1
|
Zhao A, Zhang G, Wei H, Yan X, Gan J, Jiang X. Heat shock proteins in cerebral ischemia-reperfusion injury: Mechanisms and therapeutic implications. Exp Neurol 2025; 390:115284. [PMID: 40318821 DOI: 10.1016/j.expneurol.2025.115284] [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: 03/03/2025] [Revised: 04/24/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025]
Abstract
Cerebral ischemia-reperfusion injury (CIRI) remains a significant challenge in ischemic stroke treatment. Heat shock proteins (HSPs), a cadre of molecular chaperones, have emerged as pivotal regulators in this pathological cascade. This review synthesizes the latest research on HSPs in CIRI from 2013 to 2024 focusing on their multifaceted roles and therapeutic potential. We explore the diverse cellular functions of HSPs, including regulation of oxidative stress, apoptosis, necroptosis, ferroptosis, autophagy, neuroinflammation, and blood-brain barrier integrity. Key HSPs, such as HSP90, HSP70, HSP32, HSP60, HSP47, and small HSPs, are investigated for their specific mechanisms of action in CIRI. Potential therapeutic strategies targeting HSPs, including HSP inhibitors, traditional Chinese medicine components, and gene therapy, are discussed. This review provides a comprehensive understanding of HSPs in CIRI and offers insights into the development of innovative neuroprotective treatments.
Collapse
Affiliation(s)
- Anliu Zhao
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Guangming Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Huayuan Wei
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xu Yan
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jiali Gan
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
2
|
Reginensi D, Ortiz DA, Denis B, Castillo S, Burillo A, Khoury N, Xu J, Dam ML, Escobar AAH, Dave KR, Perez-Pinzon MA, Gittens RA. Region-specific brain decellularized extracellular matrix promotes cell recovery in an in vitro model of stroke. Sci Rep 2025; 15:11921. [PMID: 40195414 PMCID: PMC11976941 DOI: 10.1038/s41598-025-95656-w] [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: 09/21/2024] [Accepted: 03/24/2025] [Indexed: 04/09/2025] Open
Abstract
Brain decellularized extracellular matrix (ECM) can be an attractive scaffold capable of mimicking the native ecosystem of the central nervous system tissue. We studied the in vitro response of neural cultures exposed to region-specific brain decellularized ECM scaffolds from three distinct neuroanatomical sections: cortex, cerebellum and remaining areas. First, each brain region was evaluated with the isotropic fractionator method to understand the cellular composition of the different cerebral areas. Second, the cerebral regions were subjected to the decellularization process and their respective characterization using molecular, histological, and ultrastructural techniques. Third, the levels of neurotrophic factors in the decellularized brain scaffold were analyzed. Fourth, we studied the region-specific brain decellularized ECM as a mimetic platform for the maturation of PC12 cells, as a unidirectional model of differentiation. Finally, in vitro studies were carried out to evaluate the cell recovery capacity of brain decellularized ECM under stroke-mimetic conditions. Our results show that region-specific brain decellularized ECM can serve as a biomimetic scaffold capable of promoting the growth of neural lineage cells and, in addition, it possesses a combination of structural and biochemical signals (e.g., neurotrophic factors) that are capable of inducing cell phenotypic changes and promote viability and cell recovery in a stroke/ischemia model in vitro.
