID: 26199608 PMCID: PMC4498353 DOI: 10.4103/1673-5374.158356] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
In the aging brain, cognitive function gradually declines and causes a progressive reduction in the structural and functional plasticity of the hippocampus. Transcranial magnetic stimulation is an emerging and novel neurological and psychiatric tool used to investigate the neurobiology of cognitive function. Recent studies have demonstrated that low-frequency transcranial magnetic stimulation (≤1 Hz) ameliorates synaptic plasticity and spatial cognitive deficits in learning-impaired mice. However, the mechanisms by which this treatment improves these deficits during normal aging are still unknown. Therefore, the current study investigated the effects of transcranial magnetic stimulation on the brain-derived neurotrophic factor signal pathway, synaptic protein markers, and spatial memory behavior in the hippocampus of normal aged mice. The study also investigated the downstream regulator, Fyn kinase, and the downstream effectors, synaptophysin and growth-associated protein 43 (both synaptic markers), to determine the possible mechanisms by which transcranial magnetic stimulation regulates cognitive capacity. Transcranial magnetic stimulation with low intensity (110% average resting motor threshold intensity, 1 Hz) increased mRNA and protein levels of brain-derived neurotrophic factor, tropomyosin receptor kinase B, and Fyn in the hippocampus of aged mice. The treatment also upregulated the mRNA and protein expression of synaptophysin and growth-associated protein 43 in the hippocampus of these mice. In conclusion, brain-derived neurotrophic factor signaling may play an important role in sustaining and regulating structural synaptic plasticity induced by transcranial magnetic stimulation in the hippocampus of aging mice, and Fyn may be critical during this regulation. These responses may change the structural plasticity of the aging hippocampus, thereby improving cognitive function.
Collapse
Affiliation(s)
- Zhan-Chi Zhang
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Feng Luan
- Department of Otorhinolaryngology, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Chun-Yan Xie
- Second Surgical Department, Qinghe Public Hospital of Hebei Province, Xingtai, Hebei Province, China
| | - Dan-Dan Geng
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Yan-Yong Wang
- Department of Neurology, First Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China ; Hebei Key Laboratory for Brain Aging and Cognitive Neuroscience, Shijiazhuang, Hebei Province, China
| | - Jun Ma
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei Province, China ; Hebei Key Laboratory for Brain Aging and Cognitive Neuroscience, Shijiazhuang, Hebei Province, China
| |
Collapse
|
46
|
Xiong Y, Zhang Y, Mahmood A, Chopp M. Investigational agents for treatment of traumatic brain injury. Expert Opin Investig Drugs 2015; 24:743-60. [PMID: 25727893 PMCID: PMC4433440 DOI: 10.1517/13543784.2015.1021919] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
INTRODUCTION Traumatic brain injury (TBI) is a major cause of death and disability worldwide. To date, there are no pharmacologic agents proven to improve outcomes from TBI because all the Phase III clinical trials in TBI have failed. Thus, there is a compelling need to develop treatments for TBI. AREAS COVERED The following article provides an overview of select cell-based and pharmacological therapies under early development for the treatment of TBI. These therapies seek to enhance cognitive and neurological functional recovery through neuroprotective and neurorestorative strategies. EXPERT OPINION TBI elicits both complex degenerative and regenerative tissue responses in the brain. TBI can lead to cognitive, behavioral, and motor deficits. Although numerous promising neuroprotective treatment options have emerged from preclinical studies that mainly target the lesion, translation of preclinical effective neuroprotective drugs to clinical trials has proven challenging. Accumulating evidence indicates that the mammalian brain has a significant, albeit limited, capacity for both structural and functional plasticity, as well as regeneration essential for spontaneous functional recovery after injury. A new therapeutic approach is to stimulate neurovascular remodeling by enhancing angiogenesis, neurogenesis, oligodendrogenesis, and axonal sprouting, which in concert, may improve neurological functional recovery after TBI.
