1
|
Xie S, Li F. Ependymal cells: roles in central nervous system infections and therapeutic application. J Neuroinflammation 2024; 21:255. [PMID: 39385253 PMCID: PMC11465851 DOI: 10.1186/s12974-024-03240-2] [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: 07/24/2024] [Accepted: 09/23/2024] [Indexed: 10/12/2024] Open
Abstract
Ependymal cells are arranged along the inner surfaces of the ventricles and the central canal of the spinal cord, providing anatomical, physiological and immunological barriers that maintain cerebrospinal fluid (CSF) homeostasis. Based on this, studies have found that alterations in gene expression, cell junctions, cytokine secretion and metabolic disturbances can lead to dysfunction of ependymal cells, thereby participating in the onset and progression of central nervous system (CNS) infections. Additionally, ependymal cells can exhibit proliferative and regenerative potential as well as secretory functions during CNS injury, contributing to neuroprotection and post-injury recovery. Currently, studies on ependymal cell primarily focus on the basic investigations of their morphology, function and gene expression; however, there is a notable lack of clinical translational studies examining the molecular mechanisms by which ependymal cells are involved in disease onset and progression. This limits our understanding of ependymal cells in CNS infections and the development of therapeutic applications. Therefore, this review will discuss the molecular mechanism underlying the involvement of ependymal cells in CNS infections, and explore their potential for application in clinical treatment modalities.
Collapse
Affiliation(s)
- Shiqi Xie
- Department of Pulmonary and Critical Care Medicine, Shanghai Public Health Clinical Center, Fudan University, 2901 Cao Lang Road, Jinshan District, Shanghai, China
| | - Feng Li
- Department of Pulmonary and Critical Care Medicine, Shanghai Public Health Clinical Center, Fudan University, 2901 Cao Lang Road, Jinshan District, Shanghai, China.
- Shanghai Institute of Infectious Disease and Biosecurity, 130 Dong An Road, Xuhui District, Shanghai, China.
- Tuberculosis Research Center, Shanghai Public Health Clinical Center, Fudan University, 2901 Cao Lang Road, Jinshan District, Shanghai, China.
| |
Collapse
|
2
|
Chen Z, Shao J, Yang Y, Wang G, Xiong Z, Song X, Ai L, Xia Y, Zhu B. Evaluation of Functional Components of Lactobacillus plantarum AR495 on Ovariectomy-Induced Osteoporosis in Mice And RAW264.7 Cells. Foods 2024; 13:3115. [PMID: 39410150 PMCID: PMC11476097 DOI: 10.3390/foods13193115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/21/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
Osteoporosis is a disease characterized by abnormal bone metabolism, where bone resorption outpaces bone formation. In this study, we investigated the key functional components of Lactobacillus plantarum AR495 in mitigating ovariectomy (OVX)-induced osteoporosis in mice. The results indicated that both Lactobacillus plantarum AR495 and its fermentation broth significantly reduced urinary calcium and deoxypyridinoline (DPD) levels in the mice. These interventions inhibited bone resorption and improved trabecular bone architecture by modulating the nuclear factor κB (RANK)/RANK ligand (RANKL)/osteoprotegerin (OPG) signaling pathway. Additionally, the L. plantarum AR495 and fermentation broth groups inhibited the RANKL/TRAF-6 and TLR4/MYD88 pathways, leading to enhanced bone metabolism, improved intestinal barrier function, and reduced intestinal inflammation. In vitro experiments revealed that AR495 fermentation supernatant fractions larger than 100 kDa and those between 50-100 kDa significantly decreased the activity of the osteoclast marker TRAP, regulated the expression of the TLR4/MYD88 pathway, and inhibited osteoclast formation, thereby alleviating the OVX-induced osteoporosis phenotype. These findings suggest that these components may be primary functional elements of L. plantarum AR495 in the treatment of osteoporosis.
Collapse
Affiliation(s)
- Zheng Chen
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China;
| | - Junlin Shao
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai 200093, China; (J.S.); (Y.Y.); (G.W.); (Z.X.); (X.S.); (L.A.)
| | - Yijin Yang
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai 200093, China; (J.S.); (Y.Y.); (G.W.); (Z.X.); (X.S.); (L.A.)
| | - Guangqiang Wang
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai 200093, China; (J.S.); (Y.Y.); (G.W.); (Z.X.); (X.S.); (L.A.)
| | - Zhiqiang Xiong
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai 200093, China; (J.S.); (Y.Y.); (G.W.); (Z.X.); (X.S.); (L.A.)
| | - Xin Song
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai 200093, China; (J.S.); (Y.Y.); (G.W.); (Z.X.); (X.S.); (L.A.)
| | - Lianzhong Ai
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai 200093, China; (J.S.); (Y.Y.); (G.W.); (Z.X.); (X.S.); (L.A.)
| | - Yongjun Xia
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai 200093, China; (J.S.); (Y.Y.); (G.W.); (Z.X.); (X.S.); (L.A.)
| | - Beiwei Zhu
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China;
| |
Collapse
|
3
|
Eskandari S, Rezayof A, Asghari SM, Hashemizadeh S. Neurobiochemical characteristics of arginine-rich peptides explain their potential therapeutic efficacy in neurodegenerative diseases. Neuropeptides 2023; 101:102356. [PMID: 37390744 DOI: 10.1016/j.npep.2023.102356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/02/2023]
Abstract
Neurodegenerative diseases, including Alzheimer̕ s disease (AD), Parkinson̕ s disease (PD), Huntington̕ s disease (HD), and Amyotrophic Lateral Sclerosis (ALS) require special attention to find new potential treatment methods. This review aims to summarize the current knowledge of the relationship between the biochemical properties of arginine-rich peptides (ARPs) and their neuroprotective effects to deal with the harmful effects of risk factors. It seems that ARPs have portrayed a promising and fantastic landscape for treating neurodegeneration-associated disorders. With multimodal mechanisms of action, ARPs play various unprecedented roles, including as the novel delivery platforms for entering the central nervous system (CNS), the potent antagonists for calcium influx, the invader molecules for targeting mitochondria, and the protein stabilizers. Interestingly, these peptides inhibit the proteolytic enzymes and block protein aggregation to induce pro-survival signaling pathways. ARPs also serve as the scavengers of toxic molecules and the reducers of oxidative stress agents. They also have anti-inflammatory, antimicrobial, and anti-cancer properties. Moreover, by providing an efficient nucleic acid delivery system, ARPs can play an essential role in developing various fields, including gene vaccines, gene therapy, gene editing, and imaging. ARP agents and ARP/cargo therapeutics can be raised as an emergent class of neurotherapeutics for neurodegeneration. Part of the aim of this review is to present recent advances in treating neurodegenerative diseases using ARPs as an emerging and powerful therapeutic tool. The applications and progress of ARPs-based nucleic acid delivery systems have also been discussed to highlight their usefulness as a broad-acting class of drugs.
Collapse
Affiliation(s)
- Sedigheh Eskandari
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran; Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Ameneh Rezayof
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran.
| | - S Mohsen Asghari
- Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran.
| | - Shiva Hashemizadeh
- School of Cognitive Sciences, Institute for Research in Fundamental Sciences, IPM, Tehran, Iran
| |
Collapse
|
4
|
Tang L, Xu Y, Wang L, Pan J. Adipose-derived stem cell exosomes ameliorate traumatic brain injury through the NLRP3 signaling pathway. Neuroreport 2023; 34:677-684. [PMID: 37506308 PMCID: PMC10399942 DOI: 10.1097/wnr.0000000000001941] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023]
Abstract
The exosomes of mesenchymal stem cells have immunoregulatory properties and can effectively mitigate secondary neuroinflammation due to traumatic brain injury (TBI). In this study, we found that adipose-derived stem cell exosomes (ADSCs-Exo) could reduce the inflammatory response after traumatic brain injury by reducing NLRP3 inflammasome secretion by microglial. ADSCs-Exo were monitored by Western blot and electron microscopy. An in-vitro lipopolysaccharide (LPS)-caused primary microglia model and a TBI rat model were constructed. Functional recovery was examined using the modified neurological severity score and foot fault tests. Inflammasome inactivation in LPS-stimulated microglial, ADSCs-Exo can reduce the secretion of interleukin (IL)-1β, IL-6 and tumor necrosis factor α. Compared with PBS-processed controls, the sensorimotor functional recovery was significantly improved by exosome treatment after injury at 14-35 days. Additionally, NLRP3 inflammasome was stimulated within 24 h after TBI. ADSCs-Exo application led to remarkable down-expression of NLRP3 and caspase-1. ADSCs-Exo can ameliorate LPS-induced inflammatory activation by reducing microglial pro-inflammatory cytokines. Moreover, the neuroprotective effect of ADSCs-Exo may be partially attributed to the inhibition thereof on the formation of NLRP3-mediated inflammasome. Such findings imply a potential function of ADSCs-Exo in treating TBI.
Collapse
Affiliation(s)
- Linjun Tang
- Department of Neurosurgery, The Second People’s Hospital of Wuhu, Wuhu, Anhui, China
| | - Yong Xu
- Department of Neurosurgery, The Second People’s Hospital of Wuhu, Wuhu, Anhui, China
| | - Liangwei Wang
- Department of Neurosurgery, The Second People’s Hospital of Wuhu, Wuhu, Anhui, China
| | - Jingjing Pan
- Department of Neurosurgery, The Second People’s Hospital of Wuhu, Wuhu, Anhui, China
| |
Collapse
|
5
|
Govindarajulu M, Patel MY, Wilder DM, Krishnan J, LaValle C, Pandya JD, Shear DA, Hefeneider SH, Long JB, Arun P. Upregulation of multiple toll-like receptors in ferret brain after blast exposure: potential targets for treatment. Neurosci Lett 2023; 810:137364. [PMID: 37391063 DOI: 10.1016/j.neulet.2023.137364] [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: 10/21/2022] [Revised: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 07/02/2023]
Abstract
Although blast-induced traumatic brain injury (bTBI) has been designated as the signature injury of recent combat operations, its precise pathological mechanism(s) has not been identified thus far. Prior preclinical studies on bTBI demonstrated acute neuroinflammatory cascades which are known to be contributing to neurodegeneration. Danger-associated chemical patterns are released from the injured cells, which activate non-specific pattern recognition receptors, such as toll-like receptors (TLRs) leading to increased expression of inflammatory genes and release of cytokines. Upregulation of specific TLRs in the brain has been described as a mechanism of injury in diverse brain injury models unrelated to blast exposure. However, the expression profile of various TLRs in bTBI has not been investigated thus far. Hence, we have evaluated the expression of transcripts for TLR1-TLR10 in the brain of a gyrencephalic animal model of bTBI. We exposed ferrets to tightly coupled repeated blasts and determined the differential expression of TLRs (TLR1-10) by quantitative RT-PCR in multiple brain regions at 4 hr, 24 hr, 7 days and 28 days post-blast injury. The results obtained indicate that multiple TLRs are upregulated in the brain at 4 hr, 24 hr, 7 days and 28 days post-blast. Specifically, upregulation of TLR2, TLR4 and TLR9 was noted in different brain regions, suggesting that multiple TLRs might play a role in the pathophysiology of bTBI and that drugs that can inhibit multiple TLRs might have enhanced efficacy to attenuate brain damage and thereby improve bTBI outcome. Taken together, these results suggest that several TLRs are upregulated in the brain after bTBI and participate in the inflammatory response and thereby provide new insights into the disease pathogenesis. Therefore, inhibition of multiple TLRs, including TLR2, 4 and 9, simultaneously might be a potential therapeutic strategy for the treatment of bTBI.
Collapse
Affiliation(s)
- Manoj Govindarajulu
- Blast-Induced Neurotrauma Branch, Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Mital Y Patel
- Blast-Induced Neurotrauma Branch, Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Donna M Wilder
- Blast-Induced Neurotrauma Branch, Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Jishnu Krishnan
- Blast-Induced Neurotrauma Branch, Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Christina LaValle
- Blast-Induced Neurotrauma Branch, Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Jignesh D Pandya
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Deborah A Shear
- Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | | | - Joseph B Long
- Blast-Induced Neurotrauma Branch, Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Peethambaran Arun
- Blast-Induced Neurotrauma Branch, Brain Trauma Neuroprotection and Neurorestoration Branch, Center for Military Psychiatry and Neuroscience, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA.
| |
Collapse
|
6
|
Nelles DG, Hazrati LN. The pathological potential of ependymal cells in mild traumatic brain injury. Front Cell Neurosci 2023; 17:1216420. [PMID: 37396927 PMCID: PMC10312375 DOI: 10.3389/fncel.2023.1216420] [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: 05/03/2023] [Accepted: 06/05/2023] [Indexed: 07/04/2023] Open
Abstract
Mild traumatic brain injury (mTBI) is a common neurological condition affecting millions of individuals worldwide. Although the pathology of mTBI is not fully understood, ependymal cells present a promising approach for studying the pathogenesis of mTBI. Previous studies have revealed that DNA damage in the form of γH2AX accumulates in ependymal cells following mTBI, with evidence of widespread cellular senescence in the brain. Ependymal ciliary dysfunction has also been observed, leading to altered cerebrospinal fluid homeostasis. Even though ependymal cells have not been extensively studied in the context of mTBI, these observations reflect the pathological potential of ependymal cells that may underlie the neuropathological and clinical presentations of mTBI. This mini review explores the molecular and structural alterations that have been reported in ependymal cells following mTBI, as well as the potential pathological mechanisms mediated by ependymal cells that may contribute to overall dysfunction of the brain post-mTBI. Specifically, we address the topics of DNA damage-induced cellular senescence, dysregulation of cerebrospinal fluid homeostasis, and the consequences of impaired ependymal cell barriers. Moreover, we highlight potential ependymal cell-based therapies for the treatment of mTBI, with a focus on neurogenesis, ependymal cell repair, and modulation of senescence signaling pathways. Further insight and research in this field will help to establish the role of ependymal cells in the pathogenesis of mTBI and may lead to improved treatments that leverage ependymal cells to target the origins of mTBI pathology.