Collapse
Affiliation(s)
- Diego Reginensi
- Center for Biodiversity and Drug Discovery, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT-AIP), Panama, Panama
- Advanced Therapies, School of Medicine, Universidad de Panamá (UP), Panama, Panama
- Tissue Engineering, Faculty of Biosciences and Public Health, Universidad Especializada de las Américas (UDELAS), Panama, Panama
- Biomedical Engineering, Faculty of Health Sciences and Engineering, Universidad Latina de Panama (ULATINA), Panama, Panama
- Sistema Nacional de Investigación (SNI-SENACYT), Panama, Panama
| | - Didio Alberto Ortiz
- Center for Biodiversity and Drug Discovery, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT-AIP), Panama, Panama
| | - Bernardino Denis
- Center for Biodiversity and Drug Discovery, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT-AIP), Panama, Panama
- Sistema Nacional de Investigación (SNI-SENACYT), Panama, Panama
- MD-PhD Program in Clinical and Biomedical Research, School of Medicine, UP, Panama, Panama
| | - Solangel Castillo
- Advanced Therapies, School of Medicine, Universidad de Panamá (UP), Panama, Panama
- Tissue Engineering, Faculty of Biosciences and Public Health, Universidad Especializada de las Américas (UDELAS), Panama, Panama
| | - Andrea Burillo
- Center for Biodiversity and Drug Discovery, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT-AIP), Panama, Panama
| | - Nathalie Khoury
- Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, USA
- Neurology Department, The Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jing Xu
- Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, USA
- Neurology Department, The Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Maria Lucia Dam
- Center for Biodiversity and Drug Discovery, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT-AIP), Panama, Panama
| | - Anthony A Hurtado Escobar
- Center for Biodiversity and Drug Discovery, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT-AIP), Panama, Panama
- Biomedical Engineering, Faculty of Health Sciences and Engineering, Universidad Latina de Panama (ULATINA), Panama, Panama
| | - Kunjan R Dave
- Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, USA
- Neurology Department, The Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Miguel A Perez-Pinzon
- Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, USA
- Neurology Department, The Peritz Scheinberg Cerebral Vascular Disease Research Laboratories, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Rolando A Gittens
- Center for Biodiversity and Drug Discovery, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT-AIP), Panama, Panama.
- Sistema Nacional de Investigación (SNI-SENACYT), Panama, Panama.
- MD-PhD Program in Clinical and Biomedical Research, School of Medicine, UP, Panama, Panama.
- Instituto Técnico Superior Especializado (ITSE), Ave. Domingo Diaz, Tocumen Panama, Republic of Panama.
- Centro de Investigación e Innovación Educativa, Ciencia y Tecnología (CiiECYT-AIP), Panama, Panama.
| |
Collapse
|
3
|
Gu CL, Zhang L, Zhu Y, Bao TY, Zhu YT, Chen YT, Pang HQ. Exploring the cellular and molecular basis of nerve growth factor in cerebral ischemia recovery. Neuroscience 2025; 566:190-197. [PMID: 39742942 DOI: 10.1016/j.neuroscience.2024.12.049] [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: 11/04/2024] [Revised: 12/19/2024] [Accepted: 12/26/2024] [Indexed: 01/04/2025]
Abstract
Vascular obstruction often causes inadequate oxygen and nutrient supply to the brain. This deficiency results in cerebral ischemic injury, which significantly impairs neurological function. This review aimed to explore the neuroprotective and regenerative effects of nerve growth factor (NGF) in cerebral ischemic injury. NGF, a crucial neurotrophic factor, could inhibit neuronal apoptosis, reduce inflammatory responses, and promote axon regeneration and angiogenesis through its interaction with TrkA, a high-affinity receptor. These functions were closely related to the activation of Phosphatidylinositol 3-kinase/Protein kinase B (PI3K/AKT) and Mitogen-Activated Protein Kinase (MAPK) pathways. Moreover, the mechanisms of NGF in the acute and recovery phases, along with the strategies to enhance its therapeutic effects using delivery systems (such as intranasal administration, nanovesicles, and gene therapy) were also summarized. Although NGF shows great potential for clinical application, its delivery efficiency and long-term safety still need more research and improvements. Future research should focus on exploring the specific action mechanism of NGF, optimizing the delivery strategy, and evaluating its long-term efficacy and safety to facilitate its clinical transformation in cerebral ischemic stroke.
