1
|
Hrybouski S, Das SR, Xie L, Brown CA, Flamporis M, Lane J, Nasrallah IM, Detre JA, Yushkevich PA, Wolk DA. BOLD amplitude correlates of preclinical Alzheimer's disease. Neurobiol Aging 2025; 150:157-171. [PMID: 40138942 DOI: 10.1016/j.neurobiolaging.2025.03.007] [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: 11/02/2024] [Revised: 03/08/2025] [Accepted: 03/12/2025] [Indexed: 03/29/2025]
Abstract
Alzheimer's disease (AD) is characterized by a long preclinical stage during which molecular markers of amyloid beta and tau pathology rise, but there is minimal neurodegeneration or cognitive decline. Previous literature suggests that measures of brain function might be more sensitive to neuropathologic burden during the preclinical stage of AD than conventional measures of macrostructure, such as cortical thickness. Among studies that used resting-state functional Magnetic Resonance Imaging (fMRI) acquisitions with Blood Oxygenation Level Dependent (BOLD) contrast, most employed connectivity-based analytic approaches. Consequently, little is known about the effects of amyloid and tau pathology on amplitude of intrinsic BOLD signal fluctuations. To address this knowledge gap, we characterized the effects of preclinical and prodromal AD on the amplitude of low-frequency fluctuations (ALFF) of the BOLD signal both at the whole-brain level and at a more granular level focused on subregions of the medial temporal lobe. We observed reduced ALFF in both preclinical and prodromal AD. In preclinical AD, amyloid positivity was associated with a spatially diffuse ALFF reduction in the frontal, medial parietal, and lateral temporal association cortices. In contrast, tau pathology was negatively associated with ALFF in the entorhinal cortex. These ALFF effects were observed in the absence of observable macrostructural changes in preclinical AD and remained after adjusting for structural atrophy in prodromal AD, indicating that ALFF offers additional sensitivity to early disease processes beyond what is provided by traditional structural imaging biomarkers of neurodegeneration. We conclude that ALFF may be a promising imaging-based biomarker in preclinical AD.
Collapse
Affiliation(s)
- Stanislau Hrybouski
- Penn Image Computing and Science Laboratory (PICSL), University of Pennsylvania, Philadelphia, PA, United States; Department of Radiology, University of Pennsylvania, Philadelphia, PA, United States.
| | - Sandhitsu R Das
- Penn Image Computing and Science Laboratory (PICSL), University of Pennsylvania, Philadelphia, PA, United States; Department of Neurology, University of Pennsylvania, Philadelphia, PA, United States; Penn Memory Center, University of Pennsylvania, Philadelphia, PA, United States; Penn Alzheimer's Disease Research Center, University of Pennsylvania, Philadelphia, PA, United States
| | - Long Xie
- Penn Image Computing and Science Laboratory (PICSL), University of Pennsylvania, Philadelphia, PA, United States; Department of Radiology, University of Pennsylvania, Philadelphia, PA, United States
| | - Christopher A Brown
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, United States; Penn Alzheimer's Disease Research Center, University of Pennsylvania, Philadelphia, PA, United States
| | - Melissa Flamporis
- Penn Memory Center, University of Pennsylvania, Philadelphia, PA, United States
| | - Jacqueline Lane
- Penn Memory Center, University of Pennsylvania, Philadelphia, PA, United States
| | - Ilya M Nasrallah
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, United States; Penn Alzheimer's Disease Research Center, University of Pennsylvania, Philadelphia, PA, United States
| | - John A Detre
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, United States; Department of Neurology, University of Pennsylvania, Philadelphia, PA, United States
| | - Paul A Yushkevich
- Penn Image Computing and Science Laboratory (PICSL), University of Pennsylvania, Philadelphia, PA, United States; Department of Radiology, University of Pennsylvania, Philadelphia, PA, United States; Penn Alzheimer's Disease Research Center, University of Pennsylvania, Philadelphia, PA, United States
| | - David A Wolk
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, United States; Penn Memory Center, University of Pennsylvania, Philadelphia, PA, United States; Penn Alzheimer's Disease Research Center, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
2
|
Sarkar S, Porel P, Kosey S, Aran KR. Unraveling the role of CGRP in neurological diseases: a comprehensive exploration to pathological mechanisms and therapeutic implications. Mol Biol Rep 2025; 52:436. [PMID: 40299101 DOI: 10.1007/s11033-025-10542-y] [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: 03/11/2025] [Accepted: 04/24/2025] [Indexed: 04/30/2025]
Abstract
Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), Multiple sclerosis (MS), Amyotrophic lateral sclerosis (ALS), and Spinal muscular atrophy (SMA) are neurodegenerative diseases (NDDs) characterized by progressive neuronal degeneration. Recent studies provide compelling information regarding the contribution of Calcitonin Gene-Related Peptide (CGRP), a potent neuropeptide, in regulating neuroinflammation, vasodilation, and neuronal survival in these disorders. This review systematically delves into the multidimensional aspects of CGRP as both a neuroprotective agent and a neurotoxic factor in NDDs. The neuroprotective effects of CGRP include suppression of inflammation, regulation of intracellular signaling pathways, and promotion of neuronal growth and survival. However, under pathological conditions, its overexpression or dysregulation is associated with oxidative stress, excitotoxicity, and neuronal death. The therapeutic use of CGRP and its receptor antagonists in migraine provides substantial evidence for CGRP's therapeutic potential, which can be further explored for the management of NDDs. However, since the bidirectional nature of CGRP effects is evident, it is crucial to gain an accurate insight into its mechanisms to target only the neuropeptide's beneficial effects while completely avoiding the undesired consequences. Further studies should focus on understanding the context-dependent activity of CGRP in the hope of designing targeted therapy for NDDs, which could gradually transform the current pharmacological management of NDDs.
Collapse
Affiliation(s)
- Sampriti Sarkar
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Pratyush Porel
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Sourabh Kosey
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Khadga Raj Aran
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India.
| |
Collapse
|
3
|
Müller E, Schoberwalter T, Mader K, Seitz JM, Kopp A, Baranowsky A, Keller J. The Biological Effects of Magnesium-Based Implants on the Skeleton and Their Clinical Implications in Orthopedic Trauma Surgery. Biomater Res 2024; 28:0122. [PMID: 39717475 PMCID: PMC11665827 DOI: 10.34133/bmr.0122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 11/27/2024] [Accepted: 11/27/2024] [Indexed: 12/25/2024] Open
Abstract
Magnesium (Mg)-based implants have evolved as a promising innovation in orthopedic trauma surgery, with the potential to revolutionize the treatment of bone diseases, including osteoporotic fractures and bone defects. Available clinical studies mostly show excellent patient outcomes of resorbable Mg-based implants, without the need for subsequent implant removal. However, the occurrence of radiolucent zones around Mg-based implants seems to be a noticeable drawback for a more widespread clinical use. Mechanistically, both in vivo and in vitro studies demonstrated beneficial effects on the formation of new bone, a unique characteristic of Mg-based implants. In this regard, Mg has been shown to exert pleiotropic functions on osteogenic differentiation and migration of osteoblasts and their precursors. Additionally, collective evidence suggests that Mg-based implants promote angiogenesis in newly formed bone and exert immunomodulatory effects in the bone microenvironment. Likewise, Mg-based implants and their degradation products were shown to inhibit bone resorption by impairing osteoclastogenesis. The purpose of this review is to provide a state-of-the-art summary of the clinical and basic science evidence regarding the performance of currently used Mg-based implants. In addition to the status of in vivo and in vitro research and clinical applications, future challenges and perspectives of Mg-based orthopedic implants are discussed.