Collapse
Affiliation(s)
- Diana G. Nelles
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- The Hospital for Sick Children, Toronto, ON, Canada
| | - Lili-Naz Hazrati
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- The Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
7
|
Wang C, Ye H, Zheng Y, Qi Y, Zhang M, Long Y, Hu Y. Phenylethanoid Glycosides of Cistanche Improve Learning and Memory Disorders in APP/PS1 Mice by Regulating Glial Cell Activation and Inhibiting TLR4/NF-κB Signaling Pathway. Neuromolecular Med 2023; 25:75-93. [PMID: 35781783 DOI: 10.1007/s12017-022-08717-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 06/06/2022] [Indexed: 11/25/2022]
Abstract
Phenylethanoid Glycosides of Cistanche (PhGs) have a certain curative effect on AD animal model, Echinacea (ECH) and verbascoside (ACT), as the quality control standard of Cistanche deserticola Y. C. Ma and the main representative compounds of PhGs have been proved to have neuroprotective effects, but the specific mechanism needs to be further explored. This study explored the mechanisms of PhGs, ECH, and ACT in the treatment of Alzheimer's disease (AD) from the perspectives of glial cell activation, TLR4/NF-κB signaling pathway, and synaptic protein expression. We used APP/PS1 mice as AD models. After treatment with PhGs, ECH, and ACT, the learning and memory abilities of APP/PS1 mice were enhanced, and the pathological changes in brain tissue were alleviated. The expression of pro-inflammatory M1 microglia markers (CD11b, iNOS, and IL-1β) was decreased; the expression of M2 microglia markers (Arg-1 and TGF-β1) was increased, which promoted the transformation of microglia from M1 pro-inflammatory phenotype to M2 anti-inflammatory phenotype. In addition, PhGs, ECH, and ACT could down-regulate the expression of proteins related to the TLR4/NF-κB signaling pathway and up-regulate the expression of synaptic proteins. The results indicated that PhGs, ECH, and ACT played a neuroprotective role by regulating the activation of glial cells and inhibiting the TLR4/NF-κB inflammatory pathway, and improving the expression levels of synapse-related proteins.
Collapse
Affiliation(s)
- Chunhui Wang
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Department of Pharmacology, Shihezi University, Shihezi, 832000, Xinjiang, People's Republic of China
| | - Hongxia Ye
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Department of Pharmacology, Shihezi University, Shihezi, 832000, Xinjiang, People's Republic of China
| | - Yanjie Zheng
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Department of Pharmacology, Shihezi University, Shihezi, 832000, Xinjiang, People's Republic of China
| | - Yanqiang Qi
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Department of Pharmacology, Shihezi University, Shihezi, 832000, Xinjiang, People's Republic of China
| | - Mengyu Zhang
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Department of Pharmacology, Shihezi University, Shihezi, 832000, Xinjiang, People's Republic of China
| | - Yan Long
- Central Laboratory, Shenzhen Sami Medical Center, Shenzhen, China
| | - Yanli Hu
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Department of Pharmacology, Shihezi University, Shihezi, 832000, Xinjiang, People's Republic of China.
| |
Collapse
|
8
|
Mahmoud MS, El-Kott AF, AlGwaiz HIM, Fathy SM. Protective effect of Moringa oleifera Lam. leaf extract against oxidative stress, inflammation, depression, and apoptosis in a mouse model of hepatic encephalopathy. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:83783-83796. [PMID: 35771324 DOI: 10.1007/s11356-022-21453-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 06/10/2022] [Indexed: 06/15/2023]
Abstract
The present study aimed to assess the antioxidative, anti-inflammatory, antiapoptotic, and anti-depression impacts of Moringa oleifera Lam. leaf ethanolic extract (MOLE) in the hippocampus and cerebral cortex of CCl4-induced hepatic encephalopathy mouse model. High-performance liquid chromatography was used to detect marker compounds: rutin and β-sitosterol. Animals were divided into four groups: vehicle group, CCl4-treated group, MOLE-treated group, and (CCl4 + MOLE) group treated with MOLE for 14 days before CCl4-induced neurotoxicity. MOLE decreased alanine aminotransferase, aspartate aminotransferase, corticosterone, and ammonia levels in serum and improved the antioxidant status of CCl4-treated mice in the hippocampus and cerebral cortex. It reduced the expression of toll-like receptor 4 (TLR4), TLR2, myeloid differentiation primary response 88 (MYD88), and nuclear factor-kappa B (NF-κB) genes and the protein levels of the pro-inflammatory cytokines. MOLE also attenuated apoptosis, as revealed by the reduced expression of caspase3, and prevented histological deterioration. Furthermore, MOLE attenuated CCl4-induced anxiety and depression-like behavioral changes. Collectively, MOLE modulates neuroinflammation, oxidative stress, TLR4/2-MyD88/NF-κB signaling, and apoptosis in the hippocampus and cerebral cortex of the hepatic encephalopathy experimental model.
Collapse
Affiliation(s)
- Mohammed S Mahmoud
- Zoology Department, Faculty of Science, Fayoum University, Fayoum, 63514, Egypt.
| | - Attalla F El-Kott
- Department of Biology, College of Science, King Khalid University, Abha, 61421, Saudi Arabia
- Department of Zoology, Faculty of Science, Damanhour University, Damanhour, 22511, Egypt
| | - Hussah I M AlGwaiz
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, 11474, Riyadh, Saudi Arabia
| | - Samah M Fathy
- Zoology Department, Faculty of Science, Fayoum University, Fayoum, 63514, Egypt
| |
Collapse
|
9
|
Cao LX, Lin SJ, Zhao SS, Wang SQ, Zeng H, Chen WA, Lin ZW, Chen JX, Zhu MM, Zhang YM. Effects of acupuncture on microglial polarization and the TLR4/TRIF/MyD88 pathway in a rat model of traumatic brain injury. Acupunct Med 2022:9645284221108214. [PMID: 36046956 DOI: 10.1177/09645284221108214] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE Neuroinflammation caused by traumatic brain injury (TBI) can lead to neurological deficits. Acupuncture can inhibit neuroinflammation and promote nerve repair; however, the specific mechanism is still unclear. The purpose of this study was to explore whether acupuncture could modulate the M1 and M2 phenotypic polarization of microglia in a rat model of TBI via the toll-like receptor 4 (TLR4)/intracellular toll-interleukin-1 receptor (TIR) domain-containing adaptor inducing interferon-β (TRIF)/myeloid differentiation factor 88 (MyD88) pathway. METHODS A total of 90 adult male Sprague-Dawley (SD) rats, SPF grade, were randomly divided into a normal group, model group and acupuncture group. Each group was further divided into three subgroups (first, third, and fifth day groups) according to the treatment time (n = 10 rats/subgroup). We used the modified neurological severity score (mNSS) method to quantify neurological deficits before and after modeling. We used Nissl staining to observe the pathological changes in brain tissue, flow cytometry to detect the proportion of M1 and M2 polarized microglia in the injured area on the first, third and fifth day, and co-immunoprecipitation (Co-IP) to examine TLR4/TRIF/MyD88 expression in microglia on the first, third and fifth day, as well as expression of the amount of binding of TLR4 with TRIF and MyD88. RESULTS Compared to the model group, mNSS in the acupuncture group gradually decreased and pathological morphology improved. The proportion of CD11b/CD86 positive cells was decreased, while that of CD11b/CD206 was increased in the acupuncture group. Expression of IP TLR4, IP TRIF and IP MyD88 also decreased in the acupuncture group. CONCLUSION The results of this study demonstrate that one of the mechanisms through which acupuncture mitigates neuroinflammation and promotes nerve repair in TBI rats may be inhibition of M1 phenotypic polarization and promotion of M2 phenotypic polarization through inhibition of the TLR4/TRIF/MyD88 signaling pathway.
Collapse
Affiliation(s)
- Lu-Xi Cao
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China.,Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Shu-Jun Lin
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China.,Clinical Medical College of Acupuncture, Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Si-Si Zhao
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Shi-Qi Wang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Hai Zeng
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Wen-An Chen
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Zhuo-Wen Lin
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Jia-Xu Chen
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China.,Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Ming-Min Zhu
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Yi-Min Zhang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| |
Collapse
|
10
|
Activating toll-like receptor 4 after traumatic brain injury inhibits neuroinflammation and the accelerated development of seizures in rats. Exp Neurol 2022; 357:114202. [PMID: 35970203 DOI: 10.1016/j.expneurol.2022.114202] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 08/09/2022] [Indexed: 01/01/2023]
Abstract
Toll-like receptor 4 (TLR4) signaling plays a detrimental role in traumatic brain injury (TBI) pathology. Pharmacologic or genetic inactivating TLR4 diminish TBI inflammation and neurological complications. Nonetheless, TLR4 priming alleviates TBI inflammation and seizure susceptibility. We investigated impact of postconditioning with TLR4 agonist monophosphoryl lipid A (MPL) on TBI neuroinflammation and epileptogenesis in rats. TBI was induced in temporo-parietal cortex of rats by Controlled Cortical Impact device. Then rats received a single dose (0.1 μg/rat) of MPL by intracerebroventricular injection. After 24 h, CCI-injured rats received intraperitoneal injection of pentylenetetrazole 35 mg/kg once every other day until acquisition of generalized seizures. The injury size, number of survived neurons, and brain protein level of TNF-α, TGF-β, IL-10, and arginase1 (Arg1) were determined. Astrocytes and macrophage/microglia activation/polarization was assessed by double immunostaining with anti GFAP/Arg1 or anti Iba1/Arg1 antibodies. The CCI-injured rats developed generalized seizures after 5.9 ± 1.3 pentylenetetrazole injections (p < 0.001, compared to 12.3 ± 1.4 injections for sham-operated rats). MPL treatment returned the accelerated rate of epileptogenesis in TBI state to the sham-operated level. MPL did not change damage volume but attenuated number of dead neurons (p < 0.01). MPL decreased TNF-α overexpression (6 h post-TBI p < 0.0001), upregulated expression of TGF-β (48 h post-TBI, p < 0.0001), and IL-10 (48 h post-TBI, p < 0.0001) but did not change Arg1 expression. GFAP/Arg1 and Iba1/Arg1 positive cells were detected in TBI area with no significant change following MPL administration. MPL administration after TBI reduces vulnerability to seizure acquisition through down regulating neural death and inflammation, and up-regulating anti-inflammatory cytokines. This capacity along with the clinical safety, makes MPL a potential candidate for development of drugs against neurological deficits of TBI.
Collapse
|
11
|
Mohseni-Moghaddam P, Dogani M, Hatami M, Roohollahi S, Amiresmaeli A, Askari N. A behavioral and molecular study; ameliorated anxiety-like behavior and cognitive dysfunction in a rat model of chronic unpredictable stress treated with oregano extract. Brain Behav 2022; 12:e2727. [PMID: 35898162 PMCID: PMC9392516 DOI: 10.1002/brb3.2727] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/01/2022] [Accepted: 07/12/2022] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE Chronic stress is considered a severe risk factor leading to various disorders, including anxiety and cognitive decline. The present study aimed to investigate the effects of Origanum vulgare (oregano) extract on improving anxiety-like behavior and learning and memory defection caused by chronic unpredictable stress (CUS). METHOD A 10-day CUS protocol was executed on male rats, and on day 10, their anxiety, learning, and memory status were evaluated. After that, in addition to the CUS, the rats were treated with the oregano extract for 2 weeks. Then, the expression of BDNF, TrkB, and TLR2/4 genes in the hippocampus and prefrontal cortex of the rats was evaluated. Also, the liver- and kidney-related serum parameters, including triglycerides, total cholesterol, HDL, LDL, creatinine, urea, serum glucose, alanine aminotransferase, and aspartate aminotransferase were assessed. Further, the extract's lethal effect and its impact on animals' body weight were investigated. RESULTS Behavioral tests confirmed the anxiety-like behavior and learning-memory function impairment caused by CUS. In contrast, the administration of the extract could significantly alleviate the mental deficiencies and diminished anxiety-like behaviors. Molecular assessments showed that CUS could markedly decrease the BDNF and TrkB genes' expression levels while increasing that of TLR2 and TLR4. In contrast, in extract-treated animals, mRNA levels of BDNF and TrkB considerably increased, yet TLR2 and TLR4 mRNA levels reduced. Additionally, consumption of the extract caused weight gain, while having no lethality and detrimental effect on the liver and kidneys functions. CONCLUSIONS These findings indicate the anxiolytic properties of the extract and its improving effect on cognitive dysfunction.
