1
|
Bhatia A, Thakur S, Kohal R, Brar S, Gupta GD, Verma SK. A comprehensive update on phytochemistry and medicinal developments of apocynin. Fitoterapia 2025; 183:106558. [PMID: 40280248 DOI: 10.1016/j.fitote.2025.106558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 04/11/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
The natural phenolic compound apocynin, referred to as acetovanillone, generated significant attention due to its diverse pharmacological properties, especially as an NADPH oxidase inhibitor, and it was applicable orally and effectively even at small doses. During chronic inflammation, various pro-inflammatory-related factors such as nuclear factor kappa β (NF-kβ), nitrotyrosine, poly adenosine diphosphate ribose polymerase (PARP), inducible nitric oxide synthase (iNOS), cluster of differentiation 31 (CD31), intercellular adhesion molecule-1 (ICAM-1), glycoproteins granule membrane protein 140 (GMP140), tumor necrosis factor-alpha (TNFα), p38 mitogen-activated protein kinases (p38 MAPK), membrane cofactor protein (MCP), interleukin-6 (IL-6), all of which could be targeted by apocynin. Research suggests that apocynin significantly benefits conditions like diabetes, cardiovascular diseases, and neurological disorders due to its ability to mitigate inflammation and enhance endothelial function. Further investigations are essential to examine apocynin and its derivatives, mainly its long-term potency. Future research must focus on clinical trials to evaluate its safety, effectiveness, and optimal dosing in various applications. This review provides a recent update on apocynin, covering aspects such as its extraction and isolation, chemical framework, biosynthesis, synthetic derivatives, pharmacological activities, patent landscape, stability and specifications.
Collapse
Affiliation(s)
- Anchal Bhatia
- Department of Pharmacognosy, ISF College of Pharmacy, Moga 142 001, Punjab, India
| | - Shimple Thakur
- Department of Pharmacognosy, ISF College of Pharmacy, Moga 142 001, Punjab, India
| | - Rupali Kohal
- Department of Pharmacognosy, ISF College of Pharmacy, Moga 142 001, Punjab, India
| | - Seema Brar
- Department of Pharmacognosy, ISF College of Pharmacy, Moga 142 001, Punjab, India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, Moga 142 001, Punjab, India
| | - Sant Kumar Verma
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga 142 001, Punjab, India.
| |
Collapse
|
2
|
Bano N, Khan S, Ahamad S, Dar NJ, Alanazi HH, Nazir A, Bhat SA. Microglial NOX2 as a therapeutic target in traumatic brain injury: Mechanisms, consequences, and potential for neuroprotection. Ageing Res Rev 2025; 108:102735. [PMID: 40122395 DOI: 10.1016/j.arr.2025.102735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/08/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025]
Abstract
Traumatic brain injury (TBI) is a leading cause of long-term disability worldwide, with secondary injury mechanisms, including neuroinflammation and oxidative stress, driving much of its chronic pathology. While NADPH oxidase 2 (NOX2)-mediated reactive oxygen species (ROS) production is a recognized factor in TBI, the specific role of microglial NOX2 in perpetuating oxidative and inflammatory damage remains underexplored. Addressing this gap is critical, as current therapeutic approaches primarily target acute symptoms and fail to interrupt the persistent neuroinflammation that contributes to progressive neurodegeneration. Besides NOX, other ROS-generating enzymes, such as CYP1B1, COX2, and XO, also play crucial roles in triggering oxidative stress and neuroinflammatory conditions in TBI. However, this review highlights the pathophysiological role of microglial NOX2 in TBI, focusing on its activation following injury and its impact on ROS generation, neuroinflammatory signaling, and neuronal loss. These insights reveal NOX2 as a critical driver of secondary injury, linked to worsened outcomes, particularly in aged individuals where NOX2 activation is more pronounced. In addition, this review evaluates emerging therapeutic approaches targeting NOX2, such as GSK2795039 and other selective NOX2 inhibitors, which show potential in reducing ROS levels, limiting neuroinflammation, and preserving neurological functions. By highlighting the specific role of NOX2 in microglial ROS production and secondary neurodegeneration, this study advocates for NOX2 inhibition as a promising strategy to improve TBI outcomes by addressing the unmet need for therapies targeting long-term inflammation and neuroprotection. Our review highlights the potential of NOX2-targeted interventions to disrupt the cycle of oxidative stress and inflammation, ultimately offering a pathway to mitigate the chronic impact of TBI.
Collapse
Affiliation(s)
- Nargis Bano
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Sameera Khan
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Shakir Ahamad
- Department of Chemistry, Aligarh Muslim University, Aligarh 202002, India
| | - Nawab John Dar
- CNB, SALK Institute of Biological Sciences, La Jolla, CA 92037, USA
| | - Hamad H Alanazi
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, Al Jouf University 77455, Saudi Arabia
| | - Aamir Nazir
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India; Academy of Scientific and Innovative Research, New Delhi, India.
| | - Shahnawaz Ali Bhat
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India.
| |
Collapse
|
3
|
Bano N, Khan S, Ahamad S, Dar NJ, Alanazi HH, Nazir A, Bhat SA. Microglial Autophagic Dysregulation in Traumatic Brain Injury: Molecular Insights and Therapeutic Avenues. ACS Chem Neurosci 2025; 16:543-562. [PMID: 39920904 DOI: 10.1021/acschemneuro.4c00617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2025] Open
Abstract
Traumatic brain injury (TBI) is a complex and multifaceted condition that can result in cognitive and behavioral impairments. One aspect of TBI that has received increasing attention in recent years is the role of microglia, the brain-resident immune cells, in the pathophysiology of the injury. Specifically, increasing evidence suggests that dysfunction in microglial autophagy, the process by which cells degrade and recycle their own damaged components, may contribute to the development and progression of TBI-related impairments. Here, we unravel the pathways by which microglia autophagic dysregulation predisposes the brain to secondary damage and neurological deficits following TBI. An overview of the role of autophagic dysregulation in perpetuation and worsening of the inflammatory response, neuroinflammation, and neuronal cell death in TBI follows. Further, we have evaluated several signaling pathways and processes that contribute to autophagy dysfunction-mediated inflammation, neurodegeneration, and poor outcome in TBI. Additionally, a discussion on the small molecule therapeutics employed to modulate these pathways and mechanisms to treat TBI have been presented. However, additional research is required to fully understand the processes behind these underlying pathways and uncover potential therapeutic targets for restoring microglial autophagic failure in TBI.
Collapse
Affiliation(s)
- Nargis Bano
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Sameera Khan
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Shakir Ahamad
- Department of Chemistry, Aligarh Muslim University, Aligarh 202002, India
| | - Nawab John Dar
- CNB, SALK Institute of Biological Sciences, La Jolla, California 92037, United States
| | - Hamad H Alanazi
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, Al Jouf University, Sakaka 77455, Saudi Arabia
| | - Aamir Nazir
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
- Academy of Scientific and Innovative Research, New Delhi 201002, India
| | | |
Collapse
|
4
|
Tabassum S, Wu S, Lee CH, Yang BSK, Gusdon AM, Choi HA, Ren XS. Mitochondrial-targeted therapies in traumatic brain injury: From bench to bedside. Neurotherapeutics 2025; 22:e00515. [PMID: 39721917 PMCID: PMC11840356 DOI: 10.1016/j.neurot.2024.e00515] [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: 07/30/2024] [Revised: 12/03/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of morbidity and mortality worldwide, with limited effective therapeutic options currently available. Recent research has highlighted the pivotal role of mitochondrial dysfunction in the pathophysiology of TBI, making mitochondria an attractive target for therapeutic intervention. This review comprehensively examines advancements in mitochondrial-targeted therapies for TBI, bridging the gap from basic research to clinical applications. We discuss the underlying mechanisms of mitochondrial damage in TBI, including oxidative stress, impaired bioenergetics, mitochondrial dynamics, and apoptotic pathways. Furthermore, we highlight the complex interplay between mitochondrial dysfunction, inflammation, and blood-brain barrier (BBB) integrity, elucidating how these interactions exacerbate injury and impede recovery. We also evaluate various preclinical studies exploring pharmacological agents, gene therapy, and novel drug delivery systems designed to protect and restore mitochondrial function. Clinical trials and their outcomes are assessed to evaluate the translational potential of mitochondrial-targeted therapies in TBI. By integrating findings from bench to bedside, this review emphasizes promising therapeutic avenues and addresses remaining challenges. It also provides guidance for future research to pave the way for innovative treatments that improve patient outcomes in TBI.
Collapse
Affiliation(s)
- Sidra Tabassum
- Novel Treatments for Acute Brain Injury Institute, Texas Medical Center, TX, USA; Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Silin Wu
- Novel Treatments for Acute Brain Injury Institute, Texas Medical Center, TX, USA; Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Chang-Hun Lee
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Bosco Seong Kyu Yang
- Novel Treatments for Acute Brain Injury Institute, Texas Medical Center, TX, USA; Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Aaron M Gusdon
- Novel Treatments for Acute Brain Injury Institute, Texas Medical Center, TX, USA; Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Huimahn A Choi
- Novel Treatments for Acute Brain Injury Institute, Texas Medical Center, TX, USA; Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Xuefang S Ren
- Novel Treatments for Acute Brain Injury Institute, Texas Medical Center, TX, USA; Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA.
| |
Collapse
|
5
|
Gao TX, Liang Y, Li J, Zhao D, Dong BJ, Xu C, Zhao WD, Li X, Zhao CS. Knockout of neutrophil cytosolic factor 1 ameliorates neuroinflammation and motor deficit after traumatic brain injury. Exp Neurol 2024; 382:114983. [PMID: 39357591 DOI: 10.1016/j.expneurol.2024.114983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/17/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024]
Abstract
Traumatic brain injury (TBI) is a predominant cause of long-term disability in adults, yet the molecular mechanisms underpinning the neuropathological processes associated with it remain inadequately understood. Neutrophil cytosolic factor 1 (NCF1, also known as p47phox) is one of the cytosolic components of NADPH oxidase NOX2. In this study, we observed a reduction in the volume of TBI-induced brain lesions in NCF1-knockout mice compared to controls. Correspondingly, the neuronal loss induced by TBI was mitigated in the NCF1-knockout mice. Behavioral analysis also demonstrated that the motor coordination deficit following TBI was mitigated by the depletion of NCF1. Mechanistically, our findings revealed that NCF1 deficiency attenuated TBI-induced inflammatory responses by inhibiting the release of proinflammatory factors and reducing neutrophil infiltration into the brain parenchyma. Additionally, our results indicated that NCF1 deficiency significantly decreased the levels of reactive oxygen species in neutrophils. Taken together, our findings indicate that NCF1 plays a crucial role in the regulation of brain injury and secondary inflammation post-TBI.
Collapse
Affiliation(s)
- Tian-Xu Gao
- Department of Neurology, First Affiliated Hospital, China Medical University, Shenyang 110001, China; Department of Developmental Cell Biology, School of Life Sciences, China Medical University, Shenyang 110122, China
| | - Yu Liang
- Department of Developmental Cell Biology, School of Life Sciences, China Medical University, Shenyang 110122, China
| | - Jian Li
- Department of Developmental Cell Biology, School of Life Sciences, China Medical University, Shenyang 110122, China
| | - Dan Zhao
- Department of Developmental Cell Biology, School of Life Sciences, China Medical University, Shenyang 110122, China
| | - Bai-Jun Dong
- School of Basic Medical Sciences, China Medical University, Shenyang 110122, China
| | - Chen Xu
- Department of Laboratory, Xilinguole Central Hospital, Xilinhot 026000, China.
| | - Wei-Dong Zhao
- Department of Developmental Cell Biology, School of Life Sciences, China Medical University, Shenyang 110122, China.
| | - Xia Li
- Department of Gynecology and Obstetrics, Hohhot Maternal and Child Health Care Hospital, Hohhot 110000, China.
| | - Chuan-Sheng Zhao
- Department of Neurology, First Affiliated Hospital, China Medical University, Shenyang 110001, China.
| |
Collapse
|
6
|
Navabi SP, Badreh F, Khombi Shooshtari M, Hajipour S, Moradi Vastegani S, Khoshnam SE. Microglia-induced neuroinflammation in hippocampal neurogenesis following traumatic brain injury. Heliyon 2024; 10:e35869. [PMID: 39220913 PMCID: PMC11365414 DOI: 10.1016/j.heliyon.2024.e35869] [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: 11/21/2023] [Revised: 07/25/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Traumatic brain injury (TBI) is one of the most causes of death and disability among people, leading to a wide range of neurological deficits. The important process of neurogenesis in the hippocampus, which includes the production, maturation and integration of new neurons, is affected by TBI due to microglia activation and the inflammatory response. During brain development, microglia are involved in forming or removing synapses, regulating the number of neurons, and repairing damage. However, in response to injury, activated microglia release a variety of pro-inflammatory cytokines, chemokines and other neurotoxic mediators that exacerbate post-TBI injury. These microglia-related changes can negatively affect hippocampal neurogenesis and disrupt learning and memory processes. To date, the intracellular signaling pathways that trigger microglia activation following TBI, as well as the effects of microglia on hippocampal neurogenesis, are poorly understood. In this review article, we discuss the effects of microglia-induced neuroinflammation on hippocampal neurogenesis following TBI, as well as the intracellular signaling pathways of microglia activation.