Collapse
Affiliation(s)
- Chen-Lin Gu
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225009, China
| | - Lu Zhang
- The Radiology Department of Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, Taiyuan 030001, China
| | - Yan Zhu
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225009, China
| | - Ting-Yu Bao
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225009, China
| | - Yu-Ting Zhu
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225009, China
| | - Yu-Tong Chen
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225009, China
| | - Han-Qing Pang
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
4
|
Benítez-Marín MJ, Blasco-Alonso M, de Rodríguez de Fonseca F, Jiménez JS, Rivera P, González-Mesa E. Evaluating neuronal damage biomarkers at birth for predicting neurodevelopmental risks in foetal growth restriction. Acta Paediatr 2025; 114:272-284. [PMID: 39601356 DOI: 10.1111/apa.17521] [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: 07/31/2024] [Revised: 10/22/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024]
Abstract
AIM This study was based on the need to predict neurodevelopmental outcomes of children with foetal growth restriction. The aim was to systematically review the correlation between biomarkers of neural injury in children with foetal growth restriction and their neurodevelopment. METHOD Following Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines, the review included studies on growth-restricted foetuses that measured biomarkers of postpartum brain injury and assessed neurodevelopment in childhood. Studies published between 1 January 2014 and 31 March 2024 were identified through PubMed and Embase, with the study protocol registered in PROSPERO (CRD42024520254). RESULTS Only five met the inclusion criteria. Results showed that urinary S100B levels were significantly elevated in foetal growth restriction, negatively correlating with neurological development at 7 days of life. Neuron-specific enolase negatively correlated with cognitive, motor and socio-emotional development. Urinary nerve growth factor levels were significantly lower in neonates with foetal growth restriction, correlating with poor neurodevelopment. No alterations in BDNF levels were observed. Tau protein levels were lower in children with foetal growth restriction and adverse outcomes. CONCLUSION The study emphasised the need for further research on biomarkers and predictive models of neurodevelopment in children with foetal growth restriction.
Collapse
Affiliation(s)
- Mª José Benítez-Marín
- Research Group in Maternal-Fetal Medicine, Epigenetics, Women's Diseases and Reproductive Health, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma Bionand, Málaga, Spain
- Obstetrics and Gynecology Service, Regional University Hospital of Malaga, Málaga, Spain
- Obstetrics and Gynecology Service, Virgen de la Victoria University Hospital, Málaga, Spain
| | - Marta Blasco-Alonso
- Research Group in Maternal-Fetal Medicine, Epigenetics, Women's Diseases and Reproductive Health, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma Bionand, Málaga, Spain
- Obstetrics and Gynecology Service, Regional University Hospital of Malaga, Málaga, Spain
- Surgical Specialties, Biochemistry and Immunology Department, Málaga University, Málaga, Spain
| | - Fernando de Rodríguez de Fonseca
- Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, Málaga, Spain
- Servicio Neurologia, Hospital Regional Universitario de Málaga, Málaga, Spain
| | - Jesús S Jiménez
- Research Group in Maternal-Fetal Medicine, Epigenetics, Women's Diseases and Reproductive Health, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma Bionand, Málaga, Spain
- Obstetrics and Gynecology Service, Regional University Hospital of Malaga, Málaga, Spain
- Surgical Specialties, Biochemistry and Immunology Department, Málaga University, Málaga, Spain
| | - Patricia Rivera
- Instituto de Investigación Biomédica de Málaga (IBIMA)-Plataforma BIONAND, Málaga, Spain
- UGC Salud Mental, Hospital Regional Universitario de Málaga, Málaga, Spain
| | - Ernesto González-Mesa
- Research Group in Maternal-Fetal Medicine, Epigenetics, Women's Diseases and Reproductive Health, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma Bionand, Málaga, Spain
- Obstetrics and Gynecology Service, Regional University Hospital of Malaga, Málaga, Spain
- Surgical Specialties, Biochemistry and Immunology Department, Málaga University, Málaga, Spain
| |
Collapse
|
5
|
Yang Y, Hong Y, Han J, Yang Z, Huang N, Xu B, Ma Z, Wang Q. Nerve growth factor alleviates arsenic-induced testicular injury by enhancing the function of Sertoli cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 280:116578. [PMID: 38861803 DOI: 10.1016/j.ecoenv.2024.116578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 06/01/2024] [Accepted: 06/08/2024] [Indexed: 06/13/2024]
Abstract