Collapse
Affiliation(s)
- Parvaneh Mohseni-Moghaddam
- Department of Physiology, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Manijeh Dogani
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Motahare Hatami
- Department of Cardiology, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Samira Roohollahi
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Azam Amiresmaeli
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Nayereh Askari
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran.,Immunoregulation Research Center, Shahed University, Tehran, Iran
| |
Collapse
|
12
|
Jansen MI, Thomas Broome S, Castorina A. Exploring the Pro-Phagocytic and Anti-Inflammatory Functions of PACAP and VIP in Microglia: Implications for Multiple Sclerosis. Int J Mol Sci 2022; 23:ijms23094788. [PMID: 35563181 PMCID: PMC9104531 DOI: 10.3390/ijms23094788] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/22/2022] [Accepted: 04/25/2022] [Indexed: 02/04/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic neuroinflammatory and demyelinating disease of the central nervous system (CNS), characterised by the infiltration of peripheral immune cells, multifocal white-matter lesions, and neurodegeneration. In recent years, microglia have emerged as key contributors to MS pathology, acting as scavengers of toxic myelin/cell debris and modulating the inflammatory microenvironment to promote myelin repair. In this review, we explore the role of two neuropeptides, pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal peptide (VIP), as important regulators of microglial functioning during demyelination, myelin phagocytosis, and remyelination, emphasising the potential of these neuropeptides as therapeutic targets for the treatment of MS.
Collapse
|
13
|
Lin PH, Kuo LT, Luh HT. The Roles of Neurotrophins in Traumatic Brain Injury. LIFE (BASEL, SWITZERLAND) 2021; 12:life12010026. [PMID: 35054419 PMCID: PMC8780368 DOI: 10.3390/life12010026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/18/2021] [Accepted: 12/21/2021] [Indexed: 02/08/2023]
Abstract
Neurotrophins are a collection of structurally and functionally related proteins. They play important roles in many aspects of neural development, survival, and plasticity. Traumatic brain injury (TBI) leads to different levels of central nervous tissue destruction and cellular repair through various compensatory mechanisms promoted by the injured brain. Many studies have shown that neurotrophins are key modulators of neuroinflammation, apoptosis, blood–brain barrier permeability, memory capacity, and neurite regeneration. The expression of neurotrophins following TBI is affected by the severity of injury, genetic polymorphism, and different post-traumatic time points. Emerging research is focused on the potential therapeutic applications of neurotrophins in managing TBI. We conducted a comprehensive review by organizing the studies that demonstrate the role of neurotrophins in the management of TBI.
Collapse
Affiliation(s)
- Ping-Hung Lin
- Department of Medical Education, School of Medicine, National Taiwan University, Taipei 100, Taiwan;
| | - Lu-Ting Kuo
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital, Taipei 100, Taiwan;
| | - Hui-Tzung Luh
- Department of Neurosurgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, New Taipei City 235, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University, Taipei 100, Taiwan
- Correspondence: ; Tel.: +886-956279587
| |
Collapse
|
14
|
Yu J, Xia Y, Wang G, Xiong Z, Zhang H, Lai PFH, Song X, Ai L. Anti-osteoporotic potential of Lactobacillus plantarum AR237 and AR495 in ovariectomized mice. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
15
|
Rodríguez A, Velázquez J, González L, Rodríguez-Ramos T, Dixon B, Miyares FH, Morales A, González O, Estrada MP, Carpio Y. PACAP modulates the transcription of TLR-1/TLR-5/MyD88 pathway genes and boosts antimicrobial defenses in Clarias gariepinus. FISH & SHELLFISH IMMUNOLOGY 2021; 115:150-159. [PMID: 34146673 DOI: 10.1016/j.fsi.2021.06.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/10/2021] [Accepted: 06/13/2021] [Indexed: 06/12/2023]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a multifunctional neuropeptide that belongs to the secretin/glucagon/GHRH/VIP superfamily. Some of these molecules have antimicrobial activity and they are capable of stimulating the immune system. The present work studied the antibacterial and immunostimulatory activity of PACAP-38 from African catfish Clarias gariepinus against the Gram-negative bacterium Pseudomonas aeruginosa in an in vivo test. PACAP-38 improved antimicrobial activity of skin mucus molecules against P. aeruginosa. The peptide modulates the gene expression profile of TLR-1, TLR-5, MyD88, IL-1β, TNF-ɑ, IL-8, pardaxin, hepcidin and G/C-type lysozymes in skin, spleen and head kidney. The influenced exerted depended on the time after infection and tissue analyzed. This study provides the first evidence of a link between PACAP and antimicrobial peptides hepcidin and pardaxin. Our results suggest further use of PACAP as antimicrobial agent that could potentially be used to control disease in aquaculture.
Collapse
Affiliation(s)
- Alianet Rodríguez
- Veterinary Immunology Project, Animal Biotechnology Division, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Janet Velázquez
- Veterinary Immunology Project, Animal Biotechnology Division, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Luis González
- Veterinary Immunology Project, Animal Biotechnology Division, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Tania Rodríguez-Ramos
- Department of Biology, University of Waterloo, 200 University Ave W., Waterloo, ON, Canada
| | - Brian Dixon
- Department of Biology, University of Waterloo, 200 University Ave W., Waterloo, ON, Canada
| | - Fidel Herrera Miyares
- Veterinary Immunology Project, Animal Biotechnology Division, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Antonio Morales
- Veterinary Immunology Project, Animal Biotechnology Division, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Osmany González
- Veterinary Immunology Project, Animal Biotechnology Division, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Mario Pablo Estrada
- Veterinary Immunology Project, Animal Biotechnology Division, Center for Genetic Engineering and Biotechnology, Havana, Cuba.
| | - Yamila Carpio
- Veterinary Immunology Project, Animal Biotechnology Division, Center for Genetic Engineering and Biotechnology, Havana, Cuba.
| |
Collapse
|
16
|
Wu L, Ji NN, Wang H, Hua JY, Sun GL, Chen PP, Hua R, Zhang YM. Domino Effect of Interleukin-15 and CD8 T-Cell-Mediated Neuronal Apoptosis in Experimental Traumatic Brain Injury. J Neurotrauma 2021; 38:1450-1463. [PMID: 30430911 DOI: 10.1089/neu.2017.5607] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The effects of local factors on activation of immune cells infiltrating the central nervous system (CNS) in a rat model of traumatic brain injury (TBI) remain elusive. The cytokine, interleukin (IL)-15, is crucial for development and activation of CD8 T lymphocytes, a prominent lymphocytic population present in TBI lesions. We investigated whether IL-15 originates from astrocytes and whether IL-15 can evoke the CD8 T-lymphocyte response in TBI. We observed that astrocytes were activated in a rat model of TBI and that IL-15 was overexpressed on the surface of astrocytes. Further, CD8 T lymphocytes infiltrating TBI lesions colocalized with IL-15-expressing astrocytes. Activated CD8 T lymphocytes released granzyme B (Gra-b), which, in turn, activated caspase-3-induced poly(ADP-ribose) polymerase cleavage and, ultimately, neuronal apoptosis. Conversely, inhibition of astrocyte activation by pre-treatment with the specific inhibitor, fluorocitrate (FC), that reduces carbon flux through the Krebs cycle in astrocytes resulted in improved neurological function and memory. FC pre-treatment was also associated with downregulated IL-15 expression and CD8 T-cell activation as well as decreased levels of neuronal apoptosis, suggesting that IL-15 initiated a domino effect toward apoptosis. In contrast, rats pre-treated with recombinant rat IL-15 showed upregulated CD8 T-cell numbers and Gra-b levels, in addition to induction of neuronal apoptosis. Together, our results indicated that IL-15 could induce neuronal apoptosis by enhancing CD8 T-cell function in a rat model of TBI.
Collapse
Affiliation(s)
- Liang Wu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China.,Department of Anesthesiology, Xuzhou Municipal Hospital Affiliated with Xuzhou Medical University, Xuzhou, China
| | - Ning-Ning Ji
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China.,Department of Anesthesiology, Xuzhou Municipal Hospital Affiliated with Xuzhou Medical University, Xuzhou, China
| | - Hang Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Jing-Yu Hua
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Guo-Lin Sun
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Pan-Pan Chen
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| | - Rong Hua
- Faculty of Emergency Rescue Medicine, Xuzhou Medical University, Xuzhou, China.,Emergency Center of the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yong-Mei Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
17
|
Pituitary Adenylate Cyclase-Activating Polypeptide: A Potent Therapeutic Agent in Oxidative Stress. Antioxidants (Basel) 2021; 10:antiox10030354. [PMID: 33653014 PMCID: PMC7996859 DOI: 10.3390/antiox10030354] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/13/2021] [Accepted: 02/22/2021] [Indexed: 12/13/2022] Open
Abstract
Stroke is a life-threatening condition that is characterized by secondary cell death processes that occur after the initial disruption of blood flow to the brain. The inability of endogenous repair mechanisms to sufficiently support functional recovery in stroke patients and the inadequate treatment options available are cause for concern. The pathology behind oxidative stress in stroke is of particular interest due to its detrimental effects on the brain. The oxidative stress caused by ischemic stroke overwhelms the neutralization capacity of the body's endogenous antioxidant system, which leads to an overproduction of reactive oxygen species (ROS) and reactive nitrogen species (RNS) and eventually results in cell death. The overproduction of ROS compromises the functional and structural integrity of brain tissue. Therefore, it is essential to investigate the mechanisms involved in oxidative stress to help obtain adequate treatment options for stroke. Here, we focus on the latest preclinical research that details the mechanisms behind secondary cell death processes that cause many central nervous system (CNS) disorders, as well as research that relates to how the neuroprotective molecular mechanisms of pituitary adenylate cyclase-activating polypeptides (PACAPs) could make these molecules an ideal candidate for the treatment of stroke.
Collapse
|
18
|
Rui Q, Cao S, Wang X, Duan X, Iao X, Dong W, Fang Q, Zhang X, Xue Q. LMTK2 regulates inflammation in lipopolysaccharide-stimulated BV2 cells. Exp Ther Med 2021; 21:219. [PMID: 33603828 DOI: 10.3892/etm.2021.9621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 10/09/2020] [Indexed: 12/30/2022] Open
Abstract
Microglia activation plays vital roles in neuroinflammatory pathologys. Lemurs tyrosine kinase 2 (LMTK2) was reported to regulate NF-κB signals. In the present study, the roles of LMTK2 were investigated in lipopolysaccharide (LPS)-treated BV-2 cells. Reverse transcription-quantitative (RT-q)PCR and western blotting (WB) were utilized to analyze LMTK2 levels in LPS-treated BV2 cells. MTT assay determined cell viabilities. Nitric oxide (NO) and prostaglandin E2 (PGE2) levels were assessed through Griess and enzyme-linked immunosorbent assay (ELISA), respectively. The expression level of inducible NO synthase (iNOS) and cyclooxygenase-2 (COX-2) were detected through RT-qPCR and WB. The release of inflammatory mediators under LPS stimulation, tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), IL-6 and IL-10, were analyzed through ELISA. WB was used to analyze the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase 1 (HO-1)/NAD(P)H dehydrogenase quinone 1 (NQO1) signal pathway. The results showed that the levels of the inflammatory mediators, iNOS, NO, COX-2 and PGE2, along with pro-inflammatory factors, TNF-α, IL-1β and IL-6, were significantly decreased following the induction of exogenous LMTK2 expression by LMTK2 overexpression plasmids in LPS-induced BV2 microglia. In contrast, anti-inflammatory factor IL-10 showed obvious decrease. Additionally, LMTK2 overexpression induced the elevation of Nrf2 in the cytoplasm and nucleus, along with the upregulation of HO-1 and NQO1 expression. In conclusion, LMTK2 is postulated to regulate neuroinflammation possibly through Nrf2 pathway. The present study is essential to reveal the underlying function of LMTK2 and to identify novel therapeutic targets for drug development in treating neuroinflammation.