Collapse
Affiliation(s)
- Seyedeh Parisa Navabi
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Maryam Khombi Shooshtari
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Somayeh Hajipour
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sadegh Moradi Vastegani
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed Esmaeil Khoshnam
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
7
|
Yang Y, Lu D, Wang M, Liu G, Feng Y, Ren Y, Sun X, Chen Z, Wang Z. Endoplasmic reticulum stress and the unfolded protein response: emerging regulators in progression of traumatic brain injury. Cell Death Dis 2024; 15:156. [PMID: 38378666 PMCID: PMC10879178 DOI: 10.1038/s41419-024-06515-x] [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: 10/17/2023] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 02/22/2024]
Abstract
Traumatic brain injury (TBI) is a common trauma with high mortality and disability rates worldwide. However, the current management of this disease is still unsatisfactory. Therefore, it is necessary to investigate the pathophysiological mechanisms of TBI in depth to improve the treatment options. In recent decades, abundant evidence has highlighted the significance of endoplasmic reticulum stress (ERS) in advancing central nervous system (CNS) disorders, including TBI. ERS following TBI leads to the accumulation of unfolded proteins, initiating the unfolded protein response (UPR). Protein kinase RNA-like ER kinase (PERK), inositol-requiring protein 1 (IRE1), and activating transcription factor 6 (ATF6) are the three major pathways of UPR initiation that determine whether a cell survives or dies. This review focuses on the dual effects of ERS on TBI and discusses the underlying mechanisms. It is suggested that ERS may crosstalk with a series of molecular cascade responses, such as mitochondrial dysfunction, oxidative stress, neuroinflammation, autophagy, and cell death, and is thus involved in the progression of secondary injury after TBI. Hence, ERS is a promising candidate for the management of TBI.
Collapse
Affiliation(s)
- Yayi Yang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188Shizi Street, Suzhou, 215006, Jiangsu Province, China
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Dengfeng Lu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188Shizi Street, Suzhou, 215006, Jiangsu Province, China
| | - Menghan Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188Shizi Street, Suzhou, 215006, Jiangsu Province, China
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Guangjie Liu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188Shizi Street, Suzhou, 215006, Jiangsu Province, China
| | - Yun Feng
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188Shizi Street, Suzhou, 215006, Jiangsu Province, China
- Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Yubo Ren
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188Shizi Street, Suzhou, 215006, Jiangsu Province, China
| | - Xiaoou Sun
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188Shizi Street, Suzhou, 215006, Jiangsu Province, China.
| | - Zhouqing Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188Shizi Street, Suzhou, 215006, Jiangsu Province, China.
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188Shizi Street, Suzhou, 215006, Jiangsu Province, China.
| |
Collapse
|
8
|
Karakaya E, Oleinik N, Edwards J, Tomberlin J, Barker RB, Berber B, Ericsson M, Alsudani H, Ergul A, Beyaz S, Lemasters JJ, Ogretmen B, Albayram O. p17/C18-ceramide-mediated mitophagy is an endogenous neuroprotective response in preclinical and clinical brain injury. PNAS NEXUS 2024; 3:pgae018. [PMID: 38328780 PMCID: PMC10847724 DOI: 10.1093/pnasnexus/pgae018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 01/10/2024] [Indexed: 02/09/2024]
Abstract
Repeat concussions (or repetitive mild traumatic brain injury [rmTBI]) are complex pathological processes consisting of a primary insult and long-term secondary complications and are also a prerequisite for chronic traumatic encephalopathy (CTE). Recent evidence implies a significant role of autophagy-mediated dysfunctional mitochondrial clearance, mitophagy, in the cascade of secondary deleterious events resulting from TBI. C18-ceramide, a bioactive sphingolipid produced in response to cell stress and damage, and its synthesizing enzyme (CerS1) are precursors to selective stress-mediated mitophagy. A transporter, p17, mediates the trafficking of CerS1, induces C18-ceramide synthesis in the mitochondrial membrane, and acts as an elimination signal in cell survival. Whether p17-mediated mitophagy occurs in the brain and plays a causal role in mitochondrial quality control in secondary disease development after rmTBI are unknown. Using a novel repetitive less-than-mild TBI (rlmTBI) injury paradigm, ablation of mitochondrial p17/C18-ceramide trafficking in p17 knockout (KO) mice results in a loss of C18-ceramide-induced mitophagy, which contributes to susceptibility and recovery from long-term secondary complications associated with rlmTBI. Using a ceramide analog with lipid-selenium conjugate drug, LCL768 restored mitophagy and reduced long-term secondary complications, improving cognitive deficits in rlmTBI-induced p17KO mice. We obtained a significant reduction of p17 expression and a considerable decrease of CerS1 and C18-ceramide levels in cortical mitochondria of CTE human brains compared with age-matched control brains. These data demonstrated that p17/C18-ceramide trafficking is an endogenous neuroprotective mitochondrial stress response following rlmTBI, thus suggesting a novel prospective strategy to interrupt the CTE consequences of concussive TBI.
Collapse
Affiliation(s)
- Eda Karakaya
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Natalia Oleinik
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Jazlyn Edwards
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Jensen Tomberlin
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Randy Bent Barker
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Burak Berber
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Biology, Eskisehir Technical University, Tepebasi/Eskisehir 26555, Turkey
| | - Maria Ericsson
- Electron Microscopy Laboratory, Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Habeeb Alsudani
- Cancer Center, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
- College of Science, University of Basrah, Basra 61004, Iraq
| | - Adviye Ergul
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
- Ralph H. Jackson Department of Veterans Affairs Medical Center, Charleston, SC 29425, USA
| | - Semir Beyaz
- Cancer Center, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA
| | - John J Lemasters
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Besim Ogretmen
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Onder Albayram
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
- Ralph H. Jackson Department of Veterans Affairs Medical Center, Charleston, SC 29425, USA
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
9
|
Li B, Shi Y, Fu Y. Apocynin inhibits compressive force-induced apoptosis and autophagy of periodontal ligament stem cells. Oral Dis 2023; 29:2837-2844. [PMID: 35770313 DOI: 10.1111/odi.14296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 04/27/2022] [Accepted: 06/26/2022] [Indexed: 11/26/2022]
Abstract
OBJECTIVE This study aimed to evaluate the effect of Apocynin on compressive force-induced apoptosis and autophagy in periodontal ligament stem cells (PDLSCs). MATERIALS AND METHODS Periodontal ligament stem cells were subjected to a uniform compressive force of 2.0 g/cm2 for 24 h, without and with addition of 50, 100 and 200 μM Apocynin. Beclin-1 was overexpressed in PDLSCs. Flow cytometry was used to assess apoptosis and transmission electron microscopy was used to evaluate ultrastructural features of PDLSCs. Immunofluorescence was used to assess the levels of microtubule-associated protein 1 light chain 3 (LC3). The protein levels of B-cell lymphoma-2 (Bcl-2), Bcl-2 associated X (Bax), Caspase-3, LC3, and Beclin were determined by Western blot analysis. RESULTS Compressive force of PDLSCs significantly increased apoptosis, Bax, Caspase-3, the number of autophagosome or autolysosomes, Beclin-1, the ratio of LC3 II/LC3 I, and decreased Bcl-2. Apocynin was shown to inhibit apoptosis and Beclin-1-mediated autophagy. Over-expression of Beclin-1 increased apoptosis and autophagy. CONCLUSIONS Application of Apocynin attenuated long-term compressive force-induced apoptosis by regulating Beclin-1-mediated autophagy in PDLSCs. These results provide an alternative approach to improve orthodontic treatment outcomes for patients.
Collapse
Affiliation(s)
- Bin Li
- Department of Stomatology, Shengzhou People's Hospital, The First Affiliated Hospital of Zhejiang University Shengzhou Branch, Shengzhou, China
| | - Yuchun Shi
- Department of Stomatology, Shengzhou Hospital of Traditional Chinese Medicine, Shengzhou, China
| | - Yanbin Fu
- Department of Dental Implant, Yiwu Stomatological Hospital, Jinhua, China
| |
Collapse
|
10
|
Ding X, Zhang L, Zhang X, Qin Y, Yu K, Yang X. Intranasal Insulin Alleviates Traumatic Brain Injury by Inhibiting Autophagy and Endoplasmic Reticulum Stress-mediated Apoptosis Through the PI3K/Akt/mTOR Signaling Pathway. Neuroscience 2023; 529:23-36. [PMID: 37572876 DOI: 10.1016/j.neuroscience.2023.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/18/2023] [Accepted: 08/04/2023] [Indexed: 08/14/2023]
Abstract
Intranasal insulin reduces lesion size and enhances memory capacity in traumatic brain injury (TBI) models, but the molecular mechanisms behind this neuroprotective action not yet understood. Here we used Feeney's free-falling method to construct TBI mouse models and administrated intranasal insulin, rapamycin, insulin and rapamycin, or normal saline to assess their effects on neurological functions, cerebral edema, and the expression of Iba1 in microglia through immunofluorescence assay. We also measured concentrations of interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α) in the brain using enzyme immunosorbent assay, investigated apoptosis with TUNEL staining and Western blotting, and evaluated autophagy, endoplasmic reticulum (ER) stress, and PI3K/Akt/mTOR signaling pathway with Western blotting. The autophagosome was assessed through transmission electron microscopy. Our findings demonstrated that intranasal insulin promoted neurological recovery, decreased brain swelling, and reduced injury lesions on days 1, 3, and 7 post TBI. Moreover, intranasal insulin reduced microglia activation and the concentration of IL-1β or TNF-α on the same days. Through Western blotting and transmission electron microscopy, we observed that intranasal insulin suppressed autophagy while activating the PI3K/AKT/mTOR signaling pathway on days 1 and 3 post TBI. TUNEL assay and Western blotting also indicated that intranasal insulin inhibited ER stress-mediated apoptosis. Interestingly, the mTOR inhibitor rapamycin partially blocked the pro-autophagy and anti-apoptosis effects of intranasal insulin both on days 1 and 3 post TBI. Our results suggest that intranasal insulin can ameliorate TBI by regulating autophagy and ER stress-mediated apoptosis through the PI3K/AKT/mTOR signaling pathway, providing a promising therapeutic strategy for TBI.
Collapse
Affiliation(s)
- Xin Ding
- Department of Neurology, Chengdu Second People's Hospital, No. 2, Huatai Road, Chenghua District, Chengdu, Sichuan 610017, People's Republic of China
| | - Lili Zhang
- Department of Neurology, The First Affiliated Hospital of Chengdu Medical College, No, 278, Middle Baoguang Avenue, Xindu District, Chengdu, Sichuan 610050, People's Republic of China
| | - Xinping Zhang
- Department of General Medicine, Chengdu Second People's Hospital, No. 2, Huatai Road, Chenghua District, Chengdu, Sichuan 610017, People's Republic of China
| | - Yang Qin
- Department of General Medicine, The General Hospital of Western Theatre Command, No. 270, Tianhui Road, Rongdu Avenue, Jinniu District, Chengdu, Sichuan 610083, People's Republic of China.
| | - Ke Yu
- Department of General Medicine, The General Hospital of Western Theatre Command, No. 270, Tianhui Road, Rongdu Avenue, Jinniu District, Chengdu, Sichuan 610083, People's Republic of China
| | - Xiaokun Yang
- Department of Emergency, The General Hospital of Western Theatre Command, No. 270, Tianhui Road, Rongdu Avenue, Jinniu District, Chengdu, Sichuan 610083, People's Republic of China
| |
Collapse
|
11
|
Park YJ, Gil TY, Jin BR, Cha YY, An HJ. Apocynin alleviates weight gain and obesity-induced adipose tissue inflammation in high-fat diet-fed C57BL/6 mice. Phytother Res 2023; 37:3481-3494. [PMID: 37194916 DOI: 10.1002/ptr.7823] [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: 10/24/2022] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 05/18/2023]
Abstract
Obesity involves chronic low-grade inflammation within adipose tissue. Apocynin (APO) is a therapeutic agent for the treatment of inflammatory diseases. Therefore, the present study aimed to investigate whether APO can reduce weight gain and obesity-induced adipose tissue inflammation. C57BL/6 mice were administered APO or orlistat (Orli) as a positive control with a high-fat diet (HFD) for 12 weeks. Lipopolysaccharide-stimulated 3T3-L1 adipocytes were used for the in vitro study. Our results showed a significantly lower white adipose tissue (WAT) mass index in 10 mg/kg APO-treated mice than in 20 mg/kg Orli-treated mice. Moreover, the protein expression of adipose triglyceride lipase, fatty acid synthase, sterol regulatory element-binding transcription factor 1, and peroxisome proliferator-activated receptor γ was reversed in the WAT of 10 mg/kg APO-treated mice. Furthermore, APO reduced the expression of the macrophage marker F4/80, decreased the mRNA levels of tumor necrosis factor-α and monocyte chemoattractant protein-1, and increased the mRNA levels of interleukin-10 in WAT. APO decreased the phosphorylation of c-Jun N-terminal kinase, extracellular signal-regulated kinase, and p65 in vivo and in vitro. Notably, APO had a stronger effect on the amelioration of adipose tissue inflammation than Orli did. Our findings lay the foundation for research on the use of APO as an agent to ameliorate weight gain and obesity-induced inflammatory diseases.