Sertoli cells (SCs) maintain testicular homeostasis and promote spermatogenesis by forming the blood-testis barrier (BTB) and secreting growth factors. The pro-proliferative and anti-apoptotic effects of nerve growth factor (NGF) on SCs have been proved previously. It is still unclear whether the damage effect of arsenic on testis is related to the inhibition of NGF expression, and whether NGF can mitigate arsenic-induced testicular damage by decreasing the damage of SCs induced by arsenic. Here, the lower expression of NGF in testes of arsenic exposed mice (freely drinking water containing 15 mg/l of NaAsO2) was observed through detection of Western blot and Real-time PCR. Subsequently, hematoxylin and eosin (HE) staining, Evans blue staining and transmission electron microscopy were used to evaluate the pathology, BTB permeability and tight junction integrity in testes of control mice, arsenic exposed mice (freely drinking water containing 15 mg/l of NaAsO2) and arsenic + NGF treated mice (freely drinking water containing 15 mg/l of NaAsO2 + intraperitoneal injection with 30 μg/kg of NGF), respectively. Evidently, spermatogenic tubule epithelial cells in testis of arsenic exposed mice were disordered and the number of cell layers was reduced, accompanied by increased permeability and damaged integrity of the tight junction in BTB, but these changes were less obvious in testes of mice treated with arsenic + NGF. In addition, the sperm count, motility and malformation rate of mice treated with arsenic + NGF were also improved. On the basis of the above experiments, the viability and apoptosis of primary cultured SCs treated with arsenic (10 μM NaAsO2) or arsenic + NGF (10 μM NaAsO2 + 100 ng/mL NGF) were detected by Cell counting kit-8 (CCK8) and transferase-mediated DUTP-biotin nick end labeling (TUNEL) staining, respectively. It is found that NGF ameliorated the decline of growth activity and the increase of apoptosis in arsenic-induced SCs. This remarkable biological effect that NGF inhibited the increase of Bax expression and the decrease of Bcl-2 expression in arsenic-induced SCs was also determined by western blot and Real-time PCR. Moreover, the decrease in transmembrane resistance (TEER) and the expression of tight junction proteins ZO-1 and occludin was mitigated in SCs induced by arsenic due to NGF treatment. In conclusion, the above results confirmed that NGF could ameliorate the injury effects of arsenic on testis, which might be related to the function of NGF to inhibit arsenic-induced SCs injury.
Collapse
Affiliation(s)
- Yanping Yang
- Department of Histology and Embryology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou 561113, PR China
| | - Yan Hong
- Department of Histology and Embryology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou 561113, PR China
| | - Jing Han
- Department of Histology and Embryology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou 561113, PR China
| | - Zhe Yang
- Department of Histology and Embryology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou 561113, PR China
| | - Nanmin Huang
- Department of Histology and Embryology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou 561113, PR China
| | - Binwei Xu
- Department of Histology and Embryology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou 561113, PR China
| | - Zhaolei Ma
- Department of Geriatrics, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, PR China.
| | - Qi Wang
- Department of Histology and Embryology, School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou 561113, PR China.
| |
Collapse
|
6
|
Gong J, Ding G, Hao Z, Li Y, Deng A, Zhang C. Elucidating the mechanism of corneal epithelial cell repair: unraveling the impact of growth factors. Front Med (Lausanne) 2024; 11:1384500. [PMID: 38638937 PMCID: PMC11024251 DOI: 10.3389/fmed.2024.1384500] [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: 02/09/2024] [Accepted: 03/21/2024] [Indexed: 04/20/2024] Open
Abstract
The repair mechanism for corneal epithelial cell injuries encompasses migration, proliferation, and differentiation of corneal epithelial cells, and extracellular matrix remodeling of the stromal structural integrity. Furthermore, it involves the consequential impact of corneal limbal stem cells (LSCs). In recent years, as our comprehension of the mediating mechanisms underlying corneal epithelial injury repair has advanced, it has become increasingly apparent that growth factors play a pivotal role in this intricate process. These growth factors actively contribute to the restoration of corneal epithelial injuries by orchestrating responses and facilitating specific interactions at targeted sites. This article systematically summarizes the role of growth factors in corneal epithelial cell injury repair by searching relevant literature in recent years, and explores the limitations of current literature search, providing a certain scientific basis for subsequent basic research and clinical applications.