Collapse
Affiliation(s)
- Elena Müller
- Department of Trauma and Orthopedic Surgery,
University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Till Schoberwalter
- Department of Trauma and Orthopedic Surgery,
University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Konrad Mader
- Department of Trauma and Orthopedic Surgery,
University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | | | - Alexander Kopp
- Medical Magnesium GmbH, 52068 Aachen, Germany
- Meotec GmbH, 52068 Aachen, Germany
| | - Anke Baranowsky
- Department of Trauma and Orthopedic Surgery,
University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Johannes Keller
- Department of Trauma and Orthopedic Surgery,
University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| |
Collapse
|
4
|
Lin K, Stiles J, Tambo W, Ajmal E, Piao Q, Powell K, Li C. Bimodal functions of calcitonin gene-related peptide in the brain. Life Sci 2024; 359:123177. [PMID: 39486618 DOI: 10.1016/j.lfs.2024.123177] [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: 08/23/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 11/04/2024]
Abstract
AIMS Calcitonin gene-related peptide (CGRP) is a pluripotent neuropeptide crucial for maintaining vascular homeostasis, yet its full therapeutic potential remains incompletely exploited. Within the brain, CGRP demonstrates a distinct bimodal effect, contributing to neuroprotection in ischemic conditions while inducing neuronal sensitization and inflammation in non-ischemic settings. Despite extensive research on CGRP, the absence of a definitive determinant for this observed dichotomy has limited its potential for therapeutic applications in the brain. This review examines the effects of CGRP in both physiological and pathological conditions, aiming to identify a unifying factor that could enhance its therapeutic applicability. MATERIALS AND METHODS This comprehensive literature review analyzes the molecular pathways associated with CGRP and the specific cellular responses observed in these contexts. Additionally, the review investigates the psychological implications of CGRP in relation to cerebral perfusion levels, aiming to elucidate its underlying factors. KEY FINDINGS Reviewing the literature reveals that, elevated levels of CGRP in non-ischemic conditions exert detrimental effects on brain function, while they confer protective effects in the context of ischemia. These encompass anti-oxidative, anti-inflammatory, anti-apoptotic, and angiogenic properties, along with behavioral normalization. Current findings indicate promising therapeutic avenues for CGRP beyond the acute phases of cerebral injury, extending to neurodegenerative and psychological disorders associated with cerebral hypoperfusion, as well as chronic recovery following acute cerebral injuries. SIGNIFICANCE Improved understanding of CGRP's bimodal properties, alongside advancements in CGRP delivery methodologies and brain ischemia detection technologies, paves the way for realizing its untapped potential and broad therapeutic benefits in diverse pathological conditions.
Collapse
Affiliation(s)
- Kanheng Lin
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY, USA; Emory University, Atlanta, GA, USA
| | - Jacob Stiles
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY, USA; The College of William & Mary, Williamsburg, VA, USA
| | - Willians Tambo
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY, USA; Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA
| | - Erum Ajmal
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY, USA; Division of Neurosurgery, SUNY Downstate College of Medicine, Brooklyn, NY, USA
| | - Quanyu Piao
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Keren Powell
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Chunyan Li
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY, USA; Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA; Department of Neurosurgery, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA.
| |
Collapse
|
5
|
Alexoudi A, Donadio V, Karageorgiou E. The potential role of CGRP in synuclein-associated neurodegenerative disorders. Front Neurosci 2024; 18:1479830. [PMID: 39568667 PMCID: PMC11576422 DOI: 10.3389/fnins.2024.1479830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 10/21/2024] [Indexed: 11/22/2024] Open
Abstract
In this hypothesis article, the potential clinicopathological associations of Calcitonin Gene Related Peptide (CGRP) with the development of synuclein-associated neurodegenerative disorders (SAND) are discussed. The presence of α-syn and CGRP in the CNS and the ENS and the intricate role of CGRP and its related pathways in inflammation, apoptosis, metabolism, neuromodulation, and brain-gut communication are analyzed. Since this hypothesis is confirmed, modulating CGRP-potential related pathways may lead to novel disease-modifying therapies.
Collapse
Affiliation(s)
| | - Vincenzo Donadio
- IRCCS Institute of Neurological Sciences of Bologna, Bologna, Italy
| | | |
Collapse
|
6
|
Hrybouski S, Das SR, Xie L, Brown CA, Flamporis M, Lane J, Nasrallah IM, Detre JA, Yushkevich PA, Wolk DA. BOLD Amplitude Correlates of Preclinical Alzheimer's Disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.27.24316243. [PMID: 39574853 PMCID: PMC11581098 DOI: 10.1101/2024.10.27.24316243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Alzheimer's disease (AD) is characterized by a long preclinical stage during which molecular markers of amyloid beta and tau pathology rise, but there is minimal neurodegeneration or cognitive decline. Previous literature suggests that measures of brain function might be more sensitive to neuropathologic burden during the preclinical stage of AD than conventional measures of macrostructure, such as cortical thickness. However, among studies that used resting-state functional Magnetic Resonance Imaging (fMRI) acquisitions with Blood Oxygenation Level Dependent (BOLD) contrast, most employed connectivity-based analytic approaches, which discard information about the amplitude of spontaneous brain activity. Consequently, little is known about the effects of amyloid and tau pathology on BOLD amplitude. To address this knowledge gap, we characterized the effects of preclinical and prodromal AD on the amplitude of low-frequency fluctuations (ALFF) of the BOLD signal both at the whole-brain level and, at a more granular level, focused on subregions of the medial temporal lobe. We observed reduced ALFF in both preclinical and prodromal AD. In preclinical AD, amyloid positivity was associated with a spatially diffuse ALFF reduction in the frontal, medial parietal, and lateral temporal association cortices, while tau pathology was negatively associated with ALFF in the entorhinal cortex. These ALFF effects were observed in the absence of observable macrostructural changes in preclinical AD and remained after adjusting for structural atrophy in prodromal AD, indicating that ALFF offers additional sensitivity to early disease processes beyond what is provided by traditional structural imaging biomarkers of neurodegeneration. We conclude that ALFF may be a promising imaging-based biomarker for assessing the effects of amyloid-clearing immunotherapies in preclinical AD.
Collapse
|
7
|
Shahouzehi B, Masoumi-Ardakani Y, Fallah H, Aminizadeh S. Evaluation of the effect of Exercise Trainings and CGRP receptor antagonist (BIBN 4096) on mitochondrial dynamic in the hippocampus of male Wistar rats. Neurosci Lett 2024; 828:137752. [PMID: 38552868 DOI: 10.1016/j.neulet.2024.137752] [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: 12/25/2023] [Revised: 03/19/2024] [Accepted: 03/26/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND Exercise training showed beneficial effects on brain. The purpose of the present study is to evaluate the effect of six weeks of high-intensity interval training (HIIT) and Endurance training (ET) with calcitonin gene-related peptide (CGRP) receptor antagonist on the expression of genes involved in mitochondrial dynamics and apoptosis in hippocampal tissue of male Wistar rats. METHODS In this study, forty-two healthymale Wistar rats (8-week) were randomly divided into 6 groups (n = 7) as follow; 1) Control; 2) HIIT which performed 6 weeks of HIIT; 3) ET which performed 6 weeks of endurance training; 4) CGRPi received 10 mg/kg CGRP receptor antagonist every day at the last 2 weeks; 5) CGRPi-HIIT performed HIIT and received CGRP receptor antagonist; 6) CGRPi-ET performed ET and received CGRP receptor antagonist. Real-time PCR (2-ΔΔCT) and western blotting were employedto measure the expression of genes and protein, respectively. RESULTS HIIT and ET significantly increased Bcl-2, Pgc-1α, Sirt3, and Nrf-1 gene expression in the hippocampal tissue (p < 0.05, p < 0.01, p < 0.01, and p < 0.001, respectively). ET-CGRPi and HIIT-CGRPi significantly increased Sirt3, Pgc-1α, and Nrf-1 gene expression compared to the control group (p < 0.05, p < 0.01, and p < 0.05, respectively). CONCLUSION ET and HIIT-induced physiological alterations in the hippocampus. In fact, this modulation showed protective properties in the hippocampusvia up regulation of Bcl-2, Pgc-1α, Nrf-1, and Sirt3 gene expression. CGRPi did not cause gene or protein changes harmful to mitochondrial dynamic balance and apoptosis in the hippocampus of rats.
Collapse
Affiliation(s)
- Beydolah Shahouzehi
- Cardiovascular Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran; Gastroenterology and Hepatology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Yaser Masoumi-Ardakani
- Physiology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Hossein Fallah
- Applied Cellular and Molecular Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Soheil Aminizadeh
- Department of Physiology and Pharmacology, Afzalipour School of Medicine, and Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
8
|
Swinford CG, Risacher SL, Vosmeier A, Deardorff R, Chumin EJ, Dzemidzic M, Wu YC, Gao S, McDonald BC, Yoder KK, Unverzagt FW, Wang S, Farlow MR, Brosch JR, Clark DG, Apostolova LG, Sims J, Wang DJ, Saykin AJ. Amyloid and tau pathology are associated with cerebral blood flow in a mixed sample of nondemented older adults with and without vascular risk factors for Alzheimer's disease. Neurobiol Aging 2023; 130:103-113. [PMID: 37499587 PMCID: PMC10529454 DOI: 10.1016/j.neurobiolaging.2023.06.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 05/30/2023] [Accepted: 06/21/2023] [Indexed: 07/29/2023]
Abstract
Identification of biomarkers for the early stages of Alzheimer's disease (AD) is an imperative step in developing effective treatments. Cerebral blood flow (CBF) is a potential early biomarker for AD; generally, older adults with AD have decreased CBF compared to normally aging peers. CBF deviates as the disease process and symptoms progress. However, further characterization of the relationships between CBF and AD risk factors and pathologies is still needed. We assessed the relationships between CBF quantified by arterial spin-labeled magnetic resonance imaging, hypertension, APOEε4, and tau and amyloid positron emission tomography in 77 older adults: cognitively normal, subjective cognitive decline, and mild cognitive impairment. Tau and amyloid aggregation were related to altered CBF, and some of these relationships were dependent on hypertension or APOEε4 status. Our findings suggest a complex relationship between risk factors, AD pathologies, and CBF that warrants future studies of CBF as a potential early biomarker for AD.