Collapse
Affiliation(s)
- Yea-Jin Park
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
| | - Tae-Young Gil
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
| | - Bo-Ram Jin
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
| | - Yun-Yeop Cha
- Department of Rehabilitative Medicine of Korean Medicine and Neuropsychiatry, College of Korean Medicine, Sangji University, Wonju, Gangwon-do, Republic of Korea
| | - Hyo-Jin An
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
- Department of Integrated Drug Development and Natural Products, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
12
|
Hassanein EHM, Sayed AM, El-Ghafar OAMA, Omar ZMM, Rashwan EK, Mohammedsaleh ZM, Kyung SY, Park JH, Kim HS, Ali FEM. Apocynin abrogates methotrexate-induced nephrotoxicity: role of TLR4/NF-κB-p65/p38-MAPK, IL-6/STAT-3, PPAR-γ, and SIRT1/FOXO3 signaling pathways. Arch Pharm Res 2023; 46:339-359. [PMID: 36913116 DOI: 10.1007/s12272-023-01436-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 02/25/2023] [Indexed: 03/14/2023]
Abstract
The present study was designed to evaluate the potential renoprotective impacts of apocynin (APC) against nephrotoxicity induced by methotrexate (MTX) administration. To fulfill this aim, rats were allocated into four groups: control; APC (100 mg/kg/day; orally); MTX (20 mg/kg; single intraperitoneal dose at the end of the 5th day of the experiment); and APC +MTX (APC was given orally for 5 days before and 5 days after induction of renal toxicity by MTX). On the 11th day, samples were collected to estimate kidney function biomarkers, oxidative stress, pro-inflammatory cytokines, and other molecular targets. Compared to the MTX control group, treatment with APC significantly decreased urea, creatinine, and KIM-1 levels and improved kidney histological alterations. Furthermore, APC restored oxidant/antioxidant balance, as evidenced by a remarkable alleviation of MDA, GSH, SOD, and MPO levels. Additionally, the iNOS, NO, p-NF-κB-p65, Ace-NF-κB-p65, TLR4, p-p38-MAPK, p-JAK1, and p-STAT-3 expressions were reduced, while the IκBα, PPAR-γ, SIRT1, and FOXO3 expressions were significantly increased. In NRK-52E cells, MTX-induced cytotoxicity was protected by APC in a concentration-dependent manner. In addition, increased expression of p-STAT-3 and p-JAK1/2 levels were reduced in MTX-treated NRK-52E cells by APC. The in vitro experiments revealed that APC-protected MTX-mediated renal tubular epithelial cells were damaged by inhibiting the JAK/STAT3 pathway. Besides, our in vivo and in vitro results were confirmed by predicting computational pharmacology results using molecular docking and network pharmacology analysis. In conclusion, our findings proved that APC could be a good candidate for MTX-induced renal damage due to its strong antioxidative and anti-inflammatory bioactivities.
Collapse
Affiliation(s)
- Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, 71524, Asyut, Egypt
| | - Ahmed M Sayed
- Biochemistry Laboratory, Chemistry Department, Faculty of Science, Assiut University, Asyut, Egypt
| | - Omnia A M Abd El-Ghafar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni- Suef, Egypt
| | - Zainab M M Omar
- Department of Pharmacology, College of Medicine, Al-Azhar University, 71524, Asyut, Egypt
| | - Eman K Rashwan
- Department of Physiology, College of Medicine, Jouf University, 42421, Sakaka, Saudi Arabia
| | - Zuhair M Mohammedsaleh
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, 71491, Tabuk, Kingdom of Saudi Arabia
| | - So Young Kyung
- Division of Toxicology, School of Pharmacy, Sungkyunkwan University, 16419, Suwon, Republic of Korea
| | - Jae Hyeon Park
- Division of Toxicology, School of Pharmacy, Sungkyunkwan University, 16419, Suwon, Republic of Korea
| | - Hyung Sik Kim
- Division of Toxicology, School of Pharmacy, Sungkyunkwan University, 16419, Suwon, Republic of Korea.
| | - Fares E M Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, 71524, Asyut, Egypt
| |
Collapse
|
13
|
Zhou J, Lin H, Lv T, Hao J, Zhang H, Sun S, Yang J, Chi J, Guo H. Inappropriate Activation of TLR4/NF-κB is a Cause of Heart Failure. CARDIOVASCULAR INNOVATIONS AND APPLICATIONS 2022. [DOI: 10.15212/cvia.2022.0020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Significance: Heart failure, a disease with extremely high incidence, is closely associated with inflammation and oxidative stress. The Toll-like receptor 4 (TLR4)/nuclear factor kappa-B (NF-κB) pathway plays an important role in the occurrence and development of heart failure.
Recent advances: Previous studies have shown that TLR4/NF-κB causes heart failure by inducing oxidative stress and inflammation; damaging the endothelia; promoting fibrosis; and inducing myocardial hypertrophy, apoptosis, pyroptosis, and autophagy.
Critical issues: Understanding the pathogenesis of heart failure is essential for the treatment of this disease. In this review, we outline the mechanisms underlying TLR4/NF-κB pathway-mediated heart failure and discuss drugs that alleviate heart failure by regulating the TLR4/NF-κB pathway.
Future directions: During TLR4/NF-κB overactivation, interventions targeting specific receptor antagonists may effectively alleviate heart failure, thus providing a basis for the development of new anti-heart failure drugs.
Collapse
Affiliation(s)
- Jiedong Zhou
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, China
| | - Hui Lin
- Department of Cardiology, Shaoxing People’s Hospital Shaoxing Hospital, Shaoxing, China
| | - Tingting Lv
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, China
| | - Jinjin Hao
- Zhejiang University School of Medicine, Hangzhou, China
| | - Hanlin Zhang
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Shimin Sun
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Juntao Yang
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, China
| | - Jufang Chi
- Department of Cardiology, Shaoxing People’s Hospital Shaoxing Hospital, Shaoxing, China
| | - Hangyuan Guo
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, China
| |
Collapse
|
14
|
Zheng RZ, Lee KY, Qi ZX, Wang Z, Xu ZY, Wu XH, Mao Y. Neuroinflammation Following Traumatic Brain Injury: Take It Seriously or Not. Front Immunol 2022; 13:855701. [PMID: 35392083 PMCID: PMC8981520 DOI: 10.3389/fimmu.2022.855701] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 02/23/2022] [Indexed: 12/30/2022] Open
Abstract
Traumatic brain injury (TBI) is associated with high mortality and disability, with a substantial socioeconomic burden. With the standardization of the treatment process, there is increasing interest in the role that the secondary insult of TBI plays in outcome heterogeneity. The secondary insult is neither detrimental nor beneficial in an absolute sense, among which the inflammatory response was a complex cascade of events and can thus be regarded as a double-edged sword. Therefore, clinicians should take the generation and balance of neuroinflammation following TBI seriously. In this review, we summarize the current human and animal model studies of neuroinflammation and provide a better understanding of the inflammatory response in the different stages of TBI. In particular, advances in neuroinflammation using proteomic and transcriptomic techniques have enabled us to identify a functional specific delineation of the immune cell in TBI patients. Based on recent advances in our understanding of immune cell activation, we present the difference between diffuse axonal injury and focal brain injury. In addition, we give a figurative profiling of the general paradigm in the pre- and post-injury inflammatory settings employing a bow-tie framework.
Collapse
Affiliation(s)
- Rui-Zhe Zheng
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.,State Key Laboratory of Medical Neurobiology and Ministry of Education (MOE) Frontiers Center for Brain Science, School of Basic Medical Sciences and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Kuin-Yu Lee
- Department of Integrative Medicine and Neurobiology, Institute of Integrative Medicine of Fudan University Institute of Brain Science, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Zeng-Xin Qi
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.,State Key Laboratory of Medical Neurobiology and Ministry of Education (MOE) Frontiers Center for Brain Science, School of Basic Medical Sciences and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Zhe Wang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.,State Key Laboratory of Medical Neurobiology and Ministry of Education (MOE) Frontiers Center for Brain Science, School of Basic Medical Sciences and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Ze-Yu Xu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.,State Key Laboratory of Medical Neurobiology and Ministry of Education (MOE) Frontiers Center for Brain Science, School of Basic Medical Sciences and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Xue-Hai Wu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.,State Key Laboratory of Medical Neurobiology and Ministry of Education (MOE) Frontiers Center for Brain Science, School of Basic Medical Sciences and Institutes of Brain Science, Fudan University, Shanghai, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China.,Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.,State Key Laboratory of Medical Neurobiology and Ministry of Education (MOE) Frontiers Center for Brain Science, School of Basic Medical Sciences and Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
15
|
Neural stem cell therapy in conjunction with curcumin loaded in niosomal nanoparticles enhanced recovery from traumatic brain injury. Sci Rep 2022; 12:3572. [PMID: 35246564 PMCID: PMC8897489 DOI: 10.1038/s41598-022-07367-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 02/16/2022] [Indexed: 12/28/2022] Open
Abstract
Despite a great amount of effort, there is still a need for reliable treatments of traumatic brain injury (TBI). Recently, stem cell therapy has emerged as a new avenue to address neuronal regeneration after TBI. However, the environment of TBI lesions exerts negative effects on the stem cells efficacy. Therefore, to maximize the beneficial effects of stem cells in the course of TBI, we evaluated the effect of human neural stem/progenitor cells (hNS/PCs) and curcumin-loaded niosome nanoparticles (CM-NPs) on behavioral changes, brain edema, gliosis, and inflammatory responses in a rat model of TBI. After TBI, hNS/PCs were transplanted within the injury site and CM-NPs were orally administered for 10 days. Finally, the effect of combination therapy was compared to several control groups. Our results indicated a significant improvement of general locomotor activity in the hNS/PCs + CM-NPs treatment group compared to the control groups. We also observed a significant improvement in brain edema in the hNS/PCs + CM-NPs treatment group compared to the other groups. Furthermore, a significant decrease in astrogliosis was seen in the combined treatment group. Moreover, TLR4-, NF-κB-, and TNF-α- positive cells were significantly decreased in hNS/PCs + CM-NPs group compared to the control groups. Taken together, this study indicated that combination therapy of stem cells with CM-NPs can be an effective therapy for TBI.
Collapse
|
16
|
Tanyeli A, Guzel Erdogan D, Comakli S, Polat E, Guler MC, Eraslan E, Doganay S. Therapeutic effects of apocynin on ovarian ischemia-reperfusion induced lung injury. Biotech Histochem 2022; 97:536-545. [PMID: 35152781 DOI: 10.1080/10520295.2022.2036368] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Ovarian ischemia-reperfusion (I-R) injury may damage remote organs, including the lungs. We investigated whether apocynin, a NADPH oxidase inhibitor, might protect against ovarian I-R induced apoptosis in the lungs of rats. Bilateral ovarian I-R was induced for 3 h, then apocynin was applied at two concentrations. Lung tissue was evaluated using spectrophotometric and immunohistochemical methods. We found that I-R increased total oxidant status (TOS), oxidative stress index (OSI) and myeloperoxidase (MPO) levels, and immunostaining of nuclear factor kappa-B (NF-κB), light chain 3B (LC3B), interleukin 1-beta (IL-1β), caspase-3 and tumor necrosis factor-alpha (TNF-α), but decreased superoxide dismutase (SOD) values. Apocynin application to I-R injured rats enhanced recovery of lung tissue oxidants and improved both histology and frequency of apoptosis.