Collapse
Affiliation(s)
- Qianyun Rui
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Shugang Cao
- Department of Neurology, The Second People's Hospital of Hefei, Hefei, Anhui 230011, P.R. China
| | - Xiaozhu Wang
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Xiaoyu Duan
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Xinyi Iao
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Wanli Dong
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Qi Fang
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China.,Suzhou Clinical Medical Center of Neurology, Suzhou, Jiangsu 215004, P.R. China
| | - Xueguang Zhang
- Institute of Clinical Immunology, Jiangsu Key Laboratory of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006 P.R. China
| | - Qun Xue
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China.,Suzhou Clinical Medical Center of Neurology, Suzhou, Jiangsu 215004, P.R. China.,Institute of Clinical Immunology, Jiangsu Key Laboratory of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006 P.R. China
| |
Collapse
|
19
|
Hou Q, Chen H, Liu Q, Yan X. FGF10 Attenuates Experimental Traumatic Brain Injury through TLR4/MyD88/NF-κB Pathway. Cells Tissues Organs 2021; 209:248-256. [PMID: 33440393 DOI: 10.1159/000511381] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 08/27/2020] [Indexed: 11/19/2022] Open
Abstract
Traumatic brain injury (TBI) can induce neuronal apoptosis and neuroinflammation, resulting in substantial neuronal damage and behavioral disorders. Fibroblast growth factors (FGFs) have been shown to be critical mediators in tissue repair. However, the role of FGF10 in experimental TBI remains unknown. In this study, mice with TBI were established via weight-loss model and validated by increase of modified neurological severity scores (mNSS) and brain water content. Secondly, FGF10 levels were elevated in mice after TBI, whereas intraventricular injection of Ad-FGF10 decreased mNSS score and brain water content, indicating the remittance of neurological deficit and cerebral edema in TBI mice. In addition, neuronal damage could also be ameliorated by stereotactic injection of Ad-FGF10. Overexpression of FGF10 increased protein expression of Bcl-2, while it decreased Bax and cleaved caspase-3/PARP, and improved neuronal apoptosis in TBI mice. In addition, Ad-FGF10 relieved neuroinflammation induced by TBI and significantly reduced the level of interleukin 1β/6, tumor necrosis factor α, and monocyte chemoattractant protein-1. Moreover, Ad-FGF10 injection decreased the protein expression level of Toll-like receptor 4 (TLR4), MyD88, and phosphorylation of NF-κB (p-NF-κB), suggesting the inactivation of the TLR4/MyD88/NF-κB pathway. In conclusion, overexpression of FGF10 could ameliorate neurological deficit, neuronal apoptosis, and neuroinflammation through inhibition of the TLR4/MyD88/NF-κB pathway, providing a potential therapeutic strategy for brain injury in the future.
Collapse
Affiliation(s)
- Qinhan Hou
- Department of Neurosurgery, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou City, China
| | - Hongmou Chen
- Department of Neurosurgery, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou City, China,
| | - Quan Liu
- Department of Neurosurgery, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou City, China
| | - Xianlei Yan
- Department of Neurosurgery, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou City, China
| |
Collapse
|
20
|
Hu Y, Sun X, Wang S, Zhou C, Lin L, Ding X, Han J, Zhou Y, Jin G, Wang Y, Zhang W, Shi H, Zhang Z, Yang X, Hua F. Toll-like receptor-2 gene knockout results in neurobehavioral dysfunctions and multiple brain structural and functional abnormalities in mice. Brain Behav Immun 2021; 91:257-266. [PMID: 33069798 DOI: 10.1016/j.bbi.2020.10.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 10/01/2020] [Accepted: 10/06/2020] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVE Toll-like receptor-2 (TLR2), a member of TLR family, plays an important role in the induction and regulation of immune/inflammation. TLR2 gene knockout (TLR2KO) mice have been widely used for animal models of neurological diseases. Since there is close relationship between immune system and neurobehavioral functions, it is important to clarify the exact role of TLR2 defect itself in neurobehavioral functions. The present study aimed to investigate the effect of TLR2KO on neurobehavioral functions in mice and the mechanisms underlying the observed changes. METHODS Male TLR2KO and wild type (WT) mice aged 3, 7, and 12 months were used for neurobehavioral testing and detection of protein expression by Western blot. Brain magnetic resonance imaging (MRI), electrophysiological recording, and Evans blue (EB) assay were applied to evaluate regional cerebral blood flow (rCBF), synaptic function, and blood-brain barrier (BBB) integrity in 12-month-old TLR2KO and age-matched WT mice. RESULTS Compared to WT mice, TLR2KO mice showed decreased cognitive function and locomotor activity, as well as increased anxiety, which developed from middle age (before 7-month-old) to old age. In addition, significantly reduced regional cerebral blood flow (rCBF), inhibited long-term potentiation (LTP), and increased blood-brain barrier (BBB) permeability were observed in 12-month-old TLR2KO mice. Furthermore, compared with age-matched WT mice, significant reduction in protein levels of tight junction proteins (ZO-1, Occludin, and Claudin-5) and increased neurofilament protein (SMI32) were observed in 7 and 12-month-old TLR2KO mice, and that myelin basic protein (MBP) decreased in 12-month-old TLR2KO mice. CONCLUSION Our data demonstrated that TLR2 defect resulted in significantly observable neurobehavioral dysfunctions in mice starting from middle age, as well as multiple abnormalities in brain structure, function, and molecular metabolism.
Collapse
Affiliation(s)
- Yuting Hu
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, China; Institute of Neurological Diseases, Xuzhou Medical University, China
| | - Xiaoyu Sun
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, China; Institute of Neurological Diseases, Xuzhou Medical University, China
| | - Shang Wang
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, China; Institute of Neurological Diseases, Xuzhou Medical University, China
| | - Chao Zhou
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, China; Institute of Neurological Diseases, Xuzhou Medical University, China
| | - Li Lin
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, China; Institute of Neurological Diseases, Xuzhou Medical University, China
| | - Xiaohui Ding
- Department of Histology and Embryology, Shenyang Medical College, China
| | - Jingjing Han
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, China; Institute of Neurological Diseases, Xuzhou Medical University, China
| | - Yan Zhou
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, China; Institute of Neurological Diseases, Xuzhou Medical University, China
| | - Guoliang Jin
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, China; Institute of Neurological Diseases, Xuzhou Medical University, China
| | - Yuqiao Wang
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, China; Institute of Neurological Diseases, Xuzhou Medical University, China
| | - Wei Zhang
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, China; Institute of Neurological Diseases, Xuzhou Medical University, China
| | - Hongjuan Shi
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, China; Institute of Neurological Diseases, Xuzhou Medical University, China
| | - Zuohui Zhang
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, China; Institute of Neurological Diseases, Xuzhou Medical University, China
| | - Xinxin Yang
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, China; Institute of Neurological Diseases, Xuzhou Medical University, China
| | - Fang Hua
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, China; Institute of Neurological Diseases, Xuzhou Medical University, China.
| |
Collapse
|
21
|
Pituitary Adenylate Cyclase-Activating Polypeptide Alleviates Intestinal, Extra-Intestinal and Systemic Inflammatory Responses during Acute Campylobacter jejuni-induced Enterocolitis in Mice. Pathogens 2020; 9:pathogens9100805. [PMID: 33007819 PMCID: PMC7650764 DOI: 10.3390/pathogens9100805] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/15/2020] [Accepted: 09/28/2020] [Indexed: 01/08/2023] Open
Abstract
Human Campylobacter jejuni infections are emerging, and constitute a significant health burden worldwide. The ubiquitously expressed pituitary adenylate cyclase-activating polypeptide (PACAP) is well-known for its cell-protective and immunomodulatory effects. In our actual intervention study, we used an acute campylobacteriosis model and assessed the potential disease-alleviating effects of exogenous PACAP. Therefore, secondary abiotic IL-10-/- mice were perorally infected with C. jejuni and treated with synthetic PACAP38 intraperitoneally from day 2 until day 5 post-infection. Whereas PACAP did not interfere with the gastrointestinal colonization of the pathogen, mice from the PACAP group exhibited less severe clinical signs of C. jejuni-induced disease, as compared to mock controls, which were paralleled by alleviated apoptotic, but enhanced cell proliferative responses in colonic epithelia on day 6 post-infection. Furthermore, PACAP dampened the accumulation of macrophages and monocytes, but enhanced regulatory T cell responses in the colon, which were accompanied by less IFN-γ secretion in intestinal compartments in PACAP versus mock-treated mice. Remarkably, the inflammation-dampening properties of PACAP could also be observed in extra-intestinal organs, and strikingly, even the systemic circulation on day 6 post-infection. For the first time, we provide evidence that synthetic PACAP might be a promising candidate to combat acute campylobacteriosis and post-infectious sequelae.
Collapse
|
22
|
Nonaka N, Banks WA, Shioda S. Pituitary adenylate cyclase-activating polypeptide: Protective effects in stroke and dementia. Peptides 2020; 130:170332. [PMID: 32445876 DOI: 10.1016/j.peptides.2020.170332] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 04/24/2020] [Accepted: 05/18/2020] [Indexed: 01/05/2023]
Abstract
Evidence shows that pituitary adenylate cyclase-activating polypeptide (PACAP) improves stroke outcomes and dementia. The blood-brain barrier (BBB) controls the peptide and regulatory protein exchange between the central nervous system and the blood; the transport of these regulatory substances across the BBB has been altered in animal models of stroke and Alzheimer's disease (AD). PACAP is a powerful neurotrophin that can cross the BBB, which may aid in the therapy of neurodegenerative diseases, including stroke and AD. PACAP may function as a potential drug in the treatment, prevention, or management of stroke and AD and other neurodegenerative conditions. Here, we review the effects of PACAP in studies on stroke and dementias.
Collapse
Affiliation(s)
- Naoko Nonaka
- Department of Oral Anatomy and Developmental Biology, Showa University School of Dentistry, Shinagawa-ku, Tokyo, Japan.
| | - William A Banks
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA.
| | - Seiji Shioda
- Global Research Center for Innovative Life Science, Peptide Drug Innovation, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, Shinagawa-ku, Tokyo, Japan.
| |
Collapse
|
23
|
Korgaonkar AA, Nguyen S, Li Y, Sekhar D, Subramanian D, Guevarra J, Pang KCH, Santhakumar V. Distinct cellular mediators drive the Janus faces of toll-like receptor 4 regulation of network excitability which impacts working memory performance after brain injury. Brain Behav Immun 2020; 88:381-395. [PMID: 32259563 PMCID: PMC7415537 DOI: 10.1016/j.bbi.2020.03.035] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 03/30/2020] [Accepted: 03/31/2020] [Indexed: 01/15/2023] Open
Abstract
The mechanisms by which the neurophysiological and inflammatory responses to brain injury contribute to memory impairments are not fully understood. Recently, we reported that the innate immune receptor, toll-like receptor 4 (TLR4) enhances AMPA receptor (AMPAR) currents and excitability in the dentate gyrus after fluid percussion brain injury (FPI) while limiting excitability in controls. Here, we examine the cellular mediators underlying TLR4 regulation of dentate excitability and its impact on memory performance. In ex vivo slices, astrocytic and microglial metabolic inhibitors selectively abolished TLR4 antagonist modulation of excitability in controls, but not in rats after FPI, demonstrating that glial signaling contributes to TLR4 regulation of excitability in controls. In glia-depleted neuronal cultures from naïve mice, TLR4 ligands bidirectionally modulated AMPAR charge transfer consistent with neuronal TLR4 regulation of excitability, as observed after brain injury. In vivo TLR4 antagonism reduced early post-injury increases in mediators of MyD88-dependent and independent TLR4 signaling without altering expression in controls. Blocking TNFα, a downstream effector of TLR4, mimicked effects of TLR4 antagonist and occluded TLR4 agonist modulation of excitability in slices from both control and FPI rats. Functionally, transiently blocking TLR4 in vivo improved impairments in working memory observed one week and one month after FPI, while the same treatment impaired memory function in uninjured controls. Together these data identify that distinct cellular signaling mechanisms converge on TNFα to mediate TLR4 modulation of network excitability in the uninjured and injured brain and demonstrate a role for TLR4 in regulation of working memory function.
Collapse
Affiliation(s)
- Akshata A. Korgaonkar
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, New Jersey 07103,,Correspondence: Akshata Korgaonkar, PhD, Department of Neurology, Washington University School of Medicine, 660 South Euclid Ave, Campus box 8111, St Louis, MO 63110, Phone (Off): 314.362.2999,
| | - Susan Nguyen
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, California 92521
| | - Ying Li
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, New Jersey 07103
| | - Dipika Sekhar
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, New Jersey 07103,,Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, California 92521
| | - Deepak Subramanian
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, New Jersey 07103,,Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, California 92521
| | - Jenieve Guevarra
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, New Jersey 07103
| | - Kevin C H Pang
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, New Jersey 07103,,Neurobehavioral Research Lab, Department of Veteran Affairs Medical Center–New Jersey Health Care System, East Orange, New Jersey
| | - Vijayalakshmi Santhakumar
- Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, New Jersey 07103,,Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, California 92521
| |
Collapse
|
24
|
Polysialic acid and Siglec-E orchestrate negative feedback regulation of microglia activation. Cell Mol Life Sci 2020; 78:1637-1653. [PMID: 32725371 PMCID: PMC7904730 DOI: 10.1007/s00018-020-03601-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 06/23/2020] [Accepted: 07/17/2020] [Indexed: 11/09/2022]
Abstract
Polysialic acid (polySia) emerges as a novel regulator of microglia activity. We recently identified polysialylated proteins in the Golgi compartment of murine microglia that are released in response to inflammatory stimulation. Since exogenously added polySia is able to attenuate the inflammatory response, we proposed that the release of polysialylated proteins constitutes a mechanism for negative feedback regulation of microglia activation. Here, we demonstrate that translocation of polySia from the Golgi to the cell surface can be induced by calcium depletion of the Golgi compartment and that polysialylated proteins are continuously released for at least 24 h after the onset of inflammatory stimulation. The latter was unexpected, because polySia signals detected by immunocytochemistry are rapidly depleted. However, it indicates that the amount of released polySia is much higher than anticipated based on immunostaining. This may be crucial for microglial responses during traumatic brain injury (TBI), as we detected polySia signals in activated microglia around a stab wound in the adult mouse brain. In BV2 microglia, the putative polySia receptor Siglec-E is internalized during lipopolysaccharide (LPS)-induced activation and in response to polySia exposure, indicating interaction. Correspondingly, CRISPR/Cas9-mediated Siglec-E knockout prevents inhibition of pro inflammatory activation by exogenously added polySia and leads to a strong increase of the LPS response. A comparable increase of LPS-induced activation has been observed in microglia with abolished polySia synthesis. Together, these results indicate that the release of the microglia-intrinsic polySia pool, as implicated in TBI, inhibits the inflammatory response by acting as a trans-activating ligand of Siglec-E.