Collapse
Affiliation(s)
- Jinjin Gong
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
- Department of Ophthalmology, Jinan Second People’s Hospital, Jinan, China
| | - Gang Ding
- Department of Ophthalmology, Jinan Second People’s Hospital, Jinan, China
| | - Zhongkai Hao
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
- Department of Ophthalmology, Jinan Second People’s Hospital, Jinan, China
| | - Yuchun Li
- Wuxi No. 2 Chinese Medicine Hospital, Wuxi, China
| | - Aijun Deng
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Chenming Zhang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
- Department of Ophthalmology, Jinan Second People’s Hospital, Jinan, China
| |
Collapse
|
7
|
Chen X, Zhang L, Chai W, Tian P, Kim J, Ding J, Zhang H, Liu C, Wang D, Cui X, Pan H. Hypoxic Microenvironment Reconstruction with Synergistic Biofunctional Ions Promotes Diabetic Wound Healing. Adv Healthc Mater 2023; 12:e2301984. [PMID: 37740829 DOI: 10.1002/adhm.202301984] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/14/2023] [Indexed: 09/25/2023]
Abstract
Chronic hypoxia and ischemia make diabetic wounds non-healing. Cellular functions of diabetic chronic wounds are inhibited under a pathological environment. Therefore, this work develops a composite hydrogel system to promote diabetic wound healing. The composite hydrogel system consists of ε-poly-lysine (EPL), calcium peroxide (CP), and borosilicate glass (BG). The hydrogel supplies continuous dissolved oxygen molecules to the wound that can penetrate the skin tissue to restore normal cellular function and promote vascular regeneration. Biofunctional ions released from BGs can recruit more macrophages through neovascularization and modulate macrophage phenotypic transformation. Combining oxygen-mediated vascular regeneration and ion-mediated inflammatory regulation significantly accelerated diabetic wound healing. These findings indicate that this composite hydrogel system holds promise as a novel tissue engineering material.
Collapse
Affiliation(s)
- Xiaochen Chen
- School of materials science and engineering, Tongji University, Shanghai, 201804, P. R. China
| | - Liyan Zhang
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Wenwen Chai
- School of materials science and engineering, Tongji University, Shanghai, 201804, P. R. China
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Pengfei Tian
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Jua Kim
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Jingxin Ding
- School of materials science and engineering, Tongji University, Shanghai, 201804, P. R. China
| | - Hao Zhang
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Chunyu Liu
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Deping Wang
- School of materials science and engineering, Tongji University, Shanghai, 201804, P. R. China
| | - Xu Cui
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Haobo Pan
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
- Shenzhen Healthemes Biotechnology Co., Ltd, Shenzhen, 518071, P. R. China
| |
Collapse
|
8
|
Challa NVD, Chen S, Yuan H, Duncan MR, Moreno WJ, Bramlett H, Dietrich WD, Benny M, Schmidt AF, Young K, Wu S. GSDMD gene knockout alleviates hyperoxia-induced hippocampal brain injury in neonatal mice. J Neuroinflammation 2023; 20:205. [PMID: 37679766 PMCID: PMC10486051 DOI: 10.1186/s12974-023-02878-8] [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: 06/12/2023] [Accepted: 08/19/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND Neonatal hyperoxia exposure is associated with brain injury and poor neurodevelopment outcomes in preterm infants. Our previous studies in neonatal rodent models have shown that hyperoxia stimulates the brain's inflammasome pathway, leading to the activation of gasdermin D (GSDMD), a key executor of pyroptotic inflammatory cell death. Moreover, we found pharmacological inhibition of caspase-1, which blocks GSDMD activation, attenuates hyperoxia-induced brain injury in neonatal mice. We hypothesized that GSDMD plays a pathogenic role in hyperoxia-induced neonatal brain injury and that GSDMD gene knockout (KO) will alleviate hyperoxia-induced brain injury. METHODS Newborn GSDMD knockout mice and their wildtype (WT) littermates were randomized within 24 h after birth to be exposed to room air or hyperoxia (85% O2) from postnatal days 1 to 14. Hippocampal brain inflammatory injury was assessed in brain sections by immunohistology for allograft inflammatory factor 1 (AIF1) and CD68, markers of microglial activation. Cell proliferation was evaluated by Ki-67 staining, and cell death was determined by TUNEL assay. RNA sequencing of the hippocampus was performed to identify the transcriptional effects of hyperoxia and GSDMD-KO, and qRT-PCR was performed to confirm some of the significantly regulated genes. RESULTS Hyperoxia-exposed WT mice had increased microglia consistent with activation, which was associated with decreased cell proliferation and increased cell death in the hippocampal area. Conversely, hyperoxia-exposed GSDMD-KO mice exhibited considerable resistance to hyperoxia as O2 exposure did not increase AIF1 + , CD68 + , or TUNEL + cell numbers or decrease cell proliferation. Hyperoxia exposure differentially regulated 258 genes in WT and only 16 in GSDMD-KO mice compared to room air-exposed WT and GSDMD-KO, respectively. Gene set enrichment analysis showed that in the WT brain, hyperoxia differentially regulated genes associated with neuronal and vascular development and differentiation, axonogenesis, glial cell differentiation, hypoxia-induced factor 1 pathway, and neuronal growth factor pathways. These changes were prevented by GSDMD-KO. CONCLUSIONS GSDMD-KO alleviates hyperoxia-induced inflammatory injury, cell survival and death, and alterations of transcriptional gene expression of pathways involved in neuronal growth, development, and differentiation in the hippocampus of neonatal mice. This suggests that GSDMD plays a pathogenic role in preterm brain injury, and targeting GSDMD may be beneficial in preventing and treating brain injury and poor neurodevelopmental outcomes in preterm infants.