Collapse
Affiliation(s)
- Ayhan Tanyeli
- Department of Physiology, Faculty of Medicine, Atatürk University, Erzurum, Turkey
| | - Derya Guzel Erdogan
- Department of Physiology, Faculty of Medicine, Sakarya University, Sakarya, Turkey
| | - Selim Comakli
- Department of Pathology, Veterinary Faculty, Atatürk University, Erzurum, Turkey
| | - Elif Polat
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Erzurum Technical University, Erzurum, Turkey
| | - Mustafa Can Guler
- Department of Physiology, Faculty of Medicine, Atatürk University, Erzurum, Turkey
| | - Ersen Eraslan
- Department of Physiology, Faculty of Medicine, Yozgat Bozok University, Yozgat, Turkey
| | - Songul Doganay
- Department of Physiology, Faculty of Medicine, Sakarya University, Sakarya, Turkey
| |
Collapse
|
17
|
Zhu L, Dong C, Yue X, Ge P, Zheng G, Ye Z, Pan B. Silencing of TRIM44 Inhibits Inflammation and Alleviates Traumatic Brain Injury in Rats by Downregulating TLR4-NF-κB Signaling. Neuroimmunomodulation 2022; 29:439-449. [PMID: 35609523 DOI: 10.1159/000524536] [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: 12/03/2021] [Accepted: 04/05/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Neuroinflammation subsequent to traumatic brain injury (TBI) is important for the recovery of patients and is associated with neurodegenerative changes post-TBI. The tripartite motif containing 44 (TRIM44) protein is an E3 ligase involved in the regulation of immune function with no previously known link to TBI. This study explores the connection between TRIM44 and TBI. METHODS After induction of TBI in rats by control cortex injury, TRIM44 expressions were determined with quantitative real-time reverse transcription polymerase chain reaction and Western blot, and Toll-like receptor 4 (TLR4)-NF-κB signaling was examined by the expression of TLR4, p65 phosphorylation, and the specific NF-κB transcription activity. The effects of TRIM44 knockdown on inflammation, neurological function, and TLR4-NF-κB signaling in TBI rats were revealed by the detection of proinflammatory cytokines and TLR4-NF-κB signaling molecules, modified neurological severity score, brain water content, and Evans blue permeability. RESULTS We found that TRIM44 expression was significantly increased following TBI induction along with TLR4-NF-κB activation. Silencing of TRIM44 suppressed proinflammatory cytokine production, improved neurological outcomes, alleviated brain edema, and inhibited TLR4-NF-κB signaling in TBI rats. CONCLUSION Our findings suggest that suppressing TRIM44 or modulation of relevant pathways may be a therapeutic strategy for TBI.
Collapse
Affiliation(s)
- Lin Zhu
- Hebei General Hospital, Shijiazhuang, China
| | - Ce Dong
- Hebei General Hospital, Shijiazhuang, China
| | | | | | | | | | - Baogen Pan
- Hebei General Hospital, Shijiazhuang, China
| |
Collapse
|
18
|
Davis CK, Vemuganti R. Antioxidant therapies in traumatic brain injury. Neurochem Int 2022; 152:105255. [PMID: 34915062 PMCID: PMC11884749 DOI: 10.1016/j.neuint.2021.105255] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 12/07/2021] [Accepted: 12/10/2021] [Indexed: 12/24/2022]
Abstract
Oxidative stress plays a crucial role in traumatic brain injury (TBI) pathogenesis. Reactive oxygen species (ROS) and reactive nitrogen species (RNS) formed in excess after TBI synergistically contribute to secondary brain damage together with lipid peroxidation products (reactive aldehydes) and inflammatory mediators. Furthermore, oxidative stress, endoplasmic reticulum stress and inflammation potentiate each other. Following TBI, excessive oxidative stress overloads the endogenous cellular antioxidant system leading to cell death. To combat oxidative stress, several antioxidant therapies were tested in preclinical animal models of TBI. These include free radical scavengers, activators of antioxidant systems, Inhibitors of free radical generating enzymes and antioxidant enzymes. Many of these therapies showed promising outcomes including reduced edema, blood-brain barrier (BBB) protection, smaller contusion volume, and less inflammation. In addition, many antioxidant therapies also promoted better sensory, motor, and cognitive functional recovery after TBI. Overall, preventing oxidative stress is a viable therapeutic option to minimize the secondary damage and to improve the quality of life after TBI.
Collapse
Affiliation(s)
- Charles K Davis
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA; William S. Middleton Memorial Veteran Administration Hospital, Madison, WI, USA.
| |
Collapse
|
19
|
Xing Z, Zhen T, Jie F, Jie Y, Shiqi L, Kaiyi Z, Zhicui O, Mingyan H. Early Toll-like receptor 4 inhibition improves immune dysfunction in the hippocampus after hypoxic-ischemic brain damage. Int J Med Sci 2022; 19:142-151. [PMID: 34975308 PMCID: PMC8692118 DOI: 10.7150/ijms.66494] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 11/21/2021] [Indexed: 11/25/2022] Open
Abstract
Background: Toll-like receptor 4 (TLR4) is implicated in neonatal hypoxic-ischemic brain damage (HIBD), but the underlying mechanism is unclear. Hypothesis: We hypothesized that TLR4 mediates brain damage after hypoxic ischemia (HI) by inducing abnormal neuroimmune responses, including activation of immune cells and expression disorder of immune factors, while early inhibition of TLR4 can alleviate the neuroimmune dysfunction. Method: Postnatal day 7 rats were randomized into control, HI, and HI+TAK-242 (TAK-242) groups. The HIBD model was developed using the Rice-Vannucci method (the left side was the ipsilateral side of HI). TAK-242 (0.5 mg/kg) was given to rat pups in the TAK-242 group at 30 min before modeling. Immunofluorescence, immunohistochemistry, and western blotting were used to determine the TLR4 expression; the number of Iba-1+, GFAP+, CD161+, MPO+, and CD3+ cells; ICAM-1 and C3a expression; and interleukin (IL)-1β, tumor necrosis factor (TNF)-α, and IL-10 expression in the hippocampal CA1 region. Result: Significantly increased TLR4 expression was observed in the left hippocampus, and was alleviated by TAK-242. The significant increases in Iba-1+, MPO+, and CD161+ cells at 24 h and 7 days after HI and in GFAP+ and CD3+ T cells at 7 days after HI were also counteracted by TAK-242, but no significant differences were observed among groups at 24 h after HI. ICAM-1 expression increased 24 h after HI, while C3a expression decreased; TAK-242 also alleviated these changes. TNF-α and IL-1β expression increased, while IL-10 expression decreased at 24 h and 7 days after HI; TAK-242 counteracted the increased TNF-α and IL-1β expression at 24 h and the changes in IL-1β and IL-10 at 7 days, but induced no significant differences in IL-10 expression at 24 h and TNF-α expression at 7 days. Conclusion: Early TLR4 inhibition can alleviate hippocampal immune dysfunction after neonatal HIBD.
Collapse
Affiliation(s)
- Zhu Xing
- Department of Neonatology, Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, 100045 China
| | - Tang Zhen
- Department of Neonatology, Affiliated Hospital of Guilin Medical College, Guilin, Guangxi, 541001 China.,Department of Pediatrics, the Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013 China
| | - Fan Jie
- Department of Neonatology, East Hospital of Shaoyang Central Hospital, Shaoyang, Hunan, 422000 China
| | - Yu Jie
- Department of Neonatology, Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, 100045 China
| | - Liu Shiqi
- Department of Neonatology, Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, 100045 China
| | - Zhu Kaiyi
- Department of Neonatology, Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, 100045 China
| | - OuYang Zhicui
- Department of Neonatology, Affiliated Hospital of Guilin Medical College, Guilin, Guangxi, 541001 China
| | - Hei Mingyan
- Department of Neonatology, Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, 100045 China
| |
Collapse
|
20
|
Bu Y, Li WS, Lin J, Wei YW, Sun QY, Zhu SJ, Tang ZS. Electroacupuncture Attenuates Immune-Inflammatory Response in Hippocampus of Rats with Vascular Dementia by Inhibiting TLR4/MyD88 Signaling Pathway. Chin J Integr Med 2021; 28:153-161. [PMID: 34913150 PMCID: PMC8672855 DOI: 10.1007/s11655-021-3350-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/22/2021] [Indexed: 11/30/2022]
Abstract
Objective To investigate whether electroacupuncture (EA) alleviates cognitive impairment by suppressing the toll-like receptor 4 (TLR4)/myeloid differentiation factor 88 (MyD88) signaling pathway, which triggers immune-inflammatory responses in the hippocampus of rats with vascular dementia (VaD). Methods The experiments were conducted in 3 parts and in total the Sprague-Dawley rats were randomly divided into 8 groups by a random number table, including sham, four-vessel occlusion (4-VO), 4-VO+EA, 4-VO+non-EA, sham+EA, 4-VO+lipopolysaccharide (LPS), 4-VO+LPS+EA, and 4-VO+TAK-242 groups. The VaD model was established by the 4-VO method. Seven days later, rats were treated with EA at 5 acupoints of Baihui (DV 20), Danzhong (RN 17), Geshu (BL 17), Qihai (RN 6) and Sanyinjiao (SP 6), once per day for 3 consecutive weeks. Lymphocyte subsets, lymphocyte transformation rates, and inflammatory cytokines interleukin-6 (IL-6) and tumor necrosis factor α(TNF-α) were measured to assess immune function and inflammation in VaD rats. Transmission electron microscopy was used to observe the ultrastructure of nerve cells in the hippocampus. The levels of TLR4, MyD88, IL-6, and TNF-α were detected after EA treatment. TLR4/MyD88 signaling and cognitive function were also assessed after intracerebroventricular injection of TLR4 antagonist TAK-242 or TLR4 agonist LPS with or without EA. Results Compared with the 4-VO group, EA notably improved immune function of rats in the 4-VO+EA group, inhibited the protein and mRNA expressions of TLR4 and MyD88 in the hippocampus of rats, reduced the expressions of serum IL-6 and TNF-α (all P<0.05 or P<0.01), and led to neuronal repair in the hippocampus. There were no significant differences between the 4-VO+LPS+EA and 4-VO+EA groups, nor between the 4-VO+TAK-242 and 4-VO+EA groups (P>0.05). Conclusions EA attenuated cognitive impairment associated with immune inflammation by inhibition of the TLR4/MyD88 signaling pathway. Thus, EA may be a promising alternative therapy for the treatment of VaD. Electronic Supplementary Material Supplementary material (Appendixes 1–4) is available in the online version of this article at 10.1007/s11655-021-3350-5.
Collapse
Affiliation(s)
- Yu Bu
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Wen-Shuang Li
- Department of Hematology, the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Ji Lin
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Yu-Wei Wei
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Qiu-Ying Sun
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Shi-Jie Zhu
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Zhong-Sheng Tang
- School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China.
| |
Collapse
|
21
|
Kumar SP, Babu PP. NADPH Oxidase: a Possible Therapeutic Target for Cognitive Impairment in Experimental Cerebral Malaria. Mol Neurobiol 2021; 59:800-820. [PMID: 34782951 DOI: 10.1007/s12035-021-02598-1] [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: 06/02/2021] [Accepted: 10/12/2021] [Indexed: 12/19/2022]
Abstract
Long-term cognitive impairment associated with seizure-induced hippocampal damage is the key feature of cerebral malaria (CM) pathogenesis. One-fourth of child survivors of CM suffer from long-lasting neurological deficits and behavioral anomalies. However, mechanisms on hippocampal dysfunction are unclear. In this study, we elucidated whether gp91phox isoform of nicotinamide adenine dinucleotide phosphate oxidase 2 (NOX2) (a potent marker of oxidative stress) mediates hippocampal neuronal abnormalities and cognitive dysfunction in experimental CM (ECM). Mice symptomatic to CM were rescue treated with artemether monotherapy (ARM) and in combination with apocynin (ARM + APO) adjunctive based on scores of Rapid Murine Come behavior Scale (RMCBS). After a 30-day survivability period, we performed Barnes maze, T-maze, and novel object recognition cognitive tests to evaluate working and reference memory in all the experimental groups except CM. Sensorimotor tests were conducted in all the cohorts to assess motor coordination. We performed Golgi-Cox staining to illustrate cornu ammonis-1 (CA1) pyramidal neuronal morphology and study overall hippocampal neuronal density changes. Further, expression of NOX2, NeuN (neuronal marker) in hippocampal CA1 and dentate gyrus was determined using double immunofluorescence experiments in all the experimental groups. Mice administered with ARM monotherapy and APO adjunctive treatment exhibited similar survivability. The latter showed better locomotor and cognitive functions, reduced ROS levels, and hippocampal NOX2 immunoreactivity in ECM. Our results show a substantial increase in hippocampal NeuN immunoreactivity and dendritic arborization in ARM + APO cohorts compared to ARM-treated brain samples. Overall, our study suggests that overexpression of NOX2 could result in loss of hippocampal neuronal density and dendritic spines of CA1 neurons affecting the spatial working and reference memory during ECM. Notably, ARM + APO adjunctive therapy reversed the altered neuronal morphology and oxidative damage in hippocampal neurons restoring long-term cognitive functions after CM.