Collapse
Affiliation(s)
- Cecily G Swinford
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana Alzheimer's Disease Research Center, Indianapolis, IN, USA
| | - Shannon L Risacher
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana Alzheimer's Disease Research Center, Indianapolis, IN, USA
| | - Aaron Vosmeier
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana Alzheimer's Disease Research Center, Indianapolis, IN, USA
| | - Rachael Deardorff
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana Alzheimer's Disease Research Center, Indianapolis, IN, USA
| | - Evgeny J Chumin
- Indiana Alzheimer's Disease Research Center, Indianapolis, IN, USA; Indiana University Network Science Institute, Bloomington, IN, USA; Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Mario Dzemidzic
- Indiana Alzheimer's Disease Research Center, Indianapolis, IN, USA; Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yu-Chien Wu
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana Alzheimer's Disease Research Center, Indianapolis, IN, USA
| | - Sujuan Gao
- Indiana Alzheimer's Disease Research Center, Indianapolis, IN, USA; Department of Biostatistics and Health Data Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Brenna C McDonald
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana Alzheimer's Disease Research Center, Indianapolis, IN, USA; Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Karmen K Yoder
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana Alzheimer's Disease Research Center, Indianapolis, IN, USA
| | - Frederick W Unverzagt
- Indiana Alzheimer's Disease Research Center, Indianapolis, IN, USA; Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sophia Wang
- Indiana Alzheimer's Disease Research Center, Indianapolis, IN, USA; Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Martin R Farlow
- Indiana Alzheimer's Disease Research Center, Indianapolis, IN, USA; Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jared R Brosch
- Indiana Alzheimer's Disease Research Center, Indianapolis, IN, USA; Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - David G Clark
- Indiana Alzheimer's Disease Research Center, Indianapolis, IN, USA; Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Liana G Apostolova
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana Alzheimer's Disease Research Center, Indianapolis, IN, USA; Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana University Network Science Institute, Bloomington, IN, USA
| | - Justin Sims
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Danny J Wang
- Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana Alzheimer's Disease Research Center, Indianapolis, IN, USA; Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA; Indiana University Network Science Institute, Bloomington, IN, USA.
| |
Collapse
|
9
|
Overeem LH, Raffaelli B, Fleischmann R, Süße M, Vogelgesang A, Maceski AM, Papadopoulou A, Ruprecht K, Su W, Koch M, Siebert A, Arkuszewski M, Tenenbaum N, Kuhle J, Reuter U. Serum tau protein elevation in migraine: a cross-sectional case-control study. J Headache Pain 2023; 24:130. [PMID: 37726712 PMCID: PMC10507851 DOI: 10.1186/s10194-023-01663-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/01/2023] [Indexed: 09/21/2023] Open
Abstract
BACKGROUND Migraine is a disorder associated with neuropeptide release, pain and inflammation. Tau protein has recently been linked to inflammatory diseases and can be influenced by neuropeptides such as CGRP, a key neurotransmitter in migraine. Here, we report serum concentrations of total-tau protein in migraine patients and healthy controls. METHODS In this cross-sectional study, interictal blood samples from n = 92 patients with episodic migraine (EM), n = 93 patients with chronic migraine (CM), and n = 42 healthy matched controls (HC) were studied. We assessed serum total-tau protein (t-tau) and for comparison neurofilament light chain protein (NfL), glial fibrillary acidic protein (GFAP), and ubiquitin carboxy-terminal hydrolase L (UCH-L1) concentrations using the Neurology 4-plex kit, on a single molecule array HD-X Analyzer (Quanterix Corp Lexington, MA). Matched serum/cerebrospinal fluid (CSF) samples were used for post-hoc evaluations of a central nervous system (CNS) source of relevant findings. We applied non-parametric tests to compare groups and assess correlations. RESULTS Serum t-tau concentrations were elevated in EM [0.320 (0.204 to 0.466) pg/mL] and CM [0.304 (0.158 to 0.406) pg/mL] patients compared to HC [0.200 (0.114 to 0.288) pg/mL] (p = 0.002 vs. EM; p = 0.025 vs. CM). EM with aura [0.291 (0.184 to 0.486 pg/mL); p = 0.013] and EM without aura [0.332 (0.234 to 0.449) pg/mL; p = 0.008] patients had higher t-tau levels than HC but did not differ between each other. Subgroup analysis of CM with/without preventive treatment revealed elevated t-tau levels compared to HC only in the non-prevention group [0.322 (0.181 to 0.463) pg/mL; p = 0.009]. T-tau was elevated in serum (p = 0.028) but not in cerebrospinal fluid (p = 0.760). In contrast to t-tau, all proteins associated with cell damage (NfL, GFAP, and UCH-L1), did not differ between groups. DISCUSSION Migraine is associated with t-tau elevation in serum but not in the CSF. Our clinical study identifies t-tau as a new target for migraine research.
Collapse
Affiliation(s)
- Lucas Hendrik Overeem
- Department of Neurology With Experimental Neurology, Charité - Universitätsmedizin Berlin, Charitéplatz 1, Berlin, 10117, Germany
- Doctoral Program, International Graduate Program Medical Neurosciences, Humboldt Graduate School, Berlin, 10117, Germany
| | - Bianca Raffaelli
- Department of Neurology With Experimental Neurology, Charité - Universitätsmedizin Berlin, Charitéplatz 1, Berlin, 10117, Germany
- Clinician Scientist Program, Berlin Institute of Health (BIH), Berlin, 10117, Germany
| | - Robert Fleischmann
- Department of Neurology, Universitätsmedizin Greifswald, Greifswald, 17475, Germany
| | - Marie Süße
- Department of Neurology, Universitätsmedizin Greifswald, Greifswald, 17475, Germany
| | - Antje Vogelgesang
- Department of Neurology, Universitätsmedizin Greifswald, Greifswald, 17475, Germany
| | - Aleksandra Maleska Maceski
- Department of Neurology, University Hospital and University of Basel, Basel, 4051, Switzerland
- Multiple Sclerosis Centre and Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), Departments of Biomedicine and Clinical Research, University Hospital and University of Basel, Basel, 4051, Switzerland
| | - Athina Papadopoulou
- Department of Neurology, University Hospital and University of Basel, Basel, 4051, Switzerland
- Multiple Sclerosis Centre and Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), Departments of Biomedicine and Clinical Research, University Hospital and University of Basel, Basel, 4051, Switzerland
| | - Klemens Ruprecht
- Department of Neurology With Experimental Neurology, Charité - Universitätsmedizin Berlin, Charitéplatz 1, Berlin, 10117, Germany
| | - Wendy Su
- Novartis Pharma AG, Basel, 4056, Switzerland
| | - Mirja Koch
- Novartis Pharma AG, Basel, 4056, Switzerland
| | - Anke Siebert
- Department of Neurology With Experimental Neurology, Charité - Universitätsmedizin Berlin, Charitéplatz 1, Berlin, 10117, Germany
| | | | - Nadia Tenenbaum
- EMD Serono Research and Development Institute, New York, NY, USA
| | - Jens Kuhle
- Department of Neurology, University Hospital and University of Basel, Basel, 4051, Switzerland
- Multiple Sclerosis Centre and Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), Departments of Biomedicine and Clinical Research, University Hospital and University of Basel, Basel, 4051, Switzerland
| | - Uwe Reuter
- Department of Neurology With Experimental Neurology, Charité - Universitätsmedizin Berlin, Charitéplatz 1, Berlin, 10117, Germany.