Collapse
Affiliation(s)
- Naga Venkata Divya Challa
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Holtz Children's Hospital, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Shaoyi Chen
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Holtz Children's Hospital, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Huijun Yuan
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Holtz Children's Hospital, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Matthew R Duncan
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Holtz Children's Hospital, University of Miami Miller School of Medicine, Miami, FL, USA
| | - William Javier Moreno
- Miami Project to Cure Paralysis and Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Helen Bramlett
- Miami Project to Cure Paralysis and Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - W Dalton Dietrich
- Miami Project to Cure Paralysis and Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Merline Benny
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Holtz Children's Hospital, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Augusto F Schmidt
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Holtz Children's Hospital, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Karen Young
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Holtz Children's Hospital, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Shu Wu
- Department of Pediatrics/Division of Neonatology, Batchelor Children's Research Institute, Holtz Children's Hospital, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
9
|
Hu HL, Khatri L, Santacruz M, Church E, Moore C, Huang TT, Chao MV. Confronting the loss of trophic support. Front Mol Neurosci 2023; 16:1179209. [PMID: 37456526 PMCID: PMC10338843 DOI: 10.3389/fnmol.2023.1179209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/23/2023] [Indexed: 07/18/2023] Open
Abstract
Classic experiments with peripheral sympathetic neurons established an absolute dependence upon NGF for survival. A forgotten problem is how these neurons become resistant to deprivation of trophic factors. The question is whether and how neurons can survive in the absence of trophic support. However, the mechanism is not understood how neurons switch their phenotype to lose their dependence on trophic factors, such as NGF and BDNF. Here, we approach the problem by considering the requirements for trophic support of peripheral sympathetic neurons and hippocampal neurons from the central nervous system. We developed cellular assays to assess trophic factor dependency for sympathetic and hippocampal neurons and identified factors that rescue neurons in the absence of trophic support. They include enhanced expression of a subunit of the NGF receptor (Neurotrophin Receptor Homolog, NRH) in sympathetic neurons and an increase of the expression of the glucocorticoid receptor in hippocampal neurons. The results are significant since levels and activity of trophic factors are responsible for many neuropsychiatric conditions. Resistance of neurons to trophic factor deprivation may be relevant to the underlying basis of longevity, as well as an important element in preventing neurodegeneration.