Collapse
Affiliation(s)
- Simhadri Praveen Kumar
- F-23/71, Neuroscience Laboratory, Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, 500 046, India
| | - Phanithi Prakash Babu
- F-23/71, Neuroscience Laboratory, Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, 500 046, India.
| |
Collapse
|
22
|
Fang J, Sheng R, Qin ZH. NADPH Oxidases in the Central Nervous System: Regional and Cellular Localization and the Possible Link to Brain Diseases. Antioxid Redox Signal 2021; 35:951-973. [PMID: 34293949 DOI: 10.1089/ars.2021.0040] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Significance: The significant role of reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (Nox) in signal transduction is mediated by the production of reactive oxygen species (ROS), especially in the central nervous system (CNS). The pathogenesis of some neurologic and psychiatric diseases is regulated by ROS, acting as a second messenger or pathogen. Recent Advances: In the CNS, the involvement of Nox-derived ROS has been implicated in the regulation of multiple signals, including cell survival/apoptosis, neuroinflammation, migration, differentiation, proliferation, and synaptic plasticity, as well as the integrity of the blood/brain barrier. In these processes, the intracellular signals mediated by the members of the Nox family vary among different tissues. The present review illuminates the regions and cellular, subcellular localization of Nox isoforms in the brain, the signal transduction, and the role of NOX enzymes in pathophysiology, respectively. Critical Issues: Different signal transduction cascades are coupled to ROS derived from various Nox homologues with varying degrees. Therefore, a critical issue worth noting is the varied role of the homologues of NOX enzymes in different signaling pathways and also they mediate different phenotypes in the diverse pathophysiological condition. This substantiates the effectiveness of selective Nox inhibitors in the CNS. Future Directions: Further investigation to elucidate the role of various homologues of NOX enzymes in acute and chronic brain diseases and signaling mechanisms, and the development of more specific NOX inhibitors for the treatment of CNS disease are urgently needed. Antioxid. Redox Signal. 35, 951-973.
Collapse
Affiliation(s)
- Jie Fang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, China
| | - Rui Sheng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, China
| | - Zheng-Hong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, China
| |
Collapse
|
23
|
Chen YH, Chen YC, Hwang LL, Yang LY, Lu DY. Deficiency in Androgen Receptor Aggravates Traumatic Brain Injury-Induced Pathophysiology and Motor Deficits in Mice. Molecules 2021; 26:6250. [PMID: 34684832 PMCID: PMC8537172 DOI: 10.3390/molecules26206250] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/12/2021] [Accepted: 10/13/2021] [Indexed: 11/16/2022] Open
Abstract
Androgens have been shown to have a beneficial effect on brain injury and lower reactive astrocyte expression after TBI. Androgen receptors (ARs) are known to mediate the neuroprotective effects of androgens. However, whether ARs play a crucial role in TBI remains unknown. In this study, we investigated the role of ARs in TBI pathophysiology, using AR knockout (ARKO) mice. We used the controlled cortical impact model to produce primary and mechanical brain injuries and assessed motor function and brain-lesion volume. In addition, the AR knockout effects on necrosis and autophagy were evaluated after TBI. AR knockout significantly increased TBI-induced expression of the necrosis marker alpha-II-spectrin breakdown product 150 and astrogliosis marker glial fibrillary acidic protein. In addition, the TBI-induced astrogliosis increase in ARKO mice lasted for three weeks after a TBI. The autophagy marker Beclin-1 was also enhanced in ARKO mice compared with wild-type mice after TBI. Our results also indicated that ARKO mice showed a more unsatisfactory performance than wild-type mice in a motor function test following TBI. Further, they were observed to have more severe lesions than wild-type mice after injury. These findings strongly suggest that ARs play a role in TBI.
Collapse
Affiliation(s)
- Yu-Hsin Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-H.C.); (Y.-C.C.); (L.-L.H.)
| | - Yen-Chou Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-H.C.); (Y.-C.C.); (L.-L.H.)
| | - Ling-Ling Hwang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (Y.-H.C.); (Y.-C.C.); (L.-L.H.)
| | - Liang-Yo Yang
- Department of Physiology, School of Medicine, College of Medicine, China Medical University, Taichung 40402, Taiwan
- Laboratory of Neural Repair, Department of Medical Research, China Medical University Hospital, Taichung 40447, Taiwan
| | - Dah-Yuu Lu
- Department of Pharmacology, School of Medicine, China Medical University, Taichung 404333, Taiwan
- Department of Photonics and Communication Engineering, Asia University, Taichung 404333, Taiwan
| |
Collapse
|
24
|
McCarty MF, Lerner A. The second phase of brain trauma can be controlled by nutraceuticals that suppress DAMP-mediated microglial activation. Expert Rev Neurother 2021; 21:559-570. [PMID: 33749495 DOI: 10.1080/14737175.2021.1907182] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION A delayed second wave of brain trauma is mediated in large part by microglia that are activated to a pro-inflammatory M1 phenotype by DAMP proteins released by dying neurons. These microglia can promote apoptosis or necrosis in neighboring neurons by producing a range of pro-inflammatory cytokines and the deadly oxidant peroxynitrite. This second wave could therefore be mitigated with agents that blunt the post-traumatic M1 activation of microglia and that preferentially promote a pro-healing M2 phenotype. AREAS COVERED The literature on nutraceuticals that might have clinical potential in this regard. EXPERT OPINION The chief signaling pathway whereby DAMPs promote M1 microglial activation involves activation of toll-like receptor 4 (TLR4), NADPH oxidase, NF-kappaB, and the stress activated kinases JNK and p38. The green tea catechin EGCG can suppress TLR4 expression. Phycocyanobilin can inhibit NOX2-dependent NADPH oxidase, ferulate and melatonin can oppose pro-inflammatory signal modulation by NADPH oxidase-derived oxidants. Long-chain omega-3 fatty acids, the soy isoflavone genistein, the AMPK activator berberine, glucosamine, and ketone bodies can down-regulate NF-kappaB activation. Vitamin D activity can oppose JNK/p38 activation. A sophisticated program of nutraceutical supplementation may have important potential for mitigating the second phase of neuronal death and aiding subsequent healing.
Collapse
Affiliation(s)
- Mark F McCarty
- Department of research, Catalytic Longevity Foundation, San Diego, California, USA
| | - Aaron Lerner
- Chaim Sheba Medical Center, The Zabludowicz Research Center for Autoimmune Diseases, Tel Hashomer, Israel
| |
Collapse
|
25
|
Zhao M, Wang Y, Li L, Liu S, Wang C, Yuan Y, Yang G, Chen Y, Cheng J, Lu Y, Liu J. Mitochondrial ROS promote mitochondrial dysfunction and inflammation in ischemic acute kidney injury by disrupting TFAM-mediated mtDNA maintenance. Theranostics 2021; 11:1845-1863. [PMID: 33408785 PMCID: PMC7778599 DOI: 10.7150/thno.50905] [Citation(s) in RCA: 449] [Impact Index Per Article: 112.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/11/2020] [Indexed: 02/05/2023] Open
Abstract
Aims: Ischemia-reperfusion injury (IRI)-induced acute kidney injury (IRI-AKI) is characterized by elevated levels of reactive oxygen species (ROS), mitochondrial dysfunction, and inflammation, but the potential link among these features remains unclear. In this study, we aimed to investigate the specific role of mitochondrial ROS (mtROS) in initiating mitochondrial DNA (mtDNA) damage and inflammation during IRI-AKI. Methods: The changes in renal function, mitochondrial function, and inflammation in IRI-AKI mice with or without mtROS inhibition were analyzed in vivo. The impact of mtROS on TFAM (mitochondrial transcription factor A), Lon protease, mtDNA, mitochondrial respiration, and cytokine release was analyzed in renal tubular cells in vitro. The effects of TFAM knockdown on mtDNA, mitochondrial function, and cytokine release were also analyzed in vitro. Finally, changes in TFAM and mtDNA nucleoids were measured in kidney samples from IRI-AKI mice and patients. Results: Decreasing mtROS levels attenuated renal dysfunction, mitochondrial damage, and inflammation in IRI-AKI mice. Decreasing mtROS levels also reversed the decrease in TFAM levels and mtDNA copy number that occurs in HK2 cells under oxidative stress. mtROS reduced the abundance of mitochondrial TFAM in HK2 cells by suppressing its transcription and promoting Lon-mediated TFAM degradation. Silencing of TFAM abolished the Mito-Tempo (MT)-induced rescue of mitochondrial function and cytokine release in HK2 cells under oxidative stress. Loss of TFAM and mtDNA damage were found in kidneys from IRI-AKI mice and AKI patients. Conclusion: mtROS can promote renal injury by suppressing TFAM-mediated mtDNA maintenance, resulting in decreased mitochondrial energy metabolism and increased cytokine release. TFAM defects may be a promising target for renal repair after IRI-AKI.
Collapse
|
26
|
Sha H, Ma Y, Tong Y, Zhao J, Qin F. Apocynin inhibits placental TLR4/NF-κB signaling pathway and ameliorates preeclampsia-like symptoms in rats. Pregnancy Hypertens 2020; 22:210-215. [PMID: 33099123 DOI: 10.1016/j.preghy.2020.10.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 10/04/2020] [Accepted: 10/08/2020] [Indexed: 12/23/2022]
Abstract
OBJECTIVE We aimed to investigate the potency of apocynin in ameliorating preeclampsia and explore the underlying mechanisms. METHODS Preeclampsia model was constructed in rats by administering 200 mg/kg/day L-NAME. Apocynin was given orally in drinking water. Systolic blood pressure and proteinuria were monitored during treatment. Survival rate rate of the pups and placental weight were assessed. Serum sFlt-1, PIGF, IL-6 and placental TLR4 levels were measured using ELISA or qRT-PCR. RESULTS Apocynin dose-dependently decreased systolic blood pressure and proteinuria during gestation. Survival rate of the pups and placental weight were improved by apocynin treatment. Apocynin ameliorated the imbalance of sFlt-1 and PIGF in serum and placenta of rats with preeclampsia. Apocynin attenuated serum inflammatory cytokine expression and placental inflammation most likely due to downregulation of the placental TLR4/NF-kB pathway in L-NAME treated rats. CONCLUSIONS Apocynin potently ameliorates the L-NAME-induced preeclampsia, which is achieved by re-balancing the sFlt-1 and PIGF levels, attenuating inflammation, and inhibiting TLR4/NF-κB p65 signaling.
Collapse
Affiliation(s)
- Han Sha
- Department of Obstetrics and Gynecology, Heze Municipal Hospital, No. 2888 Caozhou Road, Mudan District, Heze 274000, Shandong, China
| | - Yanchao Ma
- Department of Obstetrics and Gynecology, Heze Municipal Hospital, No. 2888 Caozhou Road, Mudan District, Heze 274000, Shandong, China
| | - Yuli Tong
- Department of Obstetrics and Gynecology, Heze Municipal Hospital, No. 2888 Caozhou Road, Mudan District, Heze 274000, Shandong, China
| | - Jie Zhao
- Department of Obstetrics and Gynecology, Heze Municipal Hospital, No. 2888 Caozhou Road, Mudan District, Heze 274000, Shandong, China
| | - Fengzhi Qin
- Department of Obstetrics and Gynecology, Heze Municipal Hospital, No. 2888 Caozhou Road, Mudan District, Heze 274000, Shandong, China.
| |
Collapse
|
27
|
Eastman CL, D'Ambrosio R, Ganesh T. Modulating neuroinflammation and oxidative stress to prevent epilepsy and improve outcomes after traumatic brain injury. Neuropharmacology 2020; 172:107907. [PMID: 31837825 PMCID: PMC7274911 DOI: 10.1016/j.neuropharm.2019.107907] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 11/26/2019] [Accepted: 12/05/2019] [Indexed: 12/14/2022]
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability in young adults worldwide. TBI survival is associated with persistent neuropsychiatric and neurological impairments, including posttraumatic epilepsy (PTE). To date, no pharmaceutical treatment has been found to prevent PTE or ameliorate neurological/neuropsychiatric deficits after TBI. Brain trauma results in immediate mechanical damage to brain cells and blood vessels that may never be fully restored given the limited regenerative capacity of brain tissue. This primary insult unleashes cascades of events, prominently including neuroinflammation and massive oxidative stress that evolve over time, expanding the brain injury, but also clearing cellular debris and establishing homeostasis in the region of damage. Accumulating evidence suggests that oxidative stress and neuroinflammatory sequelae of TBI contribute to posttraumatic epileptogenesis. This review will focus on possible roles of reactive oxygen species (ROS), their interactions with neuroinflammation in posttraumatic epileptogenesis, and emerging therapeutic strategies after TBI. We propose that inhibitors of the professional ROS-generating enzymes, the NADPH oxygenases and myeloperoxidase alone, or combined with selective inhibition of cyclooxygenase mediated signaling may have promise for the treatment or prevention of PTE and other sequelae of TBI. This article is part of the special issue entitled 'New Epilepsy Therapies for the 21st Century - From Antiseizure Drugs to Prevention, Modification and Cure of Epilepsy'.