Collapse
|
25
|
Benemei S, Labastida-Ramírez A, Abramova E, Brunelli N, Caronna E, Diana P, Gapeshin R, Hofacker MD, Maestrini I, Pías EM, Mikulenka P, Tikhonova O, Martelletti P, MaassenVanDenBrink A. Persistent post-traumatic headache: a migrainous loop or not? The preclinical evidence. J Headache Pain 2020; 21:90. [PMID: 32664898 PMCID: PMC7362418 DOI: 10.1186/s10194-020-01135-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 05/25/2020] [Indexed: 12/11/2022] Open
Abstract
Background According to the International Classification of Headache Disorders 3, post-traumatic headache (PTH) attributed to traumatic brain injury (TBI) is a secondary headache reported to have developed within 7 days from head injury, regaining consciousness following the head injury, or discontinuation of medication(s) impairing the ability to sense or report headache following the head injury. It is one of the most common secondary headache disorders, and it is defined as persistent when it lasts more than 3 months. Main body Currently, due to the high prevalence of this disorder, several preclinical studies have been conducted using different animal models of mild TBI to reproduce conditions that engender PTH. Despite representing a simplification of a complex disorder and displaying different limitations concerning the human condition, animal models are still a mainstay to study in vivo the mechanisms of PTH and have provided valuable insight into the pathophysiology and possible treatment strategies. Different models reproduce different types of trauma and have been ideated in order to ensure maximal proximity to the human condition and optimal experimental reproducibility. Conclusion At present, despite its high prevalence, PTH is not entirely understood, and the differential contribution of pathophysiological mechanisms, also observed in other conditions like migraine, has to be clarified. Although facing limitations, animal models are needed to improve understanding of PTH. The knowledge of currently available models is necessary to all researchers who want to investigate PTH and contribute to unravel its mechanisms.
Collapse
Affiliation(s)
- Silvia Benemei
- Health Sciences Department, University of Florence and Headache Centre, Careggi University Hospital, Florence, Italy
| | - Alejandro Labastida-Ramírez
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, Dr Molewaterplein 50, 3015 GE, Rotterdam, The Netherlands
| | - Ekaterina Abramova
- Pain Clinic Unit, Department of Anesthesiology, Pirogov City Clinical Hospital, Moscow, Russia
| | | | - Edoardo Caronna
- Neurology Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Paola Diana
- Child Neuropsychiatry Unit, Department of PROMISE, University of Palermo, Palermo, Italy
| | - Roman Gapeshin
- Department of Neurology and Manual Medicine, Pavlov First Saint-Petersburg State Medical University, Saint-Petersburg, Russia
| | - Maxi Dana Hofacker
- Department of Neurology, Headache Centre, Charité Universitatsmedizin Berlin, Berlin, Germany
| | - Ilaria Maestrini
- Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
| | - Enrique Martínez Pías
- Neurology Department, Hospital Clínico Universitario of Valladolid, Valladolid, Spain
| | - Petr Mikulenka
- Department of Neurology, Third Faculty of Medicine, Charles University and University Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - Olga Tikhonova
- Department of neurology, Kazaryan Clinic of Epileptology and Neurology, Moscow, Russia
| | - Paolo Martelletti
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Antoinette MaassenVanDenBrink
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, Dr Molewaterplein 50, 3015 GE, Rotterdam, The Netherlands.
| | | |
Collapse
|
26
|
Chiu LS, Anderton RS, Clark VW, Cross JL, Knuckey NW, Meloni BP. Effect of Polyarginine Peptide R18D Following a Traumatic Brain Injury in Sprague-Dawley Rats. CURRENT THERAPEUTIC RESEARCH 2020; 92:100584. [PMID: 32322314 PMCID: PMC7163064 DOI: 10.1016/j.curtheres.2020.100584] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 03/10/2020] [Indexed: 11/23/2022]
Abstract
Cationic arginine-rich peptides (CARPs) have previously demonstrated neuroprotective efficacy in a rat model of traumatic brain injury (TBI). The CARP R18D significantly reduced the extent of axonal injury at a high dose of 1000 nmol/kg. Both high (1000 nmol/kg) and low (100 nmol/kg) doses only showed a trend in functional improvement.
Background Despite extensive studies, there are still no clinically available neuroprotective treatments for traumatic brain injury. Objectives In previous studies we demonstrated beneficial treatment effects of polyarginine peptides R18 (18-mer of arginine; 300 nmol/kg) and R18D (18-mer of D-arginine; 1000 nmol/kg) in a rat model of impact-acceleration closed-head injury. Methods We examined the efficacy of R18D when intravenously administered at a low (100 nmol/kg) and high (1000 nmol/kg) dose, 30 minutes after a closed-head injury in male Sprague-Dawley rats. Results At postinjury day 3, treatment with R18D at the high dose significantly reduced axonal injury (P = 0.044), whereas the low-dose treatment of R18D showed a trend for reduced axonal injury. Following assessment in the Barnes maze, both doses of R18D treatment appeared to improve learning and memory recovery compared with vehicle treatment at postinjury days 1 and 3, albeit not to a statistically significant level. Rotarod assessment of vestibulomotor recovery did not differ between R18D and the vehicle treatment groups. Conclusions R18D modestly decreased axonal injury only at the highest dose used but had no significant effect on functional recovery. These findings warrant further studies with additional doses to better understand peptide pharmacodynamics and provide information to guide optimal dosing.
Collapse
Affiliation(s)
- Li Shan Chiu
- Perron Institute for Neurological and Translational Science, Nedlands, Western Australia, Australia
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, Western Australia, Australia
- Address correspondence to: Li Shan Chiu, Perron Institute for Neurological and Translational Sciences, QEII Medical Centre, 8 Verdun St, RR Block, Nedlands, Western Australia, 6009, Australia.
| | - Ryan S. Anderton
- Perron Institute for Neurological and Translational Science, Nedlands, Western Australia, Australia
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, Western Australia, Australia
- School of Heath Sciences, The University Notre Dame Australia, Fremantle, Western Australia, Australia
- Institute for Health Research, The University Notre Dame Australia, Fremantle, Western Australia, Australia
| | - Vince W. Clark
- Perron Institute for Neurological and Translational Science, Nedlands, Western Australia, Australia
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, Western Australia, Australia
- Department of Neurosurgery, Sir Charles Gairdner Hospital, QEII Medical Centre, Nedlands, Western Australia, Australia
| | - Jane L. Cross
- Perron Institute for Neurological and Translational Science, Nedlands, Western Australia, Australia
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, Western Australia, Australia
- Department of Neurosurgery, Sir Charles Gairdner Hospital, QEII Medical Centre, Nedlands, Western Australia, Australia
| | - Neville W. Knuckey
- Perron Institute for Neurological and Translational Science, Nedlands, Western Australia, Australia
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, Western Australia, Australia
- Department of Neurosurgery, Sir Charles Gairdner Hospital, QEII Medical Centre, Nedlands, Western Australia, Australia
| | - Bruno P. Meloni
- Perron Institute for Neurological and Translational Science, Nedlands, Western Australia, Australia
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, Western Australia, Australia
- Department of Neurosurgery, Sir Charles Gairdner Hospital, QEII Medical Centre, Nedlands, Western Australia, Australia
| |
Collapse
|
27
|
Meloni BP, Mastaglia FL, Knuckey NW. Cationic Arginine-Rich Peptides (CARPs): A Novel Class of Neuroprotective Agents With a Multimodal Mechanism of Action. Front Neurol 2020; 11:108. [PMID: 32158425 PMCID: PMC7052017 DOI: 10.3389/fneur.2020.00108] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 01/30/2020] [Indexed: 12/17/2022] Open
Abstract
There are virtually no clinically available neuroprotective drugs for the treatment of acute and chronic neurological disorders, hence there is an urgent need for the development of new neuroprotective molecules. Cationic arginine-rich peptides (CARPs) are an expanding and relatively novel class of compounds, which possess intrinsic neuroprotective properties. Intriguingly, CARPs possess a combination of biological properties unprecedented for a neuroprotective agent including the ability to traverse cell membranes and enter the CNS, antagonize calcium influx, target mitochondria, stabilize proteins, inhibit proteolytic enzymes, induce pro-survival signaling, scavenge toxic molecules, and reduce oxidative stress as well as, having a range of anti-inflammatory, analgesic, anti-microbial, and anti-cancer actions. CARPs have also been used as carrier molecules for the delivery of other putative neuroprotective agents across the blood-brain barrier and blood-spinal cord barrier. However, there is increasing evidence that the neuroprotective efficacy of many, if not all these other agents delivered using a cationic arginine-rich cell-penetrating peptide (CCPPs) carrier (e.g., TAT) may actually be mediated largely by the properties of the carrier molecule, with overall efficacy further enhanced according to the amino acid composition of the cargo peptide, in particular its arginine content. Therefore, in reviewing the neuroprotective mechanisms of action of CARPs we also consider studies using CCPPs fused to a putative neuroprotective peptide. We review the history of CARPs in neuroprotection and discuss in detail the intrinsic biological properties that may contribute to their cytoprotective effects and their usefulness as a broad-acting class of neuroprotective drugs.
Collapse
Affiliation(s)
- Bruno P Meloni
- Department of Neurosurgery, QEII Medical Centre, Sir Charles Gairdner Hospital, Nedlands, WA, Australia.,Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, WA, Australia
| | - Frank L Mastaglia
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, WA, Australia
| | - Neville W Knuckey
- Department of Neurosurgery, QEII Medical Centre, Sir Charles Gairdner Hospital, Nedlands, WA, Australia.,Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.,Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|
28
|
Korgaonkar AA, Li Y, Sekhar D, Subramanian D, Guevarra J, Swietek B, Pallottie A, Singh S, Kella K, Elkabes S, Santhakumar V. Toll-like Receptor 4 Signaling in Neurons Enhances Calcium-Permeable α-Amino-3-Hydroxy-5-Methyl-4-Isoxazolepropionic Acid Receptor Currents and Drives Post-Traumatic Epileptogenesis. Ann Neurol 2020; 87:497-515. [PMID: 32031699 DOI: 10.1002/ana.25698] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 02/03/2020] [Accepted: 02/05/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Traumatic brain injury is a major risk factor for acquired epilepsies, and understanding the mechanisms underlying the early pathophysiology could yield viable therapeutic targets. Growing evidence indicates a role for inflammatory signaling in modifying neuronal excitability and promoting epileptogenesis. Here we examined the effect of innate immune receptor Toll-like receptor 4 (TLR4) on excitability of the hippocampal dentate gyrus and epileptogenesis after brain injury. METHODS Slice and in vivo electrophysiology and Western blots were conducted in rats subject to fluid percussion brain injury or sham injury. RESULTS The studies identify that TLR4 signaling in neurons augments dentate granule cell calcium-permeable α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor (CP-AMPAR) currents after brain injury. Blocking TLR4 signaling in vivo shortly after brain injury reduced dentate network excitability and seizure susceptibility. When blocking of TLR4 signaling after injury was delayed, however, this treatment failed to reduce postinjury seizure susceptibility. Furthermore, TLR4 signal blocking was less efficacious in limiting seizure susceptibility when AMPAR currents, downstream targets of TLR4 signaling, were transiently enhanced. Paradoxically, blocking TLR4 signaling augmented both network excitability and seizure susceptibility in uninjured controls. Despite the differential effect on seizure susceptibility, TLR4 antagonism suppressed cellular inflammatory responses after injury without impacting sham controls. INTERPRETATION These findings demonstrate that independently of glia, the immune receptor TLR4 directly regulates post-traumatic neuronal excitability. Moreover, the TLR4-dependent early increase in dentate excitability is causally associated with epileptogenesis. Identification and selective targeting of the mechanisms underlying the aberrant TLR4-mediated increase in CP-AMPAR signaling after injury may prevent epileptogenesis after brain trauma. ANN NEUROL 2020;87:497-515.