- Department of Neurology, Universitätsmedizin Greifswald, Greifswald, 17475, Germany.
| |
Collapse
|
10
|
Xiong J, Wang Z, Bai J, Cheng K, Liu Q, Ni J. Calcitonin gene-related peptide: a potential protective agent in cerebral ischemia-reperfusion injury. Front Neurosci 2023; 17:1184766. [PMID: 37529236 PMCID: PMC10387546 DOI: 10.3389/fnins.2023.1184766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 06/29/2023] [Indexed: 08/03/2023] Open
Abstract
Ischemic stroke is the most common type of cerebrovascular disease with high disability and mortality rates, which severely burdens patients, their families, and society. At present, thrombolytic therapy is mainly used for the treatment of ischemic strokes. Even though it can achieve a good effect, thrombolytic recanalization can cause reperfusion injury. Calcitonin gene-related peptide (CGRP) is a neuropeptide that plays a neuroprotective role in the process of ischemia-reperfusion injury. By combining with its specific receptors, CGRP can induce vasodilation of local cerebral ischemia by directly activating the cAMP-PKA pathway in vascular smooth muscle cells and by indirectly activating the NO-cGMP pathway in an endothelial cell-dependent manner,thus rapidly increasing ischemic local blood flow together with reperfusion. CGRP, as a key effector molecule of neurogenic inflammation, can reduce the activation of microglia, downregulates Th1 classical inflammation, and reduce the production of TNF-α, IL-2, and IFN-γ and the innate immune response of macrophages, leading to the reduction of inflammatory factors. CGRP can reduce the overexpression of the aquaporin-4 (AQP-4) protein and its mRNA in the cerebral ischemic junction, and play a role in reducing cerebral edema. CGRP can protect endothelial cells from angiotensin II by reducing the production of oxidants and protecting antioxidant defense. Furthermore, CGRP-upregulated eNOS can further induce VEGF expression, which then promotes the survival and angiogenesis of vascular endothelial cells. CGRP can also reduce apoptosis by promoting the expression of Bcl-2 and inhibiting the expression of caspase-3. These effects suggest that CGRP can reduce brain injury and repair damaged nerve function. In this review, we focused on the role of CGRP in cerebral ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Jie Xiong
- Department of Rehabilitation, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Zhiyong Wang
- Department of Rehabilitation, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Junhui Bai
- Department of Rehabilitation, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Keling Cheng
- Department of Rehabilitation, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Qicai Liu
- Department of Reproductive Medicine Centre, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Jun Ni
- Department of Rehabilitation, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
11
|
Red Ginger Oil Affects COX-2 and NMDAR Expression During Inflammatory- or Neuropathy-Induced Chronic Pain in Mice. Jundishapur J Nat Pharm Prod 2021. [DOI: 10.5812/jjnpp.112353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background: Chronic pain treatment until now is still challenging because of its complex pathopgysiology. Previously, red ginger oil (RGO) reduced pain behavior in a mouse model of chronic pain, but the mechanisms were unclear. Objectives: This study examined the effect of RGO on cyclooxygenase (COX)-2 and the N-methyl-D-aspartate receptor (NMDAR) using inflammatory- or neuropathy-induced chronic pain in mice. Methods: Red ginger was distilled with composition 1:2 using water. The acute toxicity of RGO was evaluated using OECD guidelines 423. Chronic pain was induced in 48 mice by either (1) intraplantar injection of complete Freund’s adjuvant (CFA) (inflammatory group) or (2) partial sciatic nerve ligation (PSNL) (neuropathy group). After seven days, mice were randomly divided into sham, CFA/PSNL, or RGO (at doses of 100, 200, 400, or 600 mg/kg) treatment groups. Treatments were given orally once daily until day 14. On day 15, mice were euthanized, and the brains and spinal cords were removed and fixed in 10% formalin. Hyperalgesia behavior was evaluated using hot plate test. Spinal cord morphology was analyzed via hematoxylin and eosin staining. COX-2 and NMDAR expressions were evaluated by immunohistochemistry. Results: RGO treatment improved spinal cord morphology after the induction of chronic pain. RGO at 600 mg/kg also reduced COX-2 expression in the spinal cord and brain, and reduced NMDAR2B in the spinal cord. However, RGO at 600 mg/kg increased NMDAR2A expression in the spinal cord. Conclusions: RGO administration diminished hyperalgesia in chronic pain models through inhibition of COX-2 and NMDAR2B.
Collapse
|
12
|
Acute administration of metformin prior to cardiac ischemia/reperfusion injury protects brain injury. Eur J Pharmacol 2020; 885:173418. [DOI: 10.1016/j.ejphar.2020.173418] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/23/2020] [Accepted: 07/23/2020] [Indexed: 11/19/2022]
|
13
|
Borkum JM. CGRP and Brain Functioning: Cautions for Migraine Treatment. Headache 2019; 59:1339-1357. [DOI: 10.1111/head.13591] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Jonathan M. Borkum
- Department of Psychology University of Maine Orono ME USA
- Health Psych Maine Waterville ME USA
| |
Collapse
|
14
|
Acquarone E, Argyrousi EK, van den Berg M, Gulisano W, Fà M, Staniszewski A, Calcagno E, Zuccarello E, D’Adamio L, Deng SX, Puzzo D, Arancio O, Fiorito J. Synaptic and memory dysfunction induced by tau oligomers is rescued by up-regulation of the nitric oxide cascade. Mol Neurodegener 2019; 14:26. [PMID: 31248451 PMCID: PMC6598340 DOI: 10.1186/s13024-019-0326-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 06/05/2019] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Soluble aggregates of oligomeric forms of tau protein (oTau) have been associated with impairment of synaptic plasticity and memory in Alzheimer's disease. However, the molecular mechanisms underlying the synaptic and memory dysfunction induced by elevation of oTau are still unknown. METHODS This work used a combination of biochemical, electrophysiological and behavioral techniques. Biochemical methods included analysis of phosphorylation of the cAMP-responsive element binding (CREB) protein, a transcriptional factor involved in memory, histone acetylation, and expression immediate early genes c-Fos and Arc. Electrophysiological methods included assessment of long-term potentiation (LTP), a type of synaptic plasticity thought to underlie memory formation. Behavioral studies investigated both short-term spatial memory and associative memory. These phenomena were examined following oTau elevation. RESULTS Levels of phospho-CREB, histone 3 acetylation at lysine 27, and immediate early genes c-Fos and Arc, were found to be reduced after oTau elevation during memory formation. These findings led us to explore whether up-regulation of various components of the nitric oxide (NO) signaling pathway impinging onto CREB is capable of rescuing oTau-induced impairment of plasticity, memory, and CREB phosphorylation. The increase of NO levels protected against oTau-induced impairment of LTP through activation of soluble guanylyl cyclase. Similarly, the elevation of cGMP levels and stimulation of the cGMP-dependent protein kinases (PKG) re-established normal LTP after exposure to oTau. Pharmacological inhibition of cGMP degradation through inhibition of phosphodiesterase 5 (PDE5), rescued oTau-induced LTP reduction. These findings could be extrapolated to memory because PKG activation and PDE5 inhibition rescued oTau-induced memory impairment. Finally, PDE5 inhibition re-established normal elevation of CREB phosphorylation and cGMP levels after memory induction in the presence of oTau. CONCLUSIONS Up-regulation of CREB activation through agents acting on the NO cascade might be beneficial against tau-induced synaptic and memory dysfunctions.
Collapse
Affiliation(s)
- Erica Acquarone
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, 630 West 168th Street, P&S 12-420D, New York, NY 10032 USA
- DiMi Department of Internal Medicine and Medical Specialties, University of Genoa, 16132 Genoa, Italy
| | - Elentina K. Argyrousi
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, 630 West 168th Street, P&S 12-420D, New York, NY 10032 USA
- Faculty of Psychology and Neuroscience, Maastricht University, 6229 Maastricht, Netherlands
| | - Manon van den Berg
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, 630 West 168th Street, P&S 12-420D, New York, NY 10032 USA
- Faculty of Psychology and Neuroscience, Maastricht University, 6229 Maastricht, Netherlands
| | - Walter Gulisano
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, 95125 Catania, Italy
| | - Mauro Fà
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, 630 West 168th Street, P&S 12-420D, New York, NY 10032 USA
| | - Agnieszka Staniszewski
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, 630 West 168th Street, P&S 12-420D, New York, NY 10032 USA
| | - Elisa Calcagno
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, 630 West 168th Street, P&S 12-420D, New York, NY 10032 USA
- Department of Experimental Medicine, Section of General Pathology, School of Medical and Pharmaceutical Sciences, University of Genoa, 16132 Genoa, Italy
| | - Elisa Zuccarello
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, 630 West 168th Street, P&S 12-420D, New York, NY 10032 USA
| | - Luciano D’Adamio
- Department of Pharmacology, Physiology and Neuroscience, Rutgers University, Newark, NJ USA
| | - Shi-Xian Deng
- Department of Medicine, Columbia University, New York, NY 10032 USA
| | - Daniela Puzzo
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, 95125 Catania, Italy
- Oasi Research Institute-IRCCS, 94018 Troina, Italy
| | - Ottavio Arancio
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, 630 West 168th Street, P&S 12-420D, New York, NY 10032 USA
- Department of Medicine, Columbia University, New York, NY 10032 USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032 USA
| | - Jole Fiorito
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, 630 West 168th Street, P&S 12-420D, New York, NY 10032 USA
- Department of Life Sciences, New York Institute of Technology, Northern Boulevard P.O. Box 8000, Theobald Science Center, room 425, Old Westbury, NY 11568 USA
| |
Collapse
|
15
|
Sah SK, Samuel VP, Dahiya S, Singh Y, Gilhotra RM, Gupta G, Mishra A, Sharma RK, Kumar GS, SreeHarsha N, Chellappan DK, Dua K. A contemporary biological pathway of islet amyloid polypeptide for the management of diabetic dementia. Chem Biol Interact 2019; 306:117-122. [PMID: 31004596 DOI: 10.1016/j.cbi.2019.04.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/11/2019] [Accepted: 04/17/2019] [Indexed: 01/07/2023]
Abstract
Major challenges of dealing elder patients with diabetes mellitus (DM) are the individualization of consideration in persons with various comorbid types of conditions. In spite of the fact that microvascular and macrovascular problems associated with DM are well documented, there is only a few numbers of reports viewing different conditions, for example, cognitive dysfunction. Cognitive dysfunction is of specific significance due to its effect on self-care and quality of life. All in all, the etiology of cognitive dysfunction in the maturing populace is probably going to be the grouping of ischemic and degenerative pathology. It is likewise trusted that Hyperglycemia is engaged with the system of DM-related cognitive dysfunction. At present, it isn't certain in the case of enhancing glycemic control or utilizing therapeutic agents can enhance the risk of cognitive decay. Amylin was later characterized as an amyloidogenic peptide, confined from a beta cell tumor and called islet amyloid polypeptide (IAPP), and after that, amylin. Conversely, we investigate the beneficial role and hypothesizing the mechanism of amylin related expanding the level and activation of CGRP receptor to enhance the cognition declination amid diabetic dementia.