Collapse
Affiliation(s)
- Clifford L Eastman
- Department of Neurological Surgery, 325 Ninth Ave., Seattle, WA, 98104, USA.
| | - Raimondo D'Ambrosio
- Department of Neurological Surgery, 325 Ninth Ave., Seattle, WA, 98104, USA; Regional Epilepsy Center, University of Washington, 325 Ninth Ave., Seattle, WA, 98104, USA
| | - Thota Ganesh
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Rd, Atlanta, GA, 30322, Georgia.
| |
Collapse
|
28
|
TAK‑242 exerts a neuroprotective effect via suppression of the TLR4/MyD88/TRIF/NF‑κB signaling pathway in a neonatal hypoxic‑ischemic encephalopathy rat model. Mol Med Rep 2020; 22:1440-1448. [PMID: 32627010 PMCID: PMC7339810 DOI: 10.3892/mmr.2020.11220] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 04/06/2020] [Indexed: 12/29/2022] Open
Abstract
Neonatal hypoxic-ischemic encephalopathy (HIE) is one of the main causes of death and nervous system damage in neonates. The aim of the present study was to investigate the effect of the Toll-like receptor 4 (TLR4) antagonist TAK-242 on HIE. The Rice-Vannucci method was used for ligation of the left common carotid artery, followed by hypoxic treatment for 2.5 h to establish a neonatal HIE rat model. Rats were intraperitoneally injected with 7.5 ml/kg TAK-242 after hypoxia-ischemia. It was demonstrated that TAK-242 significantly reduced the infarct volume and cerebral edema content of neonatal rats after HIE, alleviating neuronal damage and neurobehavioral function deficits. Furthermore, TAK-242 decreased the protein expression levels of TLR4, MyD88, TIR-domain-containing adapter-inducing interferon-β (TRIF), NF-κB, tumor necrosis factor α (TNF-α) and interleukin-1β in the hippocampus. The present results suggested that TAK-242 may exert a neuroprotective effect after HIE by inhibiting the TLR4/MyD88/TRIF/NF-κB signaling pathway, and reducing the release of downstream inflammatory cytokines.
Collapse
|
29
|
Li Y, He Z, Lv H, Chen W, Chen J. Calpain-2 plays a pivotal role in the inhibitory effects of propofol against TNF-α-induced autophagy in mouse hippocampal neurons. J Cell Mol Med 2020; 24:9287-9299. [PMID: 32627970 PMCID: PMC7417688 DOI: 10.1111/jcmm.15577] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 06/05/2020] [Accepted: 06/10/2020] [Indexed: 12/18/2022] Open
Abstract
Calpains are calcium‐dependent proteases and play critical roles in neuronal autophagy induced by inflammation. Propofol has been reported to exert anti‐inflammatory effects in neurons. We aimed to identify whether and how propofol‐modulated calpain activity and neuron autophagy in response to tumour necrosis factor‐α (TNF‐α). Mouse hippocampal neurons were pre‐treated with propofol and exposed to TNF‐α. Autophagy was evaluated by fluorescent autophagy assay and by measuring LC3I and LC3II expression. Intracellular calcium concentration was measured by fluorescent assay. Calpain activation was measured by calpain activity assay. The protein expression of intracellular signalling molecules was detected by Western blot analysis. Compared with untreated control neurons, 40 ng/mL TNF‐α treatment for 2 hours induced neuron autophagy, which was attenuated by 25 μmol/L propofol. TNF‐α induced intracellular calcium accumulation, the phosphorylation of calcium/calmodulin‐dependent protein kinase II (CAMK II) and calpain‐2, calpain activation and lysosomal cathepsin B release as well as tyrosine kinase receptor B (TrkB) truncation. These effects were alleviated by propofol, calcium chelator, CAMK II inhibitor, calpain‐2 inhibitor, calpain‐2 siRNA transfection and N‐Methyl‐d‐aspartic acid (NMDA) receptor antagonist. Propofol, via NMDA receptor, inhibited TNF‐α‐mediated hippocampal neuron autophagy. The mechanism may involve calcium and calcium‐dependent signalling pathway, especially CAMK II and calpain‐2.
Collapse
Affiliation(s)
- Ying Li
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhiyong He
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hu Lv
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Chen
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiawei Chen
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
30
|
VX765 Attenuates Pyroptosis and HMGB1/TLR4/NF- κB Pathways to Improve Functional Outcomes in TBI Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7879629. [PMID: 32377306 PMCID: PMC7181015 DOI: 10.1155/2020/7879629] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 02/28/2020] [Accepted: 03/20/2020] [Indexed: 01/12/2023]
Abstract
Background Traumatic brain injury (TBI) refers to temporary or permanent damage to brain function caused by penetrating objects or blunt force trauma. TBI activates inflammasome-mediated pathways and other cell death pathways to remove inactive and damaged cells, however, they are also harmful to the central nervous system. The newly discovered cell death pattern termed pyroptosis has become an area of interest. It mainly relies on caspase-1-mediated pathways, leading to cell death. Methods Our research focus is VX765, a known caspase-1 inhibitor which may offer neuroprotection after the process of TBI. We established a controlled cortical impact (CCI) mouse model and then controlled the degree of pyroptosis in TBI with VX765. The effects of caspase-1 inhibition on inflammatory response, pyroptosis, blood-brain barrier (BBB), apoptosis, and microglia activation, in addition to neurological deficits, were investigated. Results We found that TBI led to NOD-like receptors (NLRs) as well as absent in melanoma 2 (AIM2) inflammasome-mediated pyroptosis in the damaged cerebral cortex. VX765 curbed the expressions of indispensable inflammatory subunits (caspase-1 as well as key downstream proinflammatory cytokines such as interleukin- (IL-) 1β and IL-18). It also inhibited gasdermin D (GSDMD) cleavage and apoptosis-associated spot-like protein (ASC) oligomerization in the injured cortex. In addition to the above, VX765 also inhibited the inflammatory activity of the high-mobility cassette -1/Toll-like receptor 4/nuclear factor-kappa B (HMGB1/TLR4/NF-kappa B) pathway. By inhibiting pyroptosis and inflammatory mediator expression, we demonstrated that VX765 can decrease blood-brain barrier (BBB) leakage, apoptosis, and microglia polarization to exhibit its neuroprotective effects. Conclusion In conclusion, VX765 can counteract neurological damage after TBI by reducing pyroptosis and HMGB1/TLR4/NF-κB pathway activities. VX765 may have a good therapeutic effect on TBI.
Collapse
|
31
|
Rana P, Rama Rao KV, Ravula A, Trivedi R, D'Souza M, Singh AK, Gupta RK, Chandra N. Oxidative stress contributes to cerebral metabolomic profile changes in animal model of blast-induced traumatic brain injury. Metabolomics 2020; 16:39. [PMID: 32166461 DOI: 10.1007/s11306-020-1649-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 02/02/2020] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Blast-induced neurotrauma (BINT) has been recognized as the common mode of traumatic brain injury amongst military and civilian personnel due to an increased insurgent activity domestically and abroad. Previous studies from this laboratory have identified three major pathological events following BINT which include blood brain barrier disruption the earliest event, followed by oxidative stress and neuroinflammation as secondary events occurring a few hours following blast. OBJECTIVES Our recent studies have also identified an increase in oxidative stress mediated by the activation of superoxide producing enzyme NADPH oxidase (NOX) in different brain regions at varying levels with neurons displaying higher oxidative stress (NOX activation) compared to any other neural cell. Since neurons have higher energy demands in brain and are more prone to oxidative damage, this study evaluated the effect of oxidative stress on blast-blast induced changes in metabolomics profiles in different brain regions. METHODS Animals were exposed to mild/moderate blast injury (180 kPa) and examined the metabolites of energy metabolism, amino acid metabolism as well as the profiles of plasma membrane metabolites in different brain regions at different time points (24 h, 3 day and 7 day) after blast using 1H NMR spectroscopy. Effect of apocynin, an inhibitor of superoxide producing enzyme NADPH oxidase on cerebral metabalomics profiles was also examined. RESULTS Several metabolomic profile changes were observed in frontal cortex and hippocampus with concomitant decrease in energy metabolism. In addition, glutamate/glutamine and other amino acid metabolism as well as metabolites involved in plasma membrane integrity were also altered. Hippocampus appears metabolically more vulnerable than the frontal cortex. A post-treatment of animals with apocynin, an inhibitor of NOX activation significantly prevented the changes in metabolite profiles. CONCLUSION Together these studies indicate that blast injury reduces both cerebral energy and neurotransmitter amino acid metabolism and that oxidative stress contributes to these processes. Thus, strategies aimed at reducing oxidative stress can have a therapeutic benefit in mitigating metabolic changes following BINT.
Collapse
Affiliation(s)
- Poonam Rana
- Metabolomics Research Facility, Division of Behavioral Neuroscience, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Kakulavarapu V Rama Rao
- Center for Injury Biomechanics, Materials and Medicine (CIBM3), Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102-1982, USA
| | - Arunreddy Ravula
- Center for Injury Biomechanics, Materials and Medicine (CIBM3), Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102-1982, USA
| | - Richa Trivedi
- Metabolomics Research Facility, Division of Behavioral Neuroscience, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Maria D'Souza
- Department of NMR, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Ajay K Singh
- Metabolomics Research Facility, Division of Behavioral Neuroscience, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Raj K Gupta
- US Department of Defense Blast Injury Research Program Coordinating Office, US Army MRMC, 504 Scott Street, Fort Detrick, MD, USA.
| | - Namas Chandra
- US Department of Defense Blast Injury Research Program Coordinating Office, US Army MRMC, 504 Scott Street, Fort Detrick, MD, USA.
- Center for Injury Biomechanics, Materials and Medicine (CIBM3), Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102-1982, USA.
| |
Collapse
|
32
|
Wang M, Luo L. An Effective NADPH Oxidase 2 Inhibitor Provides Neuroprotection and Improves Functional Outcomes in Animal Model of Traumatic Brain Injury. Neurochem Res 2020; 45:1097-1106. [PMID: 32072445 DOI: 10.1007/s11064-020-02987-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/07/2020] [Accepted: 02/11/2020] [Indexed: 12/23/2022]
Abstract
Traumatic brain injury (TBI) has become a leading cause of death and disability all over the world. Pharmacological suppression of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 (NOX2) can inhibit oxidative stress which is implicated in the pathology of TBI. GSK2795039 was reported to target NOX2 to inhibit [Formula: see text] and ROS production. The present study aimed to investigate the effect of GSK2795039 on NOX2 activity and neurological deficits in a TBI mouse model. TBI mouse model was established by a weight-drop to mouse skull. GSK2795039 at a dose of 100 mg/kg was administrated to mice 30 min before TBI. NOX2 expression and activity were detected by Western blot and biochemical method. Neurological damage and apoptosis were detected by behavioral test and terminal deoxynucleotidyl transferase dUTP nick end labeling staining. GSK2795039 significantly inhibited NOX2 expression and activity in the TBI mouse model. It also attenuated TBI-induced neurological deficits, apoptosis, and neurological recovery. The results indicate that GSK2795039 can be used as a potential drug for TBI treatment.
Collapse
Affiliation(s)
- Mengwei Wang
- Department of Emergency, The Fourth Affiliated Hospital of China Medical University, No. 4 Chongshan East Road, Huanggu District, Shenyang, 110032, Liaoning, China.
| | - Le Luo
- Shanghai Zhuole Biotechnology Center, No. 2066 Wangyuan Road, Shanghai, 201499, China
| |
Collapse
|
33
|
Jiang LJ, Xu ZX, Wu MF, Dong GQ, Zhang LL, Gao JY, Feng CX, Feng X. Resatorvid protects against hypoxic-ischemic brain damage in neonatal rats. Neural Regen Res 2020; 15:1316-1325. [PMID: 31960818 PMCID: PMC7047798 DOI: 10.4103/1673-5374.272615] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Secondary brain damage caused by hyperactivation of autophagy and inflammatory responses in neurons plays an important role in hypoxic-ischemic brain damage (HIBD). Although previous studies have implicated Toll-like receptor 4 (TLR4) and nuclear factor kappa-B (NF-κB) in the neuroinflammatory response elicited by brain injury, the role and mechanisms of the TLR4-mediated autophagy signaling pathway in neonatal HIBD are still unclear. We hypothesized that this pathway can regulate brain damage by modulating neuron autophagy and neuroinflammation in neonatal rats with HIBD. Hence, we established a neonatal HIBD rat model using the Rice-Vannucci method, and injected 0.75, 1.5, or 3 mg/kg of the TLR4 inhibitor resatorvid (TAK-242) 30 minutes after hypoxic ischemia. Our results indicate that administering TAK-242 to neonatal rats after HIBD could significantly reduce the infarct volume and the extent of cerebral edema, alleviate neuronal damage and neurobehavioral impairment, and decrease the expression levels of TLR4, phospho-NF-κB p65, Beclin-1, microtubule-associated protein l light chain 3, tumor necrosis factor-α, and interleukin-1β in the hippocampus. Thus, TAK-242 appears to exert a neuroprotective effect after HIBD by inhibiting activation of autophagy and the release of inflammatory cytokines via inhibition of the TLR4/NF-κB signaling pathway. This study was approved by the Laboratory Animal Ethics Committee of Affiliated Hospital of Yangzhou University, China (approval No. 20180114-15) on January 14, 2018.