Collapse
Affiliation(s)
- Hui-Lan Hu
- Department of Biochemistry and Molecular Pharmacology, New York University Langone School of Medicine, New York, NY, United States
| | - Latika Khatri
- Skirball Institute for Biomolecular Medicine, Neuroscience Institute, New York University Langone Medical Center, New York, NY, United States
| | - Marilyn Santacruz
- Department of Neuroscience, Pomona College, Claremont, CA, United States
| | - Emily Church
- Department of Neuroscience, Pomona College, Claremont, CA, United States
| | - Christopher Moore
- Skirball Institute for Biomolecular Medicine, Neuroscience Institute, New York University Langone Medical Center, New York, NY, United States
| | - Tony T. Huang
- Department of Biochemistry and Molecular Pharmacology, New York University Langone School of Medicine, New York, NY, United States
| | - Moses V. Chao
- Skirball Institute for Biomolecular Medicine, Neuroscience Institute, New York University Langone Medical Center, New York, NY, United States
- Department of Cell Biology, New York Langone Medical Center, New York, NY, United States
- Department of Psychiatry, New York Langone Medical Center, New York, NY, United States
- Department of Neuroscience and Physiology, New York Langone Medical Center, New York, NY, United States
| |
Collapse
|
10
|
Palasz E, Wilkaniec A, Stanaszek L, Andrzejewska A, Adamczyk A. Glia-Neurotrophic Factor Relationships: Possible Role in Pathobiology of Neuroinflammation-Related Brain Disorders. Int J Mol Sci 2023; 24:ijms24076321. [PMID: 37047292 PMCID: PMC10094105 DOI: 10.3390/ijms24076321] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/24/2023] [Accepted: 03/27/2023] [Indexed: 03/30/2023] Open
Abstract
Neurotrophic factors (NTFs) play an important role in maintaining homeostasis of the central nervous system (CNS) by regulating the survival, differentiation, maturation, and development of neurons and by participating in the regeneration of damaged tissues. Disturbances in the level and functioning of NTFs can lead to many diseases of the nervous system, including degenerative diseases, mental diseases, and neurodevelopmental disorders. Each CNS disease is characterized by a unique pathomechanism, however, the involvement of certain processes in its etiology is common, such as neuroinflammation, dysregulation of NTFs levels, or mitochondrial dysfunction. It has been shown that NTFs can control the activation of glial cells by directing them toward a neuroprotective and anti-inflammatory phenotype and activating signaling pathways responsible for neuronal survival. In this review, our goal is to outline the current state of knowledge about the processes affected by NTFs, the crosstalk between NTFs, mitochondria, and the nervous and immune systems, leading to the inhibition of neuroinflammation and oxidative stress, and thus the inhibition of the development and progression of CNS disorders.
Collapse
Affiliation(s)
- Ewelina Palasz
- Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
- Correspondence: (E.P.); (A.A.)
| | - Anna Wilkaniec
- Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Luiza Stanaszek
- Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Anna Andrzejewska
- Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
- Center for Advanced Imaging Research, Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Agata Adamczyk
- Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland
- Correspondence: (E.P.); (A.A.)
| |
Collapse
|
11
|
Perrone S, Grassi F, Caporilli C, Boscarino G, Carbone G, Petrolini C, Gambini LM, Di Peri A, Moretti S, Buonocore G, Esposito SMR. Brain Damage in Preterm and Full-Term Neonates: Serum Biomarkers for the Early Diagnosis and Intervention. Antioxidants (Basel) 2023; 12:antiox12020309. [PMID: 36829868 PMCID: PMC9952571 DOI: 10.3390/antiox12020309] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 01/14/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023] Open
Abstract
The Brain is vulnerable to numerous insults that can act in the pre-, peri-, and post-natal period. There is growing evidence that demonstrate how oxidative stress (OS) could represent the final common pathway of all these insults. Fetuses and newborns are particularly vulnerable to OS due to their inability to active the antioxidant defenses. Specific molecules involved in OS could be measured in biologic fluids as early biomarkers of neonatal brain injury with an essential role in neuroprotection. Although S-100B seems to be the most studied biomarker, its use in clinical practice is limited by the complexity of brain damage etiopathogenesis and the time of blood sampling in relation to the brain injury. Reliable early specific serum markers are currently lacking in clinical practice. It is essential to determine if there are specific biomarkers that can help caregivers to monitor the progression of the disease in order to active an early neuroprotective strategy. We aimed to describe, in an educational review, the actual evidence on serum biomarkers for the early identification of newborns at a high risk of neurological diseases. To move the biomarkers from the bench to the bedside, the assays must be not only be of a high sensitivity but suitable for the very rapid processing and return of the results for the clinical practice to act on. For the best prognosis, more studies should focus on the association of these biomarkers to the type and severity of perinatal brain damage.