Collapse
Affiliation(s)
- Akshata A Korgaonkar
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ
| | - Ying Li
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ
| | - Dipika Sekhar
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ.,Department of Molecular, Cell, and Systems Biology, University of California Riverside, Riverside, CA
| | - Deepak Subramanian
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ.,Department of Molecular, Cell, and Systems Biology, University of California Riverside, Riverside, CA
| | - Jenieve Guevarra
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ
| | - Bogumila Swietek
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ
| | - Alexandra Pallottie
- Department of Neurological Surgery, Rutgers New Jersey Medical School, Newark, NJ
| | - Sukwinder Singh
- Department of Pathology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ
| | - Kruthi Kella
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ
| | - Stella Elkabes
- Department of Neurological Surgery, Rutgers New Jersey Medical School, Newark, NJ
| | - Vijayalakshmi Santhakumar
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ.,Department of Molecular, Cell, and Systems Biology, University of California Riverside, Riverside, CA
| |
Collapse
|
29
|
Zhou Z, Hou J, Mo Y, Ren M, Yang G, Qu Z, Hu Y. Geniposidic acid ameliorates spatial learning and memory deficits and alleviates neuroinflammation via inhibiting HMGB-1 and downregulating TLR4/2 signaling pathway in APP/PS1 mice. Eur J Pharmacol 2020; 869:172857. [DOI: 10.1016/j.ejphar.2019.172857] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 11/26/2019] [Accepted: 12/09/2019] [Indexed: 12/11/2022]
|
30
|
The Neuroprotective and Biomarker Potential of PACAP in Human Traumatic Brain Injury. Int J Mol Sci 2020; 21:ijms21030827. [PMID: 32012887 PMCID: PMC7037866 DOI: 10.3390/ijms21030827] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/14/2020] [Accepted: 01/24/2020] [Indexed: 02/07/2023] Open
Abstract
Traumatic brain injury remains a growing public health concern and represents the greatest contributor to death and disability globally among all trauma-related injuries. There are limited clinical data regarding biomarkers in the diagnosis and outcome prediction of TBI. The lack of real effective treatment for recovery calls for research of TBI to be shifted into the area of prevention, treatment of secondary brain injury and neurorehabilitation. The neuropeptide pituitary adenylate cyclase activating polypeptide (PACAP) has been reported to act as a hormone, a neuromodulator, a neurotransmitter and a trophic factor, and has been implicated in a variety of developmental and regenerative processes. The importance of PACAP in neuronal regeneration lies in the upregulation of endogenous PACAP and its receptors and the protective effect of exogenous PACAP after different central nervous system injury. The aim of this minireview is to summarize both the therapeutic and biomarker potential of the neuropeptide PACAP, as a novel possible target molecule presently being investigated in several human conditions including TBI, and with encouraging results in animal models of TBI.
Collapse
|
31
|
Avila JA, Kiprowska M, Jean-Louis T, Rockwell P, Figueiredo-Pereira ME, Serrano PA. PACAP27 mitigates an age-dependent hippocampal vulnerability to PGJ2-induced spatial learning deficits and neuroinflammation in mice. Brain Behav 2020; 10:e01465. [PMID: 31769222 PMCID: PMC6955932 DOI: 10.1002/brb3.1465] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 08/20/2019] [Accepted: 10/13/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Inflammation in the brain is mediated by the cyclooxygenase pathway, which leads to the production of prostaglandins. Prostaglandin (PG) D2, the most abundant PG in the brain, increases under pathological conditions and is spontaneously metabolized to PGJ2. PGJ2 is highly neurotoxic, with the potential to transition neuroinflammation into a chronic state and contribute to neurodegeneration as seen in many neurological diseases. Conversely, PACAP27 is a lipophilic peptide that raises intracellular cAMP and is an anti-inflammatory agent. The aim of our study was to investigate the therapeutic potential of PACAP27 to counter the behavioral and neurotoxic effects of PGJ2 observed in aged subjects. METHODS PGJ2 was injected bilaterally into the hippocampal CA1 region of 53-week-old and 12-week-old C57BL/6N male mice, once per week over 3 weeks (three total infusions) and included co-infusions of PACAP27 within respective treatment groups. Our behavioral assessments looked at spatial learning and memory performance on the 8-arm radial maze, followed by histological analyses of fixed hippocampal tissue using Fluoro-Jade C and fluorescent immunohistochemistry focused on IBA-1 microglia. RESULTS Aged mice treated with PGJ2 exhibited spatial learning and long-term memory deficits, as well as neurodegeneration in CA3 pyramidal neurons. Aged mice that received co-infusions of PACAP27 exhibited remediated learning and memory performance and decreased neurodegeneration in CA3 pyramidal neurons. Moreover, microglial activation in the CA3 region was also reduced in aged mice cotreated with PACAP27. CONCLUSIONS Our data show that PGJ2 can produce a retrograde spread of damage not observed in PGJ2-treated young mice, leading to age-dependent neurodegeneration of hippocampal neurons producing learning and memory deficits. PACAP27 can remediate the behavioral and neurodegenerative effects that PGJ2 produces in aged subjects. Targeting specific neurotoxic prostaglandins, such as PGJ2, offers great promise as a new therapeutic strategy downstream of cyclooxygenases, to combat the neuronal deficits induced by chronic inflammation.
Collapse
Affiliation(s)
- Jorge A Avila
- Department of Psychology, Hunter College, City University of New York, New York, NY, USA.,The Graduate Center of CUNY, New York, NY, USA
| | - Magdalena Kiprowska
- The Graduate Center of CUNY, New York, NY, USA.,Department of Biological Sciences, Hunter College, City University of New York, New York, NY, USA
| | - Teneka Jean-Louis
- The Graduate Center of CUNY, New York, NY, USA.,Department of Biological Sciences, Hunter College, City University of New York, New York, NY, USA
| | - Patricia Rockwell
- The Graduate Center of CUNY, New York, NY, USA.,Department of Biological Sciences, Hunter College, City University of New York, New York, NY, USA
| | - Maria E Figueiredo-Pereira
- The Graduate Center of CUNY, New York, NY, USA.,Department of Biological Sciences, Hunter College, City University of New York, New York, NY, USA
| | - Peter A Serrano
- Department of Psychology, Hunter College, City University of New York, New York, NY, USA.,The Graduate Center of CUNY, New York, NY, USA
| |
Collapse
|
32
|
Hermann JK, Capadona JR. Understanding the Role of Innate Immunity in the Response to Intracortical Microelectrodes. Crit Rev Biomed Eng 2019; 46:341-367. [PMID: 30806249 DOI: 10.1615/critrevbiomedeng.2018027166] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Intracortical microelectrodes exhibit enormous potential for researching the nervous system, steering assistive devices and functional electrode stimulation systems for severely paralyzed individuals, and augmenting the brain with computing power. Unfortunately, intracortical microelectrodes often fail to consistently record signals over clinically useful periods. Biological mechanisms, such as the foreign body response to intracortical microelectrodes and self-perpetuating neuroinflammatory cascades, contribute to the inconsistencies and decline in recording performance. Unfortunately, few studies have directly correlated microelectrode performance with the neuroinflammatory response to the implanted devices. However, of those select studies that have, the role of the innate immune system remains among the most likely links capable of corroborating the results of different studies, across laboratories. Therefore, the overall goal of this review is to highlight the role of innate immunity signaling in the foreign body response to intracortical microelectrodes and hypothesize as to appropriate strategies that may become the most relevant in enabling brain-dwelling electrodes of any geometry, or location, for a range of clinical applications.
Collapse
Affiliation(s)
- John K Hermann
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr. Drive, Wickenden Bldg, Cleveland, OH 44106; Advanced Platform Technology Center, Rehabilitation Research and Development, Louis Stokes Cleveland VA Medical Center, 10701 East Blvd. Mail Stop 151 AW/APT, Cleveland, OH 44106-1702
| | - Jeffrey R Capadona
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr. Drive, Wickenden Bldg, Cleveland, OH 44106; Advanced Platform Technology Center, Rehabilitation Research and Development, Louis Stokes Cleveland VA Medical Center, 10701 East Blvd. Mail Stop 151 AW/APT, Cleveland, OH 44106-1702
| |
Collapse
|
33
|
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: 3.6] [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
|
34
|
Bereswill S, Escher U, Grunau A, Kühl AA, Dunay IR, Tamas A, Reglodi D, Heimesaat MM. Pituitary Adenylate Cyclase-Activating Polypeptide-A Neuropeptide as Novel Treatment Option for Subacute Ileitis in Mice Harboring a Human Gut Microbiota. Front Immunol 2019; 10:554. [PMID: 30967875 PMCID: PMC6438926 DOI: 10.3389/fimmu.2019.00554] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 03/01/2019] [Indexed: 12/21/2022] Open
Abstract
The neuropeptide Pituitary adenylate cyclase-activating polypeptide (PACAP) is well-known for its important functions in immunity and inflammation. Data regarding anti-inflammatory properties of PACAP in the intestinal tract are limited, however. In our present preclinical intervention study we addressed whether PACAP treatment could alleviate experimental subacute ileitis mimicking human gut microbiota conditions. Therefore, secondary abioitic mice were subjected to human fecal microbiota transplantation (FMT) and perorally infected with low-dose Toxoplasma gondii to induce subacute ileitis on day 0. From day 3 until day 8 post-infection, mice were either treated with synthetic PACAP38 or placebo. At day 9 post-infection, placebo, but not PACAP treated mice exhibited overt macroscopic sequelae of intestinal immunopathology. PACAP treatment further resulted in less distinct apoptotic responses in ileal and colonic epithelia that were accompanied by lower T cell numbers in the mucosa and lamina propria and less secretion of pro-inflammatory cytokines in intestinal ex vivo biopsies. Notably, ileitis-associated gut microbiota shifts were less distinct in PACAP as compared to placebo treated mice. Inflammation-ameliorating effects of PACAP were not restricted to the intestines, but could also be observed in extra-intestinal including systemic compartments as indicated by lower apoptotic cell counts and less pro-inflammatory cytokine secretion in liver and lungs taken from PACAP treated as compared to placebo control mice, which also held true for markedly lower serum TNF and IL-6 concentrations in the former as compared to the latter. Our preclinical intervention study provides strong evidence that synthetic PACAP alleviates subacute ileitis and extra-intestinal including systemic sequelae of T cell-driven immunopathology. These findings further support PACAP as a novel treatment option for intestinal inflammation including inflammatory bowel diseases (IBD).
Collapse
Affiliation(s)
- Stefan Bereswill
- Department of Microbiology, Infectious Diseases, and Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ulrike Escher
- Department of Microbiology, Infectious Diseases, and Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Anne Grunau
- Department of Microbiology, Infectious Diseases, and Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Anja A Kühl
- Department of Medicine I for Gastroenterology, Infectious Diseases and Rheumatology/Research Center ImmunoSciences (RCIS), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ildiko R Dunay
- Medical Faculty, Institute of Inflammation and Neurodegeneration, University Hospital Magdeburg, Magdeburg, Germany
| | - Andrea Tamas
- Department of Anatomy, MTA-PTE PACAP Research Team, Centre for Neuroscience, University of Pecs Medical School, Pecs, Hungary
| | - Dora Reglodi
- Department of Anatomy, MTA-PTE PACAP Research Team, Centre for Neuroscience, University of Pecs Medical School, Pecs, Hungary
| | - Markus M Heimesaat
- Department of Microbiology, Infectious Diseases, and Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
35
|
Kumar A, Henry RJ, Stoica BA, Loane DJ, Abulwerdi G, Bhat SA, Faden AI. Neutral Sphingomyelinase Inhibition Alleviates LPS-Induced Microglia Activation and Neuroinflammation after Experimental Traumatic Brain Injury. J Pharmacol Exp Ther 2019; 368:338-352. [PMID: 30563941 PMCID: PMC6367691 DOI: 10.1124/jpet.118.253955] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 12/14/2018] [Indexed: 12/17/2022] Open
Abstract
Neuroinflammation is one of the key secondary injury mechanisms triggered by traumatic brain injury (TBI). Microglial activation, a hallmark of brain neuroinflammation, plays a critical role in regulating immune responses after TBI and contributes to progressive neurodegeneration and neurologic deficits following brain trauma. Here we evaluated the role of neutral sphingomyelinase (nSMase) in microglial activation by examining the effects of the nSMase inhibitors altenusin and GW4869 in vitro (using BV2 microglia cells and primary microglia), as well as in a controlled cortical injury (CCI) model in adult male C57BL/6 mice. Pretreatment of altenusin or GW4869 prior to lipopolysaccharide (LPS) stimulation for 4 or 24 hours, significantly downregulated gene expression of the pro-inflammatory mediators TNF-α, IL-1β, IL-6, iNOS, and CCL2 in microglia and reduced the release of nitric oxide and TNF-α These nSMase inhibitors also attenuated the release of microparticles and phosphorylation of p38 MAPK and ERK1/2. In addition, altenusin pretreatment also reduced the gene expression of multiple inflammatory markers associated with microglial activation after experimental TBI, including TNF-α, IL-1β, IL-6, iNOS, CCL2, CD68, NOX2, and p22phox Overall, our data demonstrate that nSMase inhibitors attenuate multiple inflammatory pathways associated with microglial activation in vitro and after experimental TBI. Thus, nSMase inhibitors may represent promising therapeutics agents targeting neuroinflammation.