Collapse
Affiliation(s)
- Li-Jun Jiang
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou; Department of Neonatology, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Zhen-Xing Xu
- Department of Neonatology, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Ming-Fu Wu
- Department of Neonatology, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Gai-Qin Dong
- Department of Neonatology, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Li-Li Zhang
- Department of Neonatology, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Jun-Yan Gao
- Department of Neonatology, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Chen-Xi Feng
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Xing Feng
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
34
|
Liu T, Zheng W, Wang L, Wang L, Zhang Y. TLR4/NF-κB Signaling Pathway Participates in the Protective Effects of Apocynin on Gestational Diabetes Mellitus Induced Placental Oxidative Stress and Inflammation. Reprod Sci 2020; 27:722-730. [PMID: 32046403 DOI: 10.1007/s43032-019-00078-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 06/17/2019] [Indexed: 02/07/2023]
Abstract
Gestational diabetes mellitus (GDM) is a temporary form of diabetes during pregnancy which influences the health of both mother and child. Both inflammation and oxidative stress have been implicated in the pathophysiology of GDM. Apocynin, acetophenone with anti-oxidative and anti-inflammation activities, has been shown to protect against insulin resistance. In the current study, the effects of apocynin on GDM symptoms, productive outcomes, oxidative stress, and inflammation were evaluated and the underlying mechanisms were explored. We administrated apocynin to GDM mice and monitored the GDM symptoms including body weight, serum levels of glucose, insulin, lipid profile, and the fetal outcomes in GDM mice. We also evaluated the effects of apocynin on placental oxidative stress, inflammation, and activation of TLR4/NF-κB signaling pathway in GDM mice. Here, we reported that apocynin treatment significantly reduced serum levels of glucose, cholesterol, triglyceride, and low-density lipoprotein in GDM mice, while significantly increased serum level of insulin and high-density lipoprotein. Apocynin improved fetal outcomes in GDM mice. Apocynin ameliorated placental oxidative stress and inflammation and inhibited TLR4/NF-κB signaling pathway activation in GDM mice. We concluded that apocynin suppressed oxidative stress and inflammation in GDM by inhibiting the TLR4/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Teng Liu
- The Affiliated Hospital of Medical School of Ningbo University, No.247, Renmin Road, Jiangbei District, Ningbo, 315020, Zhejiang, China
| | - Wei Zheng
- Ningbo Medical Center Lihuili Eastern Hospital, Ningbo, 315040, Zhejiang, China
| | - Linlin Wang
- Ningbo First Hospital, Ningbo, 315010, Zhejiang, China
| | - Lin Wang
- The Affiliated Hospital of Medical School of Ningbo University, No.247, Renmin Road, Jiangbei District, Ningbo, 315020, Zhejiang, China
| | - Yanke Zhang
- The Affiliated Hospital of Medical School of Ningbo University, No.247, Renmin Road, Jiangbei District, Ningbo, 315020, Zhejiang, China.
| |
Collapse
|
35
|
Parastan RH, Christopher M, Torrys YS, Mahadewa TGB. Combined Therapy Potential of Apocynin and Tert-butylhydroquinone as a Therapeutic Agent to Prevent Secondary Progression to Traumatic Brain Injury. Asian J Neurosurg 2020; 15:10-15. [PMID: 32181166 PMCID: PMC7057894 DOI: 10.4103/ajns.ajns_231_19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 01/23/2020] [Indexed: 11/06/2022] Open
Abstract
Traumatic brain injury is caused by physical collision (primary injury). It changes the brain's biochemistry and disturbs the normal brain function such as memory loss and consciousness disturbance (secondary injury). The severity can be measured with the Glasgow Coma Scale. The secondary injury will cause oxidative stress that leads to the nervous cells death, so treatment is needed before it gets worse. Primary injury results in excess of reactive oxidative stress (ROS) which is known from NADPH oxidase 2 (Nox2). Excessive ROS is deadly to the nerve cells. Excessive ROS will activate nuclear factor erythroid 2-like 2 (Nrf2). Nrf2 will bind to antioxidant response elements, to protect multi organs against ROS, including this brain injury. However, this does not last long, so it requires handling excess ROS. Apocynin can inhibit the activation of Nox2, and reduce the neuron injuries in the hippocampus. It also protects the tissues from oxidative stress. While Nrf2 can be activated by tert-butylhydroquinone, to protect cells. The combination may reduce the secondary brain injury, improve the neurologic recovery, cognitive function, and reduce the secondary cortical lesion.
Collapse
Affiliation(s)
| | - Michael Christopher
- Department of Medicine, Faculty of Medicine, Udayana University, Bali, Indonesia
| | | | | |
Collapse
|
36
|
Singh S, Singh TG. Role of Nuclear Factor Kappa B (NF-κB) Signalling in Neurodegenerative Diseases: An Mechanistic Approach. Curr Neuropharmacol 2020; 18:918-935. [PMID: 32031074 PMCID: PMC7709146 DOI: 10.2174/1570159x18666200207120949] [Citation(s) in RCA: 172] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 05/02/2020] [Accepted: 05/02/2020] [Indexed: 12/12/2022] Open
Abstract
A transcriptional regulatory nuclear factor kappa B (NF-κB) protein is a modulator of cellular biological activity via binding to a promoter region in the nucleus and transcribing various protein genes. The recent research implicated the intensive role of nuclear factor kappa B (NF-κB) in diseases like autoimmune disorder, inflammatory, cardiovascular and neurodegenerative diseases. Therefore, targeting the nuclear factor kappa B (NF-κB) protein offers a new opportunity as a therapeutic approach. Activation of IκB kinase/NF-κB signaling pathway leads to the development of various pathological conditions in human beings, such as neurodegenerative, inflammatory disorders, autoimmune diseases, and cancer. Therefore, the transcriptional activity of IκB kinase/NF- κB is strongly regulated at various cascade pathways. The nuclear factor NF-kB pathway plays a major role in the expression of pro-inflammatory genes, including cytokines, chemokines, and adhesion molecules. In response to the diverse stimuli, the cytosolic sequestered NF-κB in an inactivated form by binding with an inhibitor molecule protein (IkB) gets phosphorylated and translocated into the nucleus further transcribing various genes necessary for modifying various cellular functions. The various researches confirmed the role of different family member proteins of NF-κB implicated in expressing various genes products and mediating various cellular cascades. MicroRNAs, as regulators of NF- κB microRNAs play important roles in the regulation of the inflammatory process. Therefore, the inhibitor of NF-κB and its family members plays a novel therapeutic target in preventing various diseases. Regulation of NF- κB signaling pathway may be a safe and effective treatment strategy for various disorders.
Collapse
Affiliation(s)
- Shareen Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | | |
Collapse
|
37
|
Zeng Z, Zhang Y, Jiang W, He L, Qu H. Modulation of autophagy in traumatic brain injury. J Cell Physiol 2019; 235:1973-1985. [PMID: 31512236 DOI: 10.1002/jcp.29173] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 08/23/2019] [Indexed: 12/20/2022]
Abstract
Traumatic brain injury (TBI) is defined as a traumatically induced structural injury or physiological disruption of brain function as a result of external forces, leading to adult disability and death. A growing body of evidence reveals that alterations in autophagy-related proteins exist extensively in both experimentally and clinically after TBI. Of note, the autophagy pathway plays an essential role in pathophysiological processes, such as oxidative stress, inflammatory response, and apoptosis, thus contributing to neurological properties of TBI. With this in mind, this review summarizes a comprehensive overview on the beneficial and detrimental effects of autophagy in pathophysiological conditions and how these activities are linked to the pathogenesis of TBI. Moreover, the relationship between oxidative stress, inflammation, apoptosis, and autophagy occur TBI. Ultimately, multiple compounds and various drugs targeting the autophagy pathway are well described in TBI. Therefore, autophagy flux represents a potential clinical therapeutic value for the treatment of TBI and its complications.
Collapse
Affiliation(s)
- Zhiqing Zeng
- Department of Neurosurgery, First Affiliated Hospital, University of South China, Hengyang, China
| | - Yao Zhang
- Department of Clinical Laboratory, The First People's Hospital of Changde City, Changde City, Hunan Province, China
| | - Weiping Jiang
- Department of Neurosurgery, First Affiliated Hospital, University of South China, Hengyang, China
| | - Lu He
- Department of Neurosurgery, First Affiliated Hospital, University of South China, Hengyang, China
| | - Hongtao Qu
- Department of Neurosurgery, First Affiliated Hospital, University of South China, Hengyang, China
| |
Collapse
|
38
|
Su X, Ye Y, Yang Y, Zhang K, Bai W, Chen H, Kang E, Kong C, He X. The Effect of SPTLC2 on Promoting Neuronal Apoptosis is Alleviated by MiR-124-3p Through TLR4 Signalling Pathway. Neurochem Res 2019; 44:2113-2122. [PMID: 31372925 DOI: 10.1007/s11064-019-02849-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 07/06/2019] [Accepted: 07/27/2019] [Indexed: 12/21/2022]
Abstract
To investigate the role and mechanism of microRNA-124-3p (miR-124-3p) and serine palmitoyltransferase long chain base subunit 2 (SPTLC2) in neuronal apoptosis induced by mechanical injury. Transient transfection was used to modify the expression of miR-124-3p and SPTLC2. After transfection, neuronal apoptosis was evaluated in an in vitro injury model of primary neurons using TUNEL staining and western blot. The correlation between miR-124-3p and SPTLC2 was identified through a dual luciferase reporter assay in HEK293 cells. A rescue experiment in primary neurons was performed to further confirm the result. To explore the downstream mechanisms, co-immunoprecipitation was performed to identify proteins that interact with SPTLC2 in toll-like receptor 4 (TLR4) signalling pathway. Subsequently, the relative expression levels of TLR4 pathway molecules were measured by western blot. Our results showed that increased miR-124-3p can inhibit neuronal apoptosis, which is opposite to the effect of SPTLC2. In addition, miR-124-3p was proved to negatively regulate SPTLC2 expression and suppress the apoptosis-promoting effect of SPTLC2 via the TLR4 signalling pathway.
Collapse
Affiliation(s)
- Xinhong Su
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 169 Changle Western Road, Xi'an, 710032, Shanxi, China
| | - Yuqin Ye
- Department of Neurosurgery, PLA 921rd Hospital, Changsha, 410000, Hunan, China
| | - Yongxiang Yang
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 169 Changle Western Road, Xi'an, 710032, Shanxi, China
| | - Kailiang Zhang
- Department of Orthopedic Surgery, Orthopedic Oncology Institute of Chinese PLA, Tangdu Hospital, Air Force Military Medical University, Xi'an, 710032, Shanxi, China
| | - Wei Bai
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 169 Changle Western Road, Xi'an, 710032, Shanxi, China
| | - Huijun Chen
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 169 Changle Western Road, Xi'an, 710032, Shanxi, China
| | - Enming Kang
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 169 Changle Western Road, Xi'an, 710032, Shanxi, China
| | - Chuiguang Kong
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 169 Changle Western Road, Xi'an, 710032, Shanxi, China
| | - Xiaosheng He
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 169 Changle Western Road, Xi'an, 710032, Shanxi, China.
| |
Collapse
|
39
|
Zhang Z, Li D, Xu L, Li HP. Sirt1 improves functional recovery by regulating autophagy of astrocyte and neuron after brain injury. Brain Res Bull 2019; 150:42-49. [PMID: 31102754 DOI: 10.1016/j.brainresbull.2019.05.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 05/06/2019] [Accepted: 05/07/2019] [Indexed: 11/26/2022]
Abstract
Traumatic brain injury (TBI) triggers neuronal death mechanisms that significantly induce neuronal loss and neurological dysfunction. Our previous study revealed that Sirt1 could improve the neuroprotective effect by reducing the astrocyte activation after TBI. Nevertheless, the underlying mechanisms of Sirt1 attenuating astrocyte activation still remain unclear. The following study examined whether the protection of Sirt1 in nigrostriatal pathway injury is associated with autophagy regulation. We established a nigrostriatal pathway injury in the mouse brain in order to mimic the traumatic brain injury and up-regulated Sirt1 expression by resveratrol. Consequently, we analyzed the effect of Sirt1 up-regulation on LC3 and monitored the LC3 localization in the astrocytes, microglial cells and neurons. We found that the Sirt1 up-regulation by resveratrol increased the expression of LC3 around the lesion site after injury. Confocal results showed that Sirt1 up-regulation increased the expression of LC3 in astrocytes and decreased the expression in the neurons, while low effect was found on the microglial cells. Moreover, compared the resveratrol treatment groups, a typical nucleocytoplasmic localization with strong distribution in the nucleus (in astrocyte and neurons) was observed in the control group (treated with DMSO). To sum up, our data suggested that regulation of Sirt1 expression could enhance autophagy in the astrocytes and decrease the expression in the neurons. This mechanism, which may probably relate to the distribution of LC3 in cytoplasm and nucleus, provides a new theoretical basis for exploring the neuroprotective mechanism of Sirt1 after brain injury.