Collapse
Affiliation(s)
| | - Vijaya Paul Samuel
- Department of Anatomy, RAK College of Medicine, RAK Medical and Health Sciences, University, Ras Al Khaimah, United Arab Emirates
| | - Sunita Dahiya
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico, USA
| | - Yogendar Singh
- Department of Pharmaceutical Sciences, Mahatma Gandhi College of Pharmaceutical Sciences, Sitapura, Jaipur, India
| | - Ritu M Gilhotra
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jagatpura, Jaipur, India
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jagatpura, Jaipur, India.
| | - Anurag Mishra
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jagatpura, Jaipur, India
| | - Rakesh Kumar Sharma
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jagatpura, Jaipur, India
| | | | - Nagaraja SreeHarsha
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur, 57000, Malaysia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney (UTS), Ultimo, NSW 2007, Australia; Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI) & School of Biomedical Sciences and Pharmacy, The University of Newcastle (UoN), Callaghan, NSW 2308, Australia.
| |
Collapse
|
16
|
Neuroprotection of Cytisine Against Cerebral Ischemia–Reperfusion Injury in Mice by Regulating NR2B-ERK/CREB Signal Pathway. Neurochem Res 2018; 43:1575-1586. [DOI: 10.1007/s11064-018-2572-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 05/09/2018] [Accepted: 06/08/2018] [Indexed: 10/28/2022]
|
17
|
Shi Z, Zhu L, Li T, Tang X, Xiang Y, Han X, Xia L, Zeng L, Nie J, Huang Y, Tsang CK, Wang Y, Lei Z, Xu Z, So KF, Ruan Y. Neuroprotective Mechanisms of Lycium barbarum Polysaccharides Against Ischemic Insults by Regulating NR2B and NR2A Containing NMDA Receptor Signaling Pathways. Front Cell Neurosci 2017; 11:288. [PMID: 29021742 PMCID: PMC5623723 DOI: 10.3389/fncel.2017.00288] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 09/04/2017] [Indexed: 01/03/2023] Open
Abstract
Glutamate excitotoxicity plays an important role in neuronal death after ischemia. However, all clinical trials using glutamate receptor inhibitors have failed. This may be related to the evidence that activation of different subunit of NMDA receptor will induce different effects. Many studies have shown that activation of the intrasynaptic NR2A subunit will stimulate survival signaling pathways, whereas upregulation of extrasynaptic NR2B will trigger apoptotic pathways. A Lycium barbarum polysaccharide (LBP) is a mixed compound extracted from Lycium barbarum fruit. Recent studies have shown that LBP protects neurons against ischemic injury by anti-oxidative effects. Here we first reported that the effect of LBP against ischemic injury can be achieved by regulating NR2B and NR2A signaling pathways. By in vivo study, we found LBP substantially reduced CA1 neurons from death after transient global ischemia and ameliorated memory deficit in ischemic rats. By in vitro study, we further confirmed that LBP increased the viability of primary cultured cortical neurons when exposed to oxygen-glucose deprivation (OGD) for 4 h. Importantly, we found that LBP antagonized increase in expression of major proteins in the NR2B signal pathway including NR2B, nNOS, Bcl-2-associated death promoter (BAD), cytochrome C (cytC) and cleaved caspase-3, and also reduced ROS level, calcium influx and mitochondrial permeability after 4 h OGD. In addition, LBP prevented the downregulation in the expression of NR2A, pAkt and pCREB, which are important cell survival pathway components. Furthermore, LBP attenuated the effects of a NR2B co-agonist and NR2A inhibitor on cell mortality under OGD conditions. Taken together, our results demonstrated that LBP is neuroprotective against ischemic injury by its dual roles in activation of NR2A and inhibition of NR2B signaling pathways, which suggests that LBP may be a superior therapeutic candidate for targeting glutamate excitotoxicity for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Zhongshan Shi
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Department of Anatomy, Jinan University School of Medicine, Guangzhou, China.,Ministry of Education CNS Regeneration International Collaborative Laboratory, Jinan University, Guangzhou, China
| | - Lihui Zhu
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Department of Anatomy, Jinan University School of Medicine, Guangzhou, China.,Ministry of Education CNS Regeneration International Collaborative Laboratory, Jinan University, Guangzhou, China
| | - Tingting Li
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Xiaoya Tang
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Yonghui Xiang
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Xinjia Han
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Luoxing Xia
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Ling Zeng
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Junhua Nie
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Yongxia Huang
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China.,Department of Ophthalmology and State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China
| | - Chi Kwan Tsang
- Clinical Neuroscience Institute, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Ying Wang
- Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, China
| | - Zhigang Lei
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Zaocheng Xu
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Kwok-Fai So
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Ministry of Education CNS Regeneration International Collaborative Laboratory, Jinan University, Guangzhou, China.,Department of Ophthalmology and State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China
| | - Yiwen Ruan
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Department of Anatomy, Jinan University School of Medicine, Guangzhou, China.,Ministry of Education CNS Regeneration International Collaborative Laboratory, Jinan University, Guangzhou, China
| |
Collapse
|
18
|
Lu CX, Qiu T, Liu ZF, Su L, Cheng B. Calcitonin gene-related peptide has protective effect on brain injury induced by heat stroke in rats. Exp Ther Med 2017; 14:4935-4941. [PMID: 29201197 PMCID: PMC5704302 DOI: 10.3892/etm.2017.5126] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 06/22/2017] [Indexed: 11/13/2022] Open
Abstract
Heat stroke often leads to multiple organ dysfunction syndrome (MODS) with a neurological morbidity of 30%. Current studies suggested that pathophysiological responses to heat stroke may be due to a systemic inflammatory response syndrome and a series of peptidergic nerve reactions. The mechanisms underlying the high neurological morbidity in heat stroke have remained largely elusive. In recent years, calcitonin gene-related peptide (CGRP) has been considered to have a positive role in central nervous system injury. The present study investigated the influence of CGRP on brain injury induced by heat stroke. A rat model of heat stroke was established in a pre-warmed artificial climate chamber with a temperature of 35.5±0.5°C and a relative humidity of 60±5%. The rectal core temperature (Tc) was monitored. Heat stress was halted at a Tc of no more than 41°C A bolus injection of CGRP was administered to each rat in the HS+CGRP group and a bolus injection of CGRP8-37 was administered to each rat in the HS+CGRP8-37 group after heat stress. After 2 h, electroencephalograms were recorded and the pathological morphology of brain tissue as well as brain cell apoptosis and caspase-3 protein levels in the brain were measured. The EEG of rats in the HS+CGRP group was characterized by a short- to long-term α-wave and low-voltage β-waves as well as a large amount of intermittent δ- and θ-waves. Compared with the HS group, the θ-wave decreased and the α-wave increased significantly (P<0.05). Slight pathological damage of nerve cells appeared in the HS+CGRP group. Greater damage was observed in HS+CGRP8-37 group with neural cell shrinkage, volume reduction, nuclear pyknosis, disappearance of part of the nuclear membrane and cell necrosis. In the HS+CGRP group, apoptotic cells and caspase-3 protein in the brain were significantly decreased when compared with those in the HS group (P<0.05), while they were significantly increased in the HS+CGRP8-37 group (P<0.05 vs. HS group). The results of the present study reflected that CGRP has a protective effect on early-stage brain injury induced by heat stroke in rats.