Collapse
Affiliation(s)
- Asit Kumar
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Rebecca J Henry
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Bogdan A Stoica
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - David J Loane
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Gelareh Abulwerdi
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Shahnawaz A Bhat
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Alan I Faden
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
36
|
Hagos FT, Adams SM, Poloyac SM, Kochanek PM, Horvat CM, Clark RSB, Empey PE. Membrane transporters in traumatic brain injury: Pathological, pharmacotherapeutic, and developmental implications. Exp Neurol 2019; 317:10-21. [PMID: 30797827 DOI: 10.1016/j.expneurol.2019.02.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 02/12/2019] [Accepted: 02/20/2019] [Indexed: 12/12/2022]
Abstract
Membrane transporters regulate the trafficking of endogenous and exogenous molecules across biological barriers and within the neurovascular unit. In traumatic brain injury (TBI), they moderate the dynamic movement of therapeutic drugs and injury mediators among neurons, endothelial cells and glial cells, thereby becoming important determinants of pathogenesis and effective pharmacotherapy after TBI. There are three ways transporters may impact outcomes in TBI. First, transporters likely play a key role in the clearance of injury mediators. Second, genetic association studies suggest transporters may be important in the transition of TBI from acute brain injury to a chronic neurological disease. Third, transporters dynamically control the brain penetration and efflux of many drugs and their distribution within and elimination from the brain, contributing to pharmacoresistance and possibly in some cases pharmacosensitivity. Understanding the nature of drugs or candidate drugs in development with respect to whether they are a transporter substrate or inhibitor is relevant to understand whether they distribute to their target in sufficient concentrations. Emerging data provide evidence of altered expression and function of transporters in humans after TBI. Genetic variability in expression and/or function of key transporters adds an additional dynamic, as shown in recent clinical studies. In this review, evidence supporting the role of individual membrane transporters in TBI are discussed as well as novel strategies for their modulation as possible therapeutic targets. Since data specifically targeting pediatric TBI are sparse, this review relies mainly on experimental studies using adult animals and clinical studies in adult patients.
Collapse
Affiliation(s)
- Fanuel T Hagos
- Center for Clinical Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, PA, United States of America
| | - Solomon M Adams
- Center for Clinical Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, PA, United States of America
| | - Samuel M Poloyac
- Center for Clinical Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, PA, United States of America; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Patrick M Kochanek
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States of America; Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America; UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, United States of America
| | - Christopher M Horvat
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States of America; Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America; UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, United States of America
| | - Robert S B Clark
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States of America; Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America; UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, United States of America.
| | - Philip E Empey
- Center for Clinical Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, PA, United States of America; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States of America.
| |
Collapse
|
37
|
Chiu LS, Anderton RS, Cross JL, Clark VW, Knuckey NW, Meloni BP. Poly-arginine Peptide R18D Reduces Neuroinflammation and Functional Deficits Following Traumatic Brain Injury in the Long-Evans Rat. Int J Pept Res Ther 2019. [DOI: 10.1007/s10989-018-09799-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
38
|
Younger D, Murugan M, Rama Rao KV, Wu LJ, Chandra N. Microglia Receptors in Animal Models of Traumatic Brain Injury. Mol Neurobiol 2018; 56:5202-5228. [DOI: 10.1007/s12035-018-1428-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/13/2018] [Indexed: 02/07/2023]
|
39
|
Wang W, Wang J. Toll-Like Receptor 4 (TLR4)/Cyclooxygenase-2 (COX-2) Regulates Prostate Cancer Cell Proliferation, Migration, and Invasion by NF-κB Activation. Med Sci Monit 2018; 24:5588-5597. [PMID: 30098292 PMCID: PMC6180953 DOI: 10.12659/msm.906857] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Background Toll-like receptor 4 (TLR4)-mediated signaling has been implicated in invasion, metastasis, and survival of various cancers. Activation of TLR4 can promote cyclooxygenase-2 (COX-2) and nuclear factor-κB (NF-κB). However, little is known about the effects of TLR4/COX-2 in prostate cancer (PCa). Material/Methods In our study, TLR4 and COX-2 expressions were detected by quantitative real-time reverse transcription PCR (qRT-PCR) in PCa tissues (n=34). Cell proliferation was measured by Cell Counting Kit-8 (CCK-8) and carboxyfluorescein succinimidyl ester (CFSE) assays. The migration and invasion abilities were detected by wound healing and Transwell assays. qRT-PCR and western blot assays were performed to detect TLR4, COX-2, matrix metalloproteinase (MMP)-2, MMP-9, tissue inhibitor of matrix metalloproteinases (TIMP)-1, epithelial-cadherin (E-cadherin), vimentin, NF-κB (p65), and p-p65 expressions. Results The results revealed that TLR4 and COX-2 were upregulated in PCa tissues; Silencing of TLR4 or COX-2 inhibited PCa cell proliferation, migration, and invasion, and TLR4 siRNAs combined with COX-2 siRNAs synergistically suppressed PCa cell proliferation, migration, and invasion. Silencing of TLR4 or COX-2 also downregulated MMP-2, MMP-9, and E-cadherin expressions, and upregulated TIMP-1 and vimentin expressions. In addition, silencing of TLR4 or COX-2 inhibited p65 phosphorylation and had a synergistic effect. Conclusions We demonstrated that TLR4/COX-2 inhibits PCa cell proliferation, migration, and invasion by regulating NF-κB.
Collapse
Affiliation(s)
- Wei Wang
- Department of Urology Surgery, Tiantai People's Hospital, Taizhou, Zhejiang, China (mainland)
| | - Jiye Wang
- Department of Urology Surgery, Tiantai People's Hospital, Taizhou, Zhejiang, China (mainland)
| |
Collapse
|
40
|
Shi H, Hua X, Kong D, Stein D, Hua F. Role of Toll-like receptor mediated signaling in traumatic brain injury. Neuropharmacology 2018; 145:259-267. [PMID: 30075158 DOI: 10.1016/j.neuropharm.2018.07.022] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 07/04/2018] [Accepted: 07/18/2018] [Indexed: 12/13/2022]
Abstract
The mechanisms underlying secondary brain damage following traumatic brain injury (TBI) remain unclear. A great many studies have demonstrated that inflammatory cascades contribute to brain damage through the activation of immune/inflammatory responses, including the increased release of cytokines and chemokines, and the recruitment of leukocytes. The cells and tissues damaged by primary mechanical injury release a number of endogenous factors acting as damage-associated molecular patterns (DAMPs), which initiate and perpetuate noninfectious inflammatory responses through transduction signaling pathways. Toll-like receptors (TLRs) are a transmembrane receptor family that can recognize the specific DAMPs released from damaged cells and recruit a set of adaptors leading to the activation of downstream kinases and nuclear factors which regulate the expression of inflammatory genes. The activation of inflammatory responses mediated by TLR signaling is closely associated with brain tissue damage and neurological dysfunction following TBI. TLRs and their downstream protein kinases may be potential targets for the treatment of TBI. Modulation of TLR-mediated signaling may attenuate brain damage and improve TBI outcome. In this review, we briefly discuss the role of TLR-mediated signaling in TBI and the new treatments targeting TLR signaling. This article is part of the Special Issue entitled "Novel Treatments for Traumatic Brain Injury".
Collapse
Affiliation(s)
- Hongjuan Shi
- Department of Neurology, The Affiliated Hospital, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Xiaodong Hua
- Augusta University/University of Georgia Medical Partnership, Athens, GA, 30606, USA; Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Delian Kong
- Department of Neurology, The Affiliated Hospital, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Donald Stein
- Brain Research Laboratory, Department of Emergency Medicine, Emory University School of Medicine, Atlanta, GA, 30032, USA
| | - Fang Hua
- Department of Neurology, The Affiliated Hospital, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China; Key Laboratory of Anesthesiology of Jiangsu Province, Xuzhou, 221002, China.
| |
Collapse
|
41
|
Li QX, Shen YX, Ahmad A, Shen YJ, Zhang YQ, Xu PK, Chen WW, Yu YQ. Mesencephalic Astrocyte-Derived Neurotrophic Factor Prevents Traumatic Brain Injury in Rats by Inhibiting Inflammatory Activation and Protecting the Blood-Brain Barrier. World Neurosurg 2018; 117:e117-e129. [PMID: 29883817 DOI: 10.1016/j.wneu.2018.05.202] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 05/26/2018] [Accepted: 05/28/2018] [Indexed: 10/14/2022]
Abstract
BACKGROUND Our previous studies have shown that mesencephalic astrocyte-derived neurotrophic factor (MANF) provides a neuroprotective effect against ischemia/reperfusion injury and is also involved in inflammatory disease models. This study investigates the potential role and mechanism of MANF in acute brain damage after traumatic brain injury (TBI). METHODS The model of TBI was induced by Feeney free falling methods with male Sprague-Dawley rats. The expression of MANF, 24 hours after TBI, was detected by the immunohistochemistry, immunofluorescence, Western blot, and reverse transcription polymerase chain reaction techniques. After treatment with recombinant human MANF after TBI, assessment was conducted 24 hours later for brain water content, cerebral edema volume in magnetic resonance imaging, neurobehavioral testing, and Evans blue extravasation. Moreover, by the techniques of Western blot and reverse transcription polymerase chain reaction, the expression of inflammatory cytokines (interleukin 1β and tumor necrosis factor α) and P65 was also analyzed to explore the underlying protective mechanism of MANF. RESULTS At 24 hours after TBI, we found that endogenous MANF was widely expressed in the rat's brain tissues and different types of cells. Treatment with a high dose of recombinant human MANF (20 μg/20 μL) significantly increased the modified Garcia score, and reduced brain water content as well as cerebral edema volume on magnetic resonance imaging. Furthermore, MANF alleviated not only the permeability of the blood-brain barrier (BBB) but also the expressions of interleukin 1β and tumor necrosis factor α messenger RNA and protein. Besides, the activation of P65 was also inhibited. CONCLUSIONS These results suggest that MANF provides a neuroprotective effect against acute brain injury after TBI, via attenuating blood-brain barrier disruption and intracranial neuroinflammation; the inhibition of the NF-κB signaling pathway might be a potential mechanism.
Collapse
Affiliation(s)
- Qing-Xin Li
- The First Affiliated Hospital, Anhui Medical University, Hefei, People's Republic of China
| | - Yu-Xian Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, People's Republic of China; Biopharmaceutical Institute, Anhui Medical University, Hefei, People's Republic of China
| | - Akhlaq Ahmad
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, People's Republic of China
| | - Yu-Jun Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, People's Republic of China; Biopharmaceutical Institute, Anhui Medical University, Hefei, People's Republic of China
| | - Yi-Quan Zhang
- The First Affiliated Hospital, Anhui Medical University, Hefei, People's Republic of China
| | - Pei-Kun Xu
- The First Affiliated Hospital, Anhui Medical University, Hefei, People's Republic of China
| | - Wei-Wei Chen
- The First Affiliated Hospital, Anhui Medical University, Hefei, People's Republic of China
| | - Yong-Qiang Yu
- The First Affiliated Hospital, Anhui Medical University, Hefei, People's Republic of China.
| |
Collapse
|
42
|
Wang W, Zinsmaier AK, Firestone E, Lin R, Yatskievych TA, Yang S, Zhang J, Bao S. Blocking Tumor Necrosis Factor-Alpha Expression Prevents Blast-Induced Excitatory/Inhibitory Synaptic Imbalance and Parvalbumin-Positive Interneuron Loss in the Hippocampus. J Neurotrauma 2018; 35:2306-2316. [PMID: 29649942 DOI: 10.1089/neu.2018.5688] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Traumatic brain injury (TBI) is a major cause of neurological disorder and death in civilian and military populations. It comprises two components-direct injury from the traumatic impact and secondary injury from ensuing neural inflammatory responses. Blocking tumor necrosis factor-alpha (TNF-α), a central regulator of neural inflammation, has been shown to improve functional recovery after TBI. However, the mechanisms underlying those therapeutic effects are still poorly understood. Here, we examined effects of 3,6'-dithiothalidomide (dTT), a potentially therapeutic TNF-α inhibitor, in mice with blast-induced TBI. We found that blast exposure resulted in elevated expression of TNF-α, activation of microglial cells, enhanced excitatory synaptic transmission, reduced inhibitory synaptic transmission, and a loss of parvalbumin-positive (PV+) inhibitory interneurons. Administration of dTT for 5 days after the blast exposure completely suppressed blast-induced increases in TNF-α transcription, largely reversed blasted-induced synaptic changes, and prevented PV+ neuron loss. However, blocking TNF-α expression by dTT failed to mitigate blast-induced microglial activation in the hippocampus, as evidenced by their non-ramified morphology. These results indicate that TNF-α plays a major role in modulating neuronal functions in blast-induced TBI and that it is a potential target for treatment of TBI-related brain disorders.