Collapse
Affiliation(s)
- Ye Xiong
- Henry Ford Hospital, Department of Neurosurgery , Education and Research Building, Room 3096, 2799 West Grand Boulevard, Detroit, MI 48202 , USA +1 313 916 4743 ; +1 313 916 9855 ;
| | | | | | | |
Collapse
|
47
|
Nanoparticle-mediated growth factor delivery systems: A new way to treat Alzheimer's disease. J Control Release 2015; 206:187-205. [DOI: 10.1016/j.jconrel.2015.03.024] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 03/19/2015] [Accepted: 03/20/2015] [Indexed: 01/03/2023]
|
48
|
Young J, Pionk T, Hiatt I, Geeck K, Smith JS. Environmental enrichment aides in functional recovery following unilateral controlled cortical impact of the forelimb sensorimotor area however intranasal administration of nerve growth factor does not. Brain Res Bull 2015; 115:17-22. [PMID: 25889001 DOI: 10.1016/j.brainresbull.2015.04.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 04/02/2015] [Accepted: 04/08/2015] [Indexed: 11/27/2022]
Abstract
PURPOSE An injury to the forelimb sensorimotor cortex results in the impairment of motor function in animals. Recent research has suggested that intranasal administration of nerve growth factor (NGF), a protein naturally found in the brain, and placement into enriched environments (EE) improves motor and cognitive function after traumatic brain injury (TBI). The purpose of this study was to determine whether NGF, EE, or the combination of both was beneficial in the recovery of motor function following TBI. RESULTS Uninjured animals had fewer foot faults than injured animals, displaying a lesion effect. Injured animals housed in EE were shown to have fewer foot faults whether or not they received NGF. Injured animals also displayed an increased reliance on the non-impaired limb further validating a lesion effect. CONCLUSION EE is an effective treatment on the recovery of motor function after a TBI. Intranasal administration of NGF was found to not be an effective treatment for functional motor recovery after a TBI.
Collapse
Affiliation(s)
- Jennica Young
- The Brain Research Laboratory, Saginaw Valley State University, University Center, MI, USA.
| | - Timothy Pionk
- The Brain Research Laboratory, Saginaw Valley State University, University Center, MI, USA.
| | - Ivy Hiatt
- The Brain Research Laboratory, Saginaw Valley State University, University Center, MI, USA.
| | - Katalin Geeck
- The Brain Research Laboratory, Saginaw Valley State University, University Center, MI, USA.
| | - Jeffrey S Smith
- The Brain Research Laboratory, Saginaw Valley State University, University Center, MI, USA.
| |
Collapse
|
49
|
Chen J, Li S, Sun W, Li J. Anti-diabetes drug pioglitazone ameliorates synaptic defects in AD transgenic mice by inhibiting cyclin-dependent kinase5 activity. PLoS One 2015; 10:e0123864. [PMID: 25875370 PMCID: PMC4396851 DOI: 10.1371/journal.pone.0123864] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 02/23/2015] [Indexed: 01/21/2023] Open
Abstract
Cyclin-dependent kinase 5 (Cdk5) is a serine/threonine kinase that is activated by the neuron specific activators p35/p39 and plays many important roles in neuronal development. However, aberrant activation of Cdk5 is believed to be associated with the pathogenesis of several neurodegenerative diseases, including Alzheimer's disease (AD) and Parkinson's disease (PD). Here in the present study, enhanced Cdk5 activity was observed in mouse models of AD; whereas soluble amyloid-β oligomers (Aβ), which contribute to synaptic failures during AD pathogenesis, induced Cdk5 hyperactivation in cultured hippocampal neurons. Inhibition of Cdk5 activity by pharmacological or genetic approaches reversed dendritic spine loss caused by soluble amyloid-β oligomers (Aβ) treatment. Interestingly, we found that the anti-diabetes drug pioglitazone could inhibit Cdk5 activity by decreasing p35 protein level. More importantly, pioglitazone treatment corrected long-term potentiation (LTP) deficit caused by Aβ exposure in cultured slices and pioglitazone administration rescued impaired LTP and spatial memory in AD mouse models. Taken together, our study describes an unanticipated role of pioglitazone in alleviating AD and reveals a potential therapeutic drug for AD curing.
Collapse
Affiliation(s)
- Jinan Chen
- Department of Neurology, Jiangning Hospital, Nanjing Medical University, Jiangsu, China
- * E-mail: (JL); (JC)
| | - Shenghua Li
- Department of Neurology, Jiangning Hospital, Nanjing Medical University, Jiangsu, China
| | - Wenshan Sun
- Department of Neurology, Jiangning Hospital, Nanjing Medical University, Jiangsu, China
| | - Junrong Li
- Department of Neurology, Jiangning Hospital, Nanjing Medical University, Jiangsu, China
- * E-mail: (JL); (JC)
| |
Collapse
|
50
|
Ivanov A, Tukhbatova G, Salozhin S, Markevich V. NGF but not BDNF overexpression protects hippocampal LTP from beta-amyloid-induced impairment. Neuroscience 2015; 289:114-22. [DOI: 10.1016/j.neuroscience.2014.12.063] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 12/22/2014] [Accepted: 12/23/2014] [Indexed: 01/19/2023]
|