Collapse
Affiliation(s)
- Cheng-Xiang Lu
- Department of Intensive Care Unit, Affiliated General Hospital of Guangzhou Military Command of Southern Medical University, Guangzhou, Guangdong 510010, P.R. China.,Department of Intensive Care Unit, Zhongshan Hospital Xiamen University, Xiamen, Fujian 361004, P.R. China
| | - Ting Qiu
- Department of Neurology, Zhongshan Hospital Xiamen University, Xiamen, Fujian 361004, P.R. China
| | - Zhi-Feng Liu
- Department of Intensive Care Unit, General Hospital of Guangzhou Military Command, Guangzhou, Guangdong 510010, P.R. China
| | - Lei Su
- Department of Intensive Care Unit, Affiliated General Hospital of Guangzhou Military Command of Southern Medical University, Guangzhou, Guangdong 510010, P.R. China
| | - Biao Cheng
- Department of Plastic Surgery, Affiliated General Hospital of Guangzhou Military Command of Southern Medical University, Guangzhou, Guangdong 510010, P.R. China
| |
Collapse
|
19
|
Sorby-Adams AJ, Marcoionni AM, Dempsey ER, Woenig JA, Turner RJ. The Role of Neurogenic Inflammation in Blood-Brain Barrier Disruption and Development of Cerebral Oedema Following Acute Central Nervous System (CNS) Injury. Int J Mol Sci 2017; 18:E1788. [PMID: 28817088 PMCID: PMC5578176 DOI: 10.3390/ijms18081788] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 08/07/2017] [Accepted: 08/15/2017] [Indexed: 12/13/2022] Open
Abstract
Acute central nervous system (CNS) injury, encompassing traumatic brain injury (TBI) and stroke, accounts for a significant burden of morbidity and mortality worldwide, largely attributable to the development of cerebral oedema and elevated intracranial pressure (ICP). Despite this, clinical treatments are limited and new therapies are urgently required to improve patient outcomes and survival. Originally characterised in peripheral tissues, such as the skin and lungs as a neurally-elicited inflammatory process that contributes to increased microvascular permeability and tissue swelling, neurogenic inflammation has now been described in acute injury to the brain where it may play a key role in the secondary injury cascades that evolve following both TBI and stroke. In particular, release of the neuropeptides substance P (SP) and calcitonin gene-related peptide (CGRP) appear to be critically involved. In particular, increased SP expression is observed in perivascular tissue following acute CNS injury, with the magnitude of SP release being related to both the frequency and degree of the insult. SP release is associated with profound blood-brain barrier disruption and the subsequent development of vasogenic oedema, as well as neuronal injury and poor functional outcomes. Inhibition of SP through use of a neurokinin 1 (NK1) antagonist is highly beneficial following both TBI and ischaemic stroke in pre-clinical models. The role of CGRP is more unclear, especially with respect to TBI, with both elevations and reductions in CGRP levels reported following trauma. However, a beneficial role has been delineated in stroke, given its potent vasodilatory effects. Thus, modulating neuropeptides represents a novel therapeutic target in the treatment of cerebral oedema following acute CNS injury.
Collapse
Affiliation(s)
- Annabel J Sorby-Adams
- Adelaide Medical School and Adelaide Centre for Neuroscience Research, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide SA 5005, Australia.
| | - Amanda M Marcoionni
- Adelaide Medical School and Adelaide Centre for Neuroscience Research, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide SA 5005, Australia.
| | - Eden R Dempsey
- Adelaide Medical School and Adelaide Centre for Neuroscience Research, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide SA 5005, Australia.
| | - Joshua A Woenig
- Adelaide Medical School and Adelaide Centre for Neuroscience Research, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide SA 5005, Australia.
| | - Renée J Turner
- Adelaide Medical School and Adelaide Centre for Neuroscience Research, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide SA 5005, Australia.
| |
Collapse
|
20
|
Singh Y, Gupta G, Shrivastava B, Dahiya R, Tiwari J, Ashwathanarayana M, Sharma RK, Agrawal M, Mishra A, Dua K. Calcitonin gene-related peptide (CGRP): A novel target for Alzheimer's disease. CNS Neurosci Ther 2017; 23:457-461. [PMID: 28417590 PMCID: PMC6492742 DOI: 10.1111/cns.12696] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 03/11/2017] [Accepted: 03/17/2017] [Indexed: 12/22/2022] Open
Abstract
Alzheimer's disease (AD) is leading cause of death among older characterized by neurofibrillary tangles, oxidative stress, progressive neuronal deficits, and increased levels of amyloid-β (Aβ) peptides. Cholinergic treatment could be the best suitable physiological therapy for AD. Calcitonin gene-related peptide (CGRP) is a thirty-seven-amino acid regulatory neuropeptide resulting from different merging of the CGRP gene, which also includes adrenomedullin, amylin, calcitonin, intermedin, and calcitonin receptor-stimulating peptide. It is a proof for a CGRP receptor within nucleus accumbens of brain that is different from either the CGRP1 or CGRP2 receptor in which it demonstrates similar high-affinity binding for salmon calcitonin, CGRP, and amylin, a possession which is not shared by any extra CGRP receptors. Binding of CGRP to its receptor increases activated cAMP-dependent pkA and PI3 kinase, resulting in N-terminal fragments that are shown to exert complex inhibitory as well facilitator actions on nAChRs. Fragments such as CGRP1-4, CGRP1-5, and CGRP1-6 rapidly as well as reversibly improve agonist sensitivity of nAChRs without straight stimulating those receptors and produce the Ca2+ -induced intracellular Ca2+ mobilization. Renin-angiotensin-aldosterone system (RAAS)-activated angiotensin-type (AT4) receptor is also beneficial in AD. It has been suggested that exogenous administration of CGRP inhibits infiltration of macrophages and expression of various inflammatory mediators such as NFkB, IL-1b, TNF-α, iNOS, matrix metalloproteinase (MMP)-9, and cell adhesion molecules like intercellular adhesion molecule (ICAM)-1 which attenuates consequence of inflammation in AD. Donepezil, a ChEI, inhibits acetylcholinesterase and produces angiogenesis and neurogenesis, in the dentate gyrus of the hippocampus of WT mice after donepezil administration. However, none of the results discovered in CGRP-knockout mice and WT mice exposed to practical denervation. Therefore, selective agonists of CGRP receptors may become the potential candidates for treatment of AD.
Collapse
Affiliation(s)
- Yogendra Singh
- School of PharmacyJaipur National UniversityJagatpuraJaipurIndia
| | - Gaurav Gupta
- School of PharmacyJaipur National UniversityJagatpuraJaipurIndia
- School of Medicine and Public HealthUniversity of NewcastleNewcastleNSWAustralia
| | | | - Rajiv Dahiya
- Laboratory of Peptide Research and DevelopmentSchool of Pharmacy, The University of the West IndiesSt. Augustine, Trinidad & TobagoWest Indies
| | - Juhi Tiwari
- School of PharmacyJaipur National UniversityJagatpuraJaipurIndia
| | | | | | - Mohit Agrawal
- School of pharmacySuresh Gyan Vihar UniversityJaipurIndia
| | - Anurag Mishra
- School of pharmacySuresh Gyan Vihar UniversityJaipurIndia
| | - Kamal Dua
- Discipline of PharmacyGraduate School of HealthUniversity of Technology SydneySydneyNSWAustralia
- School of Biomedical Sciences and PharmacyUniversity of NewcastleNewcastleNSWAustralia
- School of Pharmaceutical SciencesShoolini UniversitySolanHimachal PradeshIndia
| |
Collapse
|
21
|
Curcumin confers neuroprotection against alcohol-induced hippocampal neurodegeneration via CREB-BDNF pathway in rats. Biomed Pharmacother 2017; 87:721-740. [DOI: 10.1016/j.biopha.2016.12.020] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 11/19/2016] [Accepted: 12/05/2016] [Indexed: 02/08/2023] Open
|
22
|
Zhang Y, Xu J, Ruan YC, Yu MK, O'Laughlin M, Wise H, Chen D, Tian L, Shi D, Wang J, Chen S, Feng JQ, Chow DHK, Xie X, Zheng L, Huang L, Huang S, Leung K, Lu N, Zhao L, Li H, Zhao D, Guo X, Chan K, Witte F, Chan HC, Zheng Y, Qin L. Implant-derived magnesium induces local neuronal production of CGRP to improve bone-fracture healing in rats. Nat Med 2016; 22:1160-1169. [PMID: 27571347 PMCID: PMC5293535 DOI: 10.1038/nm.4162] [Citation(s) in RCA: 585] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 07/13/2016] [Indexed: 12/13/2022]
Abstract
Orthopedic implants containing biodegradable magnesium have been used for fracture repair with considerable efficacy; however, the underlying mechanisms by which these implants improve fracture healing remain elusive. Here we show the formation of abundant new bone at peripheral cortical sites after intramedullary implantation of a pin containing ultrapure magnesium into the intact distal femur in rats. This response was accompanied by substantial increases of neuronal calcitonin gene-related polypeptide-α (CGRP) in both the peripheral cortex of the femur and the ipsilateral dorsal root ganglia (DRG). Surgical removal of the periosteum, capsaicin denervation of sensory nerves or knockdown in vivo of the CGRP-receptor-encoding genes Calcrl or Ramp1 substantially reversed the magnesium-induced osteogenesis that we observed in this model. Overexpression of these genes, however, enhanced magnesium-induced osteogenesis. We further found that an elevation of extracellular magnesium induces magnesium transporter 1 (MAGT1)-dependent and transient receptor potential cation channel, subfamily M, member 7 (TRPM7)-dependent magnesium entry, as well as an increase in intracellular adenosine triphosphate (ATP) and the accumulation of terminal synaptic vesicles in isolated rat DRG neurons. In isolated rat periosteum-derived stem cells, CGRP induces CALCRL- and RAMP1-dependent activation of cAMP-responsive element binding protein 1 (CREB1) and SP7 (also known as osterix), and thus enhances osteogenic differentiation of these stem cells. Furthermore, we have developed an innovative, magnesium-containing intramedullary nail that facilitates femur fracture repair in rats with ovariectomy-induced osteoporosis. Taken together, these findings reveal a previously undefined role of magnesium in promoting CGRP-mediated osteogenic differentiation, which suggests the therapeutic potential of this ion in orthopedics.