Collapse
Affiliation(s)
- Weihua Wang
- 1 Department of Physiology, College of Medicine, University of Arizona , Tucson, Arizona
| | - Alexander K Zinsmaier
- 1 Department of Physiology, College of Medicine, University of Arizona , Tucson, Arizona
| | - Ethan Firestone
- 2 Department of Otolaryngology-Head and Neck Surgery and Department of Communication Sciences and Disorders, School of Medicine, Wayne State University , Detroit, Michigan
| | - Ruizhu Lin
- 1 Department of Physiology, College of Medicine, University of Arizona , Tucson, Arizona.,3 Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University , Guangzhou, China
| | - Tatiana A Yatskievych
- 1 Department of Physiology, College of Medicine, University of Arizona , Tucson, Arizona
| | - Sungchil Yang
- 4 Department of Biomedical Sciences, City University of Hong Kong , Kowloon, Hong Kong, China
| | - Jinsheng Zhang
- 2 Department of Otolaryngology-Head and Neck Surgery and Department of Communication Sciences and Disorders, School of Medicine, Wayne State University , Detroit, Michigan
| | - Shaowen Bao
- 1 Department of Physiology, College of Medicine, University of Arizona , Tucson, Arizona
| |
Collapse
|
43
|
Kamins J, Charles A. Posttraumatic Headache: Basic Mechanisms and Therapeutic Targets. Headache 2018; 58:811-826. [DOI: 10.1111/head.13312] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 03/28/2018] [Accepted: 03/29/2018] [Indexed: 12/16/2022]
Affiliation(s)
- Joshua Kamins
- UCLA Goldberg Migraine Program; David Geffen School of Medicine at UCLA; Los Angeles CA USA
- Tisch Brainsport Program; David Geffen School of Medicine at UCLA; Los Angeles CA USA
| | - Andrew Charles
- UCLA Goldberg Migraine Program; David Geffen School of Medicine at UCLA; Los Angeles CA USA
| |
Collapse
|
44
|
PENG Y, XU M, HUANG G, ZHANG H, AI K. Effect of electroacupuncture on urodynamics, PACAP-38 and PAC1R of spinal cord in rats with neurogenic bladder ✰. WORLD JOURNAL OF ACUPUNCTURE-MOXIBUSTION 2018. [DOI: 10.1016/j.wjam.2018.03.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
45
|
Rhea EM, Bullock KM, Banks WA. Effect of controlled cortical impact on the passage of pituitary adenylate cyclase activating polypeptide (PACAP) across the blood-brain barrier. Peptides 2018; 99:8-13. [PMID: 29107653 PMCID: PMC5756113 DOI: 10.1016/j.peptides.2017.10.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 10/21/2017] [Accepted: 10/23/2017] [Indexed: 01/20/2023]
Abstract
Injuries to the central nervous system can affect the blood-brain barrier (BBB), including disruption and influencing peptide transport across the BBB. Pituitary adenylate cyclase-activating polypeptide 38 (PACAP38) is a potent neurotrophic and neuroprotective peptide currently being investigated for its therapeutic role following injury to the central nervous system and can cross the BBB in a saturable manner. The goal of the current study was to investigate for the first time PACAP38 uptake by the brain following traumatic brain injury (TBI). Using radioactively labeled PACAP38, we measured the levels of PACAP38 present in the injured, ipsilateral cortex in Sham-treated mice compared to mice receiving a controlled cortical impact (CCI), a model of TBI. Experiments were conducted at 6 different time points (from 2h up to 4 weeks) following CCI to determine temporal changes in PACAP38 transport. PACAP38 uptake was increased at 2 and 72h post-CCI compared to Sham. We did not detect changes in PACAP38 uptake in the contralateral cortex and cerebellum between Sham and CCI-treatment. The rate of PACAP38 transport into the ipsilateral cortex following CCI was increased 3.6-fold 72h after compared to 2h post-CCI. In addition, the rate of transport into the cerebellum was greater than that of the cortices. The data presented here shows PACAP38 transport is temporally altered following CCI-treatment and PACAP38 uptake is greater in the cerebellum compared to the cortices.
Collapse
Affiliation(s)
- Elizabeth M Rhea
- Division of Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA, United States; Geriatric Research and Education Clinical Center, VA Puget Sound, Seattle, WA, United States
| | - Kristin M Bullock
- Geriatric Research and Education Clinical Center, VA Puget Sound, Seattle, WA, United States
| | - William A Banks
- Division of Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA, United States; Geriatric Research and Education Clinical Center, VA Puget Sound, Seattle, WA, United States
| |
Collapse
|
46
|
Tang Y, Zhou G, Yao L, Xue P, Yu D, Xu R, Shi W, Yao X, Yan Z, Duan JA. Protective effect of Ginkgo biloba leaves extract, EGb761, on myocardium injury in ischemia reperfusion rats via regulation of TLR-4/NF-κB signaling pathway. Oncotarget 2017; 8:86671-86680. [PMID: 29156826 PMCID: PMC5689716 DOI: 10.18632/oncotarget.21372] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Accepted: 08/29/2017] [Indexed: 11/25/2022] Open
Abstract
Beneficial actions of EGb 761 against ischemia/reperfusion (I/R) injury in lung, brain and renal ischemia have been described. However, the relationship between EGb 761 and signal molecules in myocardial ischemia reperfusion has not been well elucidated. In this study, we investigated the effects and mechanism of EGb 761 preconditioning on anti-myocardial I/R injuries in vivo. Meanwhile, their potential anti-oxidative stress and anti-inflammation effect were assessed. Hemodynamic parameters were monitored as left ventricular systolic pressure, LV end-diastolic pressure and maximal rate of increase and decrease of left ventricular pressure (dP/dtmax). The oxidative stress indicators and inflammatory factors were also evaluated. Western blot method was used for analysis of toll-like receptor 4 (TLR4), p-TLR4, nuclear factor-κB (NF-κB), p-NF-κB p65, Bax and Bcl-2 protein expressions. EGb 761 significantly improved cardiac function, decreased levels of creatine kinase, aspartate aminotransferase and lactate dehydrogenase. EGb 761 also restrained the oxidative stress related to myocardial ischemia injury as evidenced by decreased malondialdehyde, superoxide dismutase, catalase, glutathione-peroxidase, glutathione reductase activity. Meanwhile, the inflammatory cascade was inhibited as evidenced by decreased cytokines such as tumor necrosis factor-α, interleukin-6 and interleukin-1β. Our results still showed that EGb 761 pretreatment significantly decrease the level of cleaved Bax, and increase the level of Bcl-2 in rats subjected to I/R injury. Simultaneously, the expressions of myocardial TLR4 and NF-κB were significantly decreased. It can be concluded that EGb 761 pretreatment was protected against myocardium I/R injury by decreasing oxidative stress, repressing inflammatory cascade in vivo and inhibiting TLR4/NF-κB pathway.
Collapse
Affiliation(s)
- Yuping Tang
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xianyang, 712083, China
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Guisheng Zhou
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Lijun Yao
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Ping Xue
- Changzhou Institute for Food and Drug Control, Changzhou, 213000, China
| | - Danhong Yu
- The Children’s Hospital Affiliated to Soochow University, Suzhou, 215006, China
| | - Renjie Xu
- Shaoxing Second Hospital, Shaoxing, 312000, China
| | - Wen Shi
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xin Yao
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Zhaowei Yan
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Jin-Ao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| |
Collapse
|
47
|
Corrigan F, Arulsamy A, Collins-Praino LE, Holmes JL, Vink R. Toll like receptor 4 activation can be either detrimental or beneficial following mild repetitive traumatic brain injury depending on timing of activation. Brain Behav Immun 2017; 64:124-139. [PMID: 28412141 DOI: 10.1016/j.bbi.2017.04.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 03/30/2017] [Accepted: 04/07/2017] [Indexed: 12/14/2022] Open
Abstract
A history of repeated concussion has been linked to the later development of neurodegeneration, which is associated with the accumulation of hyperphosphorylated tau and the development of behavioral deficits. However, the role that exogenous factors, such as immune activation, may play in the development of neurodegeneration following repeated mild traumatic brain injury (rmTBI) has not yet been explored. To investigate, male Sprague-Dawley rats were administered three mTBIs 5days apart using the diffuse impact-acceleration model to generate ∼100G. Sham animals underwent surgery only. At 1 or 5days following the last injury rats were given the TLR4 agonist, lipopolysaccharide (LPS, 0.1mg/kg), or saline. TLR4 activation had differential effects following rmTBI depending on the timing of activation. When given at 1day post-injury, LPS acutely activated microglia, but decreased production of pro-inflammatory cytokines like IL-6. This was associated with a reduction in neuronal injury, both acutely, with a restoration of levels of myelin basic protein (MBP), and chronically, preventing a loss of both MBP and PSD-95. Furthermore, these animals did not develop behavioral deficits with no changes in locomotion, anxiety, depressive-like behavior or cognition at 3months post-injury. Conversely, when LPS was given at 5days post-injury, it was associated acutely with an increase in pro-inflammatory cytokine production, with an exacerbation of neuronal damage and increased levels of aggregated and phosphorylated tau. At 3months post-injury, there was a slight exacerbation of functional deficits, particularly in cognition and depressive-like behavior. This highlights the complexity of the immune response following rmTBI and the need to understand how a history of rmTBI interacts with environmental factors to influence the potential to develop later neurodegeneration.
Collapse
Affiliation(s)
- Frances Corrigan
- Discipline of Anatomy and Pathology, Adelaide Medical School, University of Adelaide, Adelaide, Australia.
| | - Alina Arulsamy
- Discipline of Anatomy and Pathology, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Lyndsey E Collins-Praino
- Discipline of Anatomy and Pathology, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Joshua L Holmes
- Discipline of Anatomy and Pathology, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Robert Vink
- Sansom Institute for Health Research, University of South Australia, Adelaide, Australia
| |
Collapse
|
48
|
Novel tactics for neuroprotection in Parkinson's disease: Role of antibiotics, polyphenols and neuropeptides. Prog Neurobiol 2017; 155:120-148. [DOI: 10.1016/j.pneurobio.2015.10.004] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Revised: 10/08/2015] [Accepted: 10/26/2015] [Indexed: 02/04/2023]
|
49
|
Yang J, Wang Z, Chen DL. Shikonin ameliorates isoproterenol (ISO)-induced myocardial damage through suppressing fibrosis, inflammation, apoptosis and ER stress. Biomed Pharmacother 2017; 93:1343-1357. [PMID: 28753907 DOI: 10.1016/j.biopha.2017.06.086] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 06/07/2017] [Accepted: 06/23/2017] [Indexed: 12/31/2022] Open
Abstract
Shikonin, isolated from the roots of herbal plant Lithospermum erythrorhizon, is a naphthoquinone. It has been reported to exert beneficial anti-inflammatory effects and anti-oxidant properties in various diseases. Isoproterenol (ISO) has been widely used to establish cardiac injury in vivo and in vitro. However, shikonin function in ISO-induced cardiac injury remains uncertain. In our study, we attempted to investigate the efficiency and possible molecular mechanism of shikonin in cardiac injury treatment induced by ISO. In vivo, C57BL6 mice were subcutaneously injected with 5mg/kg ISO to induce heart failure. And mice were given a gavage of shikonin (2 or 4mg/kg/d, for four weeks). Cardiac function, fibrosis indices, inflammation response, apoptosis and endoplasmic reticulum (ER) stress were calculated. Pathological alterations, fibrosis-, inflammation-, apoptosis- and ER stress-related molecules were examined. In ISO-induced cardiac injury, shikonin significantly ameliorated heart function, decreased myocardial fibrosis, suppressed inflammation, attenuated apoptosis and ER stress through impeding collagen accumulation, Toll like receptor 4/nuclear transcription factor κB (TLR4/NF-κB), Caspase-3 and glucose-regulated protein 78 (GRP78) signaling pathways activity, relieving heart failure in vivo. Also, in vitro, shikonin attenuated ISO-induced cardiac muscle cells by reducing fibrosis, inflammation, apoptosis and ER stress. Our findings indicated that shikonin treatment attenuated ISO-induced heart injury, providing an effective therapeutic strategy for heart failure treatment for future.
Collapse
Affiliation(s)
- Jun Yang
- Department of Cardiology, The First People's Hospital of Yunnan Province, No. 157 Jinbi Road, Kunming 650000, China
| | - Zhao Wang
- Department of Surgery, The First People's Hospital of Yunnan Province, No. 157 Jinbi Road, Kunming 650000, China
| | - Dong-Lin Chen
- Department of Cardiology, Qujing Traditional Chinese Medicine Hospital, No. 8 Jiaotong Road, Qujing 655000, China.
| |
Collapse
|
50
|
Moye LS, Pradhan AA. From blast to bench: A translational mini-review of posttraumatic headache. J Neurosci Res 2017; 95:1347-1354. [PMID: 28151589 DOI: 10.1002/jnr.24001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 11/07/2016] [Accepted: 11/28/2016] [Indexed: 12/25/2022]
Abstract
Current events within the military and professional sports have resulted in an increased recognition of the long-term and debilitating consequences of traumatic brain injury. Mild traumatic brain injury accounts for the majority of head injuries, and posttraumatic headache is the most common adverse effect. It is estimated that between 30% to 90% of traumatic brain injuries result in posttraumatic headache, and for a significant number of people this headache disorder can continue for up to and over a year post injury. Often, the most severe and chronic posttraumatic headache has a migraine-like phenotype and is difficult to resolve. In this review we discuss the preclinical findings from animal models of posttraumatic headache. We also describe potential mechanisms by which traumatic brain injury leads to chronic posttraumatic headache, including neuroinflammatory mediators and migraine-associated neuropeptides. There are surprisingly few preclinical studies that have investigated overlapping mechanisms between posttraumatic headache and migraine, especially considering the prevalence and debilitating nature of posttraumatic headache. Given this context, posttraumatic headache is a field with many emerging opportunities for growth. The frequency of posttraumatic headache in the general and military population is rising, and further preclinical research is required to understand, ameliorate, and treat this disabling disorder. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Laura S Moye
- Department of Psychiatry, University of Illinois at Chicago
| | | |
Collapse
|