Collapse
Affiliation(s)
- Zhuo Zhang
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Dan Li
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Li Xu
- Department of Ophthalmology, Forth People's Hospital, Shenyang, China.
| | - Hong-Peng Li
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China.
| |
Collapse
|
40
|
Wang Y, Tao J, Jiang M, Yao Y. Apocynin ameliorates diabetic retinopathy in rats: Involvement of TLR4/NF-κB signaling pathway. Int Immunopharmacol 2019; 73:49-56. [PMID: 31078925 DOI: 10.1016/j.intimp.2019.04.062] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 04/24/2019] [Accepted: 04/30/2019] [Indexed: 01/01/2023]
Abstract
Diabetic retinopathy is a diabetes complication. During diabetic retinopathy development, the TLR4/NF-κB pathway is up-regulated. Apocynin is a nicotinamide adenine dinucleotide phosphate oxidase blocker which can reduce the superoxide radicals. It is demonstrated that apocynin can inhibit TLR4/NF-κB pathway in rats. We aim to figure out whether apocynin treatment is benefit for the diabetic retinopathy in rat model. The diabetes in rats was induced by streptozotocin. The treatment of apocynin (16 mg/kg/day) or vehicle in diabetic rat model was maintained for 12 weeks. The expression levels of relative genes in this research were shown through Western blot and qRT-PCR. Morphology of the retinas was shown by Hematoxylin-Eosin staining. The treatment of apocynin ameliorated biochemical indexes in diabetic rats and rescued the morphology of the retinas. After a 12 weeks apocynin treatment, the cell apoptosis, oxidative stress, and inflammatory in retina was reduced in diabetic rats. TLR4/NF-κB signaling pathway activity in diabetic rat retina was inhibited by apocynin. Based on our study, the treatment of apocynin ameliorates diabetic retinopathy in rats. The TLR4/NF-κB signaling pathway inhibition by apocynin is involved in this process. This result indicated a great therapeutic potential of apocynin in diabetic retinopathy treatment.
Collapse
Affiliation(s)
- Yixin Wang
- Department of Ophthalmology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi 214023, Jiangsu, China
| | - Jianxin Tao
- Department of Ophthalmology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi 214023, Jiangsu, China
| | - Minfeng Jiang
- Department of Ophthalmology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi 214023, Jiangsu, China
| | - Yong Yao
- Department of Ophthalmology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi 214023, Jiangsu, China.
| |
Collapse
|
41
|
Chao H, Lin C, Zuo Q, Liu Y, Xiao M, Xu X, Li Z, Bao Z, Chen H, You Y, Kochanek PM, Yin H, Liu N, Kagan VE, Bayır H, Ji J. Cardiolipin-Dependent Mitophagy Guides Outcome after Traumatic Brain Injury. J Neurosci 2019; 39:1930-1943. [PMID: 30626699 PMCID: PMC6407296 DOI: 10.1523/jneurosci.3415-17.2018] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 11/21/2018] [Accepted: 12/28/2018] [Indexed: 01/14/2023] Open
Abstract
Mitochondrial energy production is essential for normal brain function. Traumatic brain injury (TBI) increases brain energy demands, results in the activation of mitochondrial respiration, associated with enhanced generation of reactive oxygen species. This chain of events triggers neuronal apoptosis via oxidation of a mitochondria-specific phospholipid, cardiolipin (CL). One pathway through which cells can avoid apoptosis is via elimination of damaged mitochondria by mitophagy. Previously, we showed that externalization of CL to the mitochondrial surface acts as an elimination signal in cells. Whether CL-mediated mitophagy occurs in vivo or its significance in the disease processes are not known. In this study, we showed that TBI leads to increased mitophagy in the human brain, which was also detected using TBI models in male rats. Knockdown of CL synthase, responsible for de novo synthesis of CL, or phospholipid scramblase-3, responsible for CL translocation to the outer mitochondrial membrane, significantly decreased TBI-induced mitophagy. Inhibition of mitochondrial clearance by 3-methyladenine, mdivi-1, or phospholipid scramblase-3 knockdown after TBI led to a worse outcome, suggesting that mitophagy is beneficial. Together, our findings indicate that TBI-induced mitophagy is an endogenous neuroprotective process that is directed by CL, which marks damaged mitochondria for elimination, thereby limiting neuronal death and behavioral deficits.SIGNIFICANCE STATEMENT Traumatic brain injury (TBI) increases energy demands leading to activation of mitochondrial respiration associated with enhanced generation of reactive oxygen species and resultant damage to mitochondria. We demonstrate that the complete elimination of irreparably damaged organelles via mitophagy is activated as an early response to TBI. This response includes translocation of mitochondria phospholipid cardiolipin from the inner membrane to the outer membrane where externalized cardiolipin mediates targeted protein light chain 3-mediated autophagy of damaged mitochondria. Our data on targeting phospholipid scramblase and cardiolipin synthase in genetically manipulated cells and animals strongly support the essential role of cardiolipin externalization mechanisms in the endogenous reparative plasticity of injured brain cells. Furthermore, successful execution and completion of mitophagy is beneficial in the context of preservation of cognitive functions after TBI.
Collapse
Affiliation(s)
- Honglu Chao
- Departments of Neurosurgery and
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15219
| | | | - Qiang Zuo
- Orthopedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | | | - Mengqing Xiao
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS), Shanghai 200031, China
- University of the Chinese Academy of Sciences, CAS, Beijing 100049, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai 201210, China
| | | | | | | | - Huimei Chen
- Department of Medical Genetics, Nanjing University School of Medicine, Nanjing 210093, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing 210029, China
| | | | - Patrick M Kochanek
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15219
| | - Huiyong Yin
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (CAS), Shanghai 200031, China
- University of the Chinese Academy of Sciences, CAS, Beijing 100049, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai 201210, China
- Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing 100022, China
| | | | - Valerian E Kagan
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health
- Laboratory of Navigational Redox Lipidomics and Department of Human Pathology, IM Sechenov Moscow State Medical University, Moscow 119991, Russian Federation, and
| | - Hülya Bayır
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health,
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15219
- Children's Neuroscience Institute, Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania 15224
| | - Jing Ji
- Departments of Neurosurgery and
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15219
| |
Collapse
|
42
|
Pan L, Qian S. Apocynin promotes neural function recovery and suppresses neuronal apoptosis by inhibiting Tlr4/NF-κB signaling pathway in a rat model of cerebral infarction. Int J Immunopathol Pharmacol 2018. [PMCID: PMC6291862 DOI: 10.1177/2058738418817700] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Occlusion of arteries in the brain is a common cause of cerebral infarction which
induces inflammatory response and oxidative stress resulting in neuronal
apoptosis and disruption of neurological function. The present study
investigated the protective roles of an nicotinamide adenine dinucleotide
phosphate oxidase inhibitor, apocynin, against cerebral infarction. Rat went
through a surgery of middle cerebral artery occlusion and a subset of rats was
treated with apocynin by intraperitoneal injection. The volume of cerebral
infarction and water content were measured. Neuronal apoptosis, inflammatory
response, and oxidative stress were assessed following middle cerebral artery
occlusion and apocynin treatment. We found that apocynin significantly improved
neurological function, increased forelimb placement test scores, and suppressed
balance beam walk latency in rats with cerebral infarction. Histological and
biochemistry analysis revealed that apocynin lead to a significant reduction in
the volume of cerebral infarction as well as cerebral water content, suppressed
neuronal apoptosis, oxidative stress, and inflammatory response induced by
middle cerebral artery occlusion. Finally, we found that apocynin suppressed
Tlr4/nuclear factor-k-gene binding signaling pathway that was upregulated in
rats with cerebral infarction. Our results indicate that apocynin may represent
a potent therapeutic strategy in alleviating neurological dysfunctions in
patients with cerebral infarction.
Collapse
Affiliation(s)
- Lemen Pan
- Department of Vascular Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shuxia Qian
- Department of Neurology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
43
|
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: 56] [Impact Index Per Article: 8.0] [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
|
44
|
Zhang L, Wang H. Autophagy in Traumatic Brain Injury: A New Target for Therapeutic Intervention. Front Mol Neurosci 2018; 11:190. [PMID: 29922127 PMCID: PMC5996030 DOI: 10.3389/fnmol.2018.00190] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 05/15/2018] [Indexed: 11/23/2022] Open
Abstract
Traumatic brain injury (TBI) is one of the most devastating forms of brain injury. Many pathological mechanisms such as oxidative stress, apoptosis and inflammation all contribute to the secondary brain damage and poor outcomes of TBI. Current therapies are often ineffective and poorly tolerated, which drive the explore of new therapeutic targets for TBI. Autophagy is a highly conserved intracellular mechanism during evolution. It plays an important role in elimination abnormal intracellular proteins or organelles to maintain cell stability. Besides, autophagy has been researched in various models including TBI. Previous studies have deciphered that regulation of autophagy by different molecules and pathways could exhibit anti-oxidative stress, anti-apoptosis and anti-inflammation effects in TBI. Hence, autophagy is a promising target for further therapeutic development in TBI. The present review provides an overview of current knowledge about the mechanism of autophagy, the frequently used methods to monitor autophagy, the functions of autophagy in TBI as well as its potential molecular mechanisms based on the pharmacological regulation of autophagy.
Collapse
Affiliation(s)
- Li Zhang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Handong Wang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| |
Collapse
|
45
|
Ma MW, Wang J, Dhandapani KM, Wang R, Brann DW. NADPH oxidases in traumatic brain injury - Promising therapeutic targets? Redox Biol 2018; 16:285-293. [PMID: 29571125 PMCID: PMC5952873 DOI: 10.1016/j.redox.2018.03.005] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 03/09/2018] [Accepted: 03/10/2018] [Indexed: 12/22/2022] Open
Abstract
Traumatic brain injury (TBI) is a major cause of death and disability worldwide. Despite intense investigation, no neuroprotective agents for TBI have yet translated to the clinic. Recent efforts have focused on identifying potential therapeutic targets that underlie the secondary TBI pathology that evolves minutes to years following the initial injury. Oxidative stress is a key player in this complex cascade of secondary injury mechanisms and prominently contributes to neurodegeneration and neuroinflammation. NADPH oxidase (NOX) is a family of enzymes whose unique function is to produce reactive oxygen species (ROS). Human post-mortem and animal studies have identified elevated NOX2 and NOX4 levels in the injured brain, suggesting that these two NOXs are involved in the pathogenesis of TBI. In support of this, NOX2 and NOX4 deletion studies have collectively revealed that targeting NOX enzymes can reduce oxidative stress, attenuate neuroinflammation, promote neuronal survival, and improve functional outcomes following TBI. In addition, NOX inhibitor studies have confirmed these findings and demonstrated an extended critical window of efficacious TBI treatment. Finally, the translational potential, caveats, and future directions of the field are highlighted and discussed throughout the review.
Collapse
Affiliation(s)
- Merry W Ma
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Jing Wang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Krishnan M Dhandapani
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Ruimin Wang
- Department of Neurobiology, North China University of Science and Technology, Tangshan, Hebei, China
| | - Darrell W Brann
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
46
|
Wolf MS, Bayır H, Kochanek PM, Clark RSB. The role of autophagy in acute brain injury: A state of flux? Neurobiol Dis 2018; 122:9-15. [PMID: 29704549 DOI: 10.1016/j.nbd.2018.04.018] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 04/18/2018] [Accepted: 04/24/2018] [Indexed: 12/22/2022] Open
Abstract
It is established that increased autophagy is readily detectable after various types of acute brain injury, including trauma, focal and global cerebral ischemia. What remains controversial, however, is whether this heightened detection of autophagy in brain represents a homeostatic or pathologic process, or an epiphenomenon. The ultimate role of autophagy after acute brain injury likely depends upon: 1) the degree of brain injury and the overall autophagic burden; 2) the capacity of individual cell types to ramp up autophagic flux; 3) the local redox state and signaling of parallel cell death pathways; 4) the capacity to eliminate damage associated molecular patterns and toxic proteins and metabolites both intra- and extracellularly; and 5) the timing of the pro- or anti-autophagic intervention. In this review, we attempt to reconcile conflicting studies that support both a beneficial and detrimental role for autophagy in models of acute brain injury.
Collapse
Affiliation(s)
- Michael S Wolf
- Department of Critical Care Medicine, Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - Hülya Bayır
- Department of Critical Care Medicine, Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, 4401 Penn Avenue, Pittsburgh, PA 15224, USA; Department of Pediatrics, University of Pittsburgh School of Medicine, 4401 Penn Avenue, Pittsburgh, PA 15224, USA; Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, 100 Technology Drive, Pittsburgh, PA 15219, USA; Brain Care Institute, Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - Patrick M Kochanek
- Department of Critical Care Medicine, Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, 4401 Penn Avenue, Pittsburgh, PA 15224, USA; Department of Pediatrics, University of Pittsburgh School of Medicine, 4401 Penn Avenue, Pittsburgh, PA 15224, USA; Brain Care Institute, Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - Robert S B Clark
- Department of Critical Care Medicine, Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, 4401 Penn Avenue, Pittsburgh, PA 15224, USA; Department of Pediatrics, University of Pittsburgh School of Medicine, 4401 Penn Avenue, Pittsburgh, PA 15224, USA; Brain Care Institute, Children's Hospital of Pittsburgh, 4401 Penn Avenue, Pittsburgh, PA 15224, USA.
| |
Collapse
|