Collapse
Affiliation(s)
- Yifeng Zhang
- Musculoskeletal Research Laboratory, Department of Orthopedics &Traumatology, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopedics &Traumatology, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Ye Chun Ruan
- Epithelial Cell Biology Research Centre, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Mei Kuen Yu
- Epithelial Cell Biology Research Centre, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Micheal O'Laughlin
- Musculoskeletal Research Laboratory, Department of Orthopedics &Traumatology, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Helen Wise
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Di Chen
- Department of Biochemistry, Rush University, Chicago, USA
| | - Li Tian
- Musculoskeletal Research Laboratory, Department of Orthopedics &Traumatology, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Dufang Shi
- Musculoskeletal Research Laboratory, Department of Orthopedics &Traumatology, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Jiali Wang
- Musculoskeletal Research Laboratory, Department of Orthopedics &Traumatology, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Sihui Chen
- Musculoskeletal Research Laboratory, Department of Orthopedics &Traumatology, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Jian Q Feng
- Department of Biomedical Sciences, Baylor College of Dentistry, Texas A&M Health Science Center, Dallas, Texas, USA
| | - Dick Ho Kiu Chow
- Musculoskeletal Research Laboratory, Department of Orthopedics &Traumatology, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Xinhui Xie
- Musculoskeletal Research Laboratory, Department of Orthopedics &Traumatology, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Lizhen Zheng
- Musculoskeletal Research Laboratory, Department of Orthopedics &Traumatology, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Le Huang
- Musculoskeletal Research Laboratory, Department of Orthopedics &Traumatology, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Shuo Huang
- Musculoskeletal Research Laboratory, Department of Orthopedics &Traumatology, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Kwoksui Leung
- Musculoskeletal Research Laboratory, Department of Orthopedics &Traumatology, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Na Lu
- Division of Life Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, PR China
| | - Lan Zhao
- Department of Biochemistry, Rush University, Chicago, USA
| | - Huafang Li
- Musculoskeletal Research Laboratory, Department of Orthopedics &Traumatology, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Dewei Zhao
- Department of Orthopedics, Dalian University Zhongshan Hospital, Dalian, PR China
| | - Xia Guo
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong, PR China
| | - Kaiming Chan
- Musculoskeletal Research Laboratory, Department of Orthopedics &Traumatology, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Frank Witte
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Hsiao Chang Chan
- Epithelial Cell Biology Research Centre, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Yufeng Zheng
- Department of Materials Science and Engineering, College of Engineering, Peking University, Beijing, PR China
| | - Ling Qin
- Musculoskeletal Research Laboratory, Department of Orthopedics &Traumatology, The Chinese University of Hong Kong, Hong Kong, PR China
- Translational Medicine Research &Development Center, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Shenzhen, PR China
| |
Collapse
|
23
|
Han F, Zhou D, Yin X, Sun Z, Han J, Ye L, Zhao W, Zhang Y, Wang Z, Zheng L. Paeoniflorin protects diabetic mice against myocardial ischemic injury via the transient receptor potential vanilloid 1/calcitonin gene-related peptide pathway. Cell Biosci 2016; 6:37. [PMID: 27252827 PMCID: PMC4888521 DOI: 10.1186/s13578-016-0085-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 02/21/2016] [Indexed: 12/13/2022] Open
Abstract
Background Diabetes mellitus has multiple complications including neuropathy and increases cardiovascular events. Paeoniflorin (PF), a monoterpene glycoside, plays an essential role in neuroprotection and ischemic heart disease. In this study, we aimed to investigate the hypothesis that PF protects mice with diabetes mellitus against myocardial ischemic injury, and determine its associated mechanisms. Results Myocardial infarction (MI) was generated in the streptozotocin-mediated diabetic mice, which were pretreated with either vehicle or PF, respectively. Myocardial infarct size, myocardial enzyme, cardiac function, circulating calcitonin gene-related peptide (CGRP) concentration, histological analysis and the expression of associated molecules were determined and compared among different experimental groups. Compared to diabetic hearts pretreated with vehicle, hearts pretreated with PF exhibited less tissue damage and better CGRP concentration in serum when subjected to myocardial ischemia. Transient receptor potential vanilloid 1(TRPV1) gene knockout attenuated PF-mediated cardioprotection. Moreover, a specific Ca2+/calmodulin-dependent protein kinase (CaMK) inhibitor, KN-93, increased tissue damage and decreased CGRP activity in serum. Meanwhile, pretreated with PF increased the phosphorylation of cAMP response element binding protein (CREB). Conclusions Taken together, these findings demonstrate that PF protects diabetic mice against MI at least partially via the TRPV1/CaMK/CREB/CGRP signaling pathway.
Collapse
Affiliation(s)
- Fei Han
- Department of Cardiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310003 China
| | - Dongchen Zhou
- Department of Cardiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310003 China
| | - Xiang Yin
- Department of Cardiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310003 China
| | - Zewei Sun
- Department of Cardiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310003 China
| | - Jie Han
- Department of Cardiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310003 China
| | - Lifang Ye
- Department of Cardiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310003 China
| | - Wengting Zhao
- Department of Cardiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310003 China
| | - Yuanyuan Zhang
- Department of Cardiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310003 China
| | - Zhen Wang
- Department of Cardiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310003 China
| | - Liangrong Zheng
- Department of Cardiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310003 China
| |
Collapse
|
24
|
Yang SI, Yuan Y, Jiao S, Luo QI, Yu J. Calcitonin gene-related peptide protects rats from cerebral ischemia/reperfusion injury via a mechanism of action in the MAPK pathway. Biomed Rep 2016; 4:699-703. [PMID: 27284409 PMCID: PMC4887836 DOI: 10.3892/br.2016.658] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 04/14/2016] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate the protective function and underlying mechanism of calcitonin gene-related peptide (CGRP) on cerebral ischemia/reperfusion damage in rats. Adult male Wistar rats were selected for the establishment of an ischemia/reperfusion injury model through the application of a middle cerebral artery occlusion. Animals were randomly divided into 6 groups of 24 animals. Drugs were administered according to the design of each group; animals were administered CGRP, CGRP8-37, PD98059 and SB20358. The neurobehavioral scores of the rat cerebral ischemia model in each group were calculated. The infarction range of the rat brain tissues was observed by 2,3,5-triphenyltetrazolium chloride staining. The expression levels of three proteins, phosphorylated c-Jun N-terminal kinase (JNK)/JNK, phosphorylated extracellular signal-regulated protein kinase (ERK)/ERK and p-p38/p38, in the mitogen-activated protein kinase (MAPK) pathway in the brain tissues was detected by western blotting. The results showed that CGRP could improve the neurobehavioral function of the ischemic rats and reduce the infarction range. Western blotting results confirmed that the function of the CGRP was mediated mainly through the reduction of the JNK and p38 phosphorylation and the promotion of ERK phosphorylation. Therefore, the present study confirmed that an increase in the exogenous CRGP could effectively improve ischemia/reperfusion injury of the brain tissue. The mechanisms of action were achieved through a reduction in JNK and p38 phosphorylation and an increase in ERL phosphorylation in the MAPK pathway. These mechanisms were interdependent.
Collapse
Affiliation(s)
- S I Yang
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China; Department of Pediatric Neurology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yongjie Yuan
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Shan Jiao
- Department of Endodontics, Stomatological Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Q I Luo
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jinlu Yu
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
25
|
Liu YR, Li PW, Suo JJ, Sun Y, Zhang BA, Lu H, Zhu HC, Zhang GB. Catalpol provides protective effects against cerebral ischaemia/reperfusion injury in gerbils. J Pharm Pharmacol 2014; 66:1265-70. [PMID: 24720795 DOI: 10.1111/jphp.12261] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 03/02/2014] [Indexed: 12/27/2022]
Abstract
Abstract
Objectives
To investigate the protective effect of catalpol on cerebral ischaemia/reperfusion (CI/R) injury in gerbils and further explore the underlying mechanism.
Methods
A gerbil model of CI/R was prepared by bilateral common carotid occlusion for 10 min followed by 6 h reperfusion. Catalpol (5, 10 or 20 mg/kg per day) was injected intraperitoneally for 3 days before the carotid occlusion. Stroke index was measured during the reperfusion. The contents of endogenous neuropeptides, endothelin-1 (ET-1) and calcitonin gene-related peptide in plasma were evaluated by radioimmunoassay. Superoxide dismutase (SOD) and malondialdehyde (MDA) in brain tissue homogenate were also examined.
Key findings
The results showed that catalpol significantly improved the stroke index compared with CI/R control group (P < 0.05 or P < 0.01). Catalpol significantly increased the activity of SOD at the doses of 10 and 20 mg/kg (P ≤ 0.05), decreased the brain MDA content and the plasma level of ET-1 at the doses of 10 and 20 mg/kg (P ≤ 0.01).
Conclusions
These data suggested that the efficacy of catalpol pretreatment on CI/R injury may be attributed to reduction of free radicals and inhibition of lipid peroxidation and ET-1 production.
Collapse
Affiliation(s)
- Yan-ru Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Peng-wei Li
- Pharmacy College, He'nan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Jian-jun Suo
- Department of Neurology, The Third People's Hospital of Sanmenxia, Sanmenxia, China
| | - Yan Sun
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bo-ai Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hong Lu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hong-can Zhu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guo-bin Zhang
- Pharmacy College, He'nan University of Traditional Chinese Medicine, Zhengzhou, China
| |
Collapse
|
26
|
Yang L, Zhao X, Sun M, Sun X, Yao L, Yu D, Ding Q, Gao C, Chai W. Delta opioid receptor agonist BW373U86 attenuates post-resuscitation brain injury in a rat model of asphyxial cardiac arrest. Resuscitation 2013; 85:299-305. [PMID: 24200890 DOI: 10.1016/j.resuscitation.2013.10.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 09/21/2013] [Accepted: 10/22/2013] [Indexed: 10/26/2022]
Abstract
OBJECTIVE The aim of this study was to investigate whether the DOR agonist BW373U86 conferred neuroprotection following ACA when given after resuscitation and to determine the long-term effects of chronic BW373U86 treatment on ACA-elicited brain injury. METHODS Animals were divided into acute and chronic treatment groups. Each group consisted of four sub-groups, including Sham, ACA, BW373U86 (BW373U86+ACA), and Naltrindole groups (Naltrindole and BW373U86+ACA). The DOR antagonist Naltrindole was used to confirm the possible receptor-dependent effects of BW373U86. ACA was induced by 8min of asphyxiation followed by resuscitation. All drugs were administered either immediately after the restoration of spontaneous circulation (ROSC) in acute-treatment groups or over 6 consecutive days in chronic-treatment groups. Alterations of cAMP response element-binding protein (CREB) and phosphorylated CREB (pCREB) were analyzed by western blot and immunohistochemistry. Neurological functions were assessed by neurological deficit score (NDS) and Morris Water Maze performance. Neurodegeneration was monitored by immunofluorescence and Nissl staining. RESULTS ACA induced massive neuron loss and serious neurological function deficits. BW373U86 significantly reduced both of these negative effects and increased CREB and pCREB expression in the hippocampus; these effects were reversed with acute Naltrindole treatment. The protective effects of BW373U86 persisted until 28d post-ROSC with chronic treatment, but these effects were not reversed by Naltrindole. CONCLUSIONS BW373U86 attenuates global cerebral ischemic injury induced by ACA through both DOR-dependent and DOR-independent mechanisms. CREB might be an important molecule in mediating these neuroprotective effects.
Collapse
Affiliation(s)
- Lu Yang
- Department of Anesthesiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province 710038, China
| | - Xiaoyong Zhao
- Department of Anesthesiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province 710038, China
| | - Meiyan Sun
- Department of Anesthesiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province 710038, China
| | - Xude Sun
- Department of Anesthesiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province 710038, China
| | - Linong Yao
- Department of Anesthesiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province 710038, China
| | - Daihua Yu
- Department of Anesthesiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province 710038, China
| | - Qian Ding
- Department of Anesthesiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province 710038, China
| | - Changjun Gao
- Department of Anesthesiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province 710038, China.
| | - Wei Chai
- Department of Anesthesiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province 710038, China.
| |
Collapse
|
27
|
Zhang W, Li H, Xing Z, Yuan H, Kindy MS, Li Z. Expression of mRNAs for PPT, CGRP, NF-200, and MAP-2 in cocultures of dissociated DRG neurons and skeletal muscle cells in administration of NGF or NT-3. Folia Histochem Cytobiol 2012; 50:312-8. [PMID: 22763971 DOI: 10.5603/fhc.2012.0041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 02/03/2012] [Indexed: 11/25/2022] Open
Abstract
Both neurotrophins (NTs) and target skeletal muscle (SKM) cells are essential for the maintenance of the function of neurons and nerve-muscle communication. However, much less is known about the association of target SKM cells with distinct NTs on the expression of mRNAs for preprotachykinin (PPT), calcitonin-gene related peptide (CGRP), neurofilament 200 (NF-200), and microtubule associated protein 2 (MAP-2) in dorsal root ganglion (DRG) sensory neurons. In the present study, a neuromuscular coculture model of dissociated dorsal root ganglion (DRG) neurons and SKM cells was established. The morphology of DRG neurons and SKM cells in coculture was observed with an inverted phase contrast microscope. The effects of nerve growth factor (NGF) or neurotrophin-3 (NT-3) on the expression of mRNAs for PPT, CGRP, NF-200, and MAP-2 was analyzed by real time-PCR assay. The morphology of DRG neuronal cell bodies and SKM cells in neuromuscular coculture at different conditions was similar. The neurons presented evidence of dense neurite outgrowth in the presence of distinct NTs in neuromuscular cocultures. NGF and NT-3 increased mRNA levels of PPT, CGRP, and NF-200, but not MAP-2, in neuromuscular cocultures. These results offer new clues towards a better understanding of the association of target SKM cells with distinct NTs on the expression of mRNAs for PPT, CGRP, NF-200 and MAP-2, and implicate the association of target SKM cells and NTs with DRG sensory neuronal phenotypes.
Collapse
Affiliation(s)
- Weiwei Zhang
- Department of Anatomy, Shandong University School of Medicine, Jinan, China
| | | | | | | | | | | |
Collapse
|
28
|
Tau and caspase 3 as targets for neuroprotection. Int J Alzheimers Dis 2012; 2012:493670. [PMID: 22693678 PMCID: PMC3369463 DOI: 10.1155/2012/493670] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Accepted: 04/01/2012] [Indexed: 11/26/2022] Open
Abstract
The peptide drug candidate NAP (davunetide) has demonstrated protective effects in various in vivo and in vitro models of neurodegeneration. NAP was shown to reduce tau hyperphosphorylation as well as to prevent caspase-3 activation and cytochrome-3 release from mitochondria, both characteristic of apoptotic cell death. Recent studies suggest that caspases may play a role in tau pathology. The purpose of this study was to evaluate the effect of NAP on tau hyperphosphorylation and caspase activity in the same biological system. Our experimental setup used primary neuronal cultures subjected to oxygen-glucose deprivation (OGD), with and without NAP or caspase inhibitor. Cell viability was assessed by measuring mitochondrial activity (MTS assay), and immunoblots were used for analyzing protein level. It was shown that apoptosis was responsible for all cell death occurring following ischemia, and NAP treatment showed a concentration-dependent protection from cell death. Ischemia caused an increase in the levels of active caspase-3 and hyperphosphorylated tau, both of which were prevented by either NAP or caspase-inhibitor treatment. Our data suggest that, in this model system, caspase activation may be an upstream event to tau hyperphosphorylation, although additional studies will be required to fully elucidate the cascade of events.
Collapse
|
29
|
Leptin attenuates cerebral ischemia/reperfusion injury partially by CGRP expression. Eur J Pharmacol 2011; 671:61-9. [DOI: 10.1016/j.ejphar.2011.09.170] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Revised: 09/14/2011] [Accepted: 09/15/2011] [Indexed: 01/08/2023]
|
30
|
Calcitonin gene-related peptide prevents blood–brain barrier injury and brain edema induced by focal cerebral ischemia reperfusion. ACTA ACUST UNITED AC 2011; 171:19-25. [DOI: 10.1016/j.regpep.2011.05.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2010] [Revised: 05/13/2011] [Accepted: 05/30/2011] [Indexed: 11/21/2022]
|
31
|
Inhibition of Neuron-Specific CREB Dephosphorylation is Involved in Propofol and Ketamine-Induced Neuroprotection Against Cerebral Ischemic Injuries of Mice. Neurochem Res 2011; 37:49-58. [DOI: 10.1007/s11064-011-0582-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2011] [Revised: 08/10/2011] [Accepted: 08/12/2011] [Indexed: 10/17/2022]
|