Collapse
Affiliation(s)
- Serafina Perrone
- Neonatology Unit, Pietro Barilla Children’s Hospital, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- Correspondence:
| | - Federica Grassi
- Pediatric Clinic, Pietro Barilla Children’s Hospital, University of Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Chiara Caporilli
- Pediatric Clinic, Pietro Barilla Children’s Hospital, University of Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Giovanni Boscarino
- Pediatric Clinic, Pietro Barilla Children’s Hospital, University of Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Giulia Carbone
- Pediatric Clinic, Pietro Barilla Children’s Hospital, University of Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Chiara Petrolini
- Neonatology Unit, Pietro Barilla Children’s Hospital, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Lucia Maria Gambini
- Neonatology Unit, Pietro Barilla Children’s Hospital, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Antonio Di Peri
- Neonatology Unit, Pietro Barilla Children’s Hospital, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Sabrina Moretti
- Neonatology Unit, Pietro Barilla Children’s Hospital, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Giuseppe Buonocore
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy
| | | |
Collapse
|
12
|
The p75 neurotrophin receptor inhibitor, LM11A-31, ameliorates acute stroke injury and modulates astrocytic proNGF. Exp Neurol 2023; 359:114161. [PMID: 35787888 DOI: 10.1016/j.expneurol.2022.114161] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 06/14/2022] [Accepted: 06/28/2022] [Indexed: 12/30/2022]
Abstract
The precursor form of nerve growth factor (proNGF) is essential to maintain NGF survival signaling. ProNGF is also among endogenous ligands for p75 neurotrophin receptor (p75ntr). Mounting evidence implies that p75ntr signaling contributes to neural damage in ischemic stroke. The present study examines the therapeutic effect of the p75ntr modulator LM11A-31. Adult mice underwent transient distal middle cerebral artery occlusion (t-dMCAO) followed by LM11A-31 treatment (25 mg/kg, i.p., twice daily) either for 72 h post-injury (acute phase) or afterward till two weeks post-stroke (subacute phase). LM11A-31 reduced blood-brain barrier permeability, cerebral tissue injury, and sensorimotor function in the acute phase of stroke. Ischemic brain samples showed repressed proNGF/P75ntr signaling and Caspase 3 activation in LM11A-31 treated mice, where we observed less reactive microglia and IL-1β production. LM11A-31 (20-80 nM) also mitigated neural injury induced by oxygen-glucose deprivation (OGD) in sandwich co-cultures of primary cortical neurons (PCN) and astrocytes. This concurred with JNK/PARP downregulation and reduced caspase-3 cleavage in the PCNs and was associated with repressed proNGF generation in astrocytes. Further in vitro experiments indicated human proNGF suppresses the pro-inflammatory phenotype in microglial cultures, as determined by a sharp decline in HMGB-1 production and moderate arginase-1 upregulation. Despite significant protection in acute stroke, LM11A-31 treatment did not improve cortical atrophy and sensorimotor function in the subacute phase. Our findings provide preclinical evidence supporting LM11A-31 as a promising therapy for acute stroke injury. Further investigations may elucidate if reduced astrocytic proNGF, an endogenous reservoir of pro-neurotrophins, may restrict the therapeutic window.
Collapse
|
13
|
You S, Wang Y, Guo Y, Guo C, Cao F, Shi W, Yang L, Mi W, Tong L. Activation of the ERK1/2 pathway mediates the neuroprotective effect provided by calycosin treatment. Neurosci Lett 2023; 792:136956. [PMID: 36347338 DOI: 10.1016/j.neulet.2022.136956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 10/25/2022] [Accepted: 11/01/2022] [Indexed: 11/08/2022]
Abstract
Calycosin is a natural product extracted from some plant families and exhibits various biological properties. But the effect of calycosin on cerebral ischemia-reperfusion injury has not been fully elucidated. In this study, the neuroprotective effect of calycosin treatment on the differentiated SH-SY5Y cells exposed to OGD was evaluated using MTT and flow cytometry. Rats that were pretreatment with calycosin were subjected to MCAO, neurological behavior scores and brain infarct volume were evaluated. The protein expression of pERK/ERK were assessed using Western blot. siRNA-pERK and U0126 were administered to investigate the impact of the ERK pathway on calycosin preconditioning. The results demonstrated the neuronal viability in the calycosin-treated SH-SY5Y cells increased significantly, and the rate of apoptosis decreased compared with the Oxygen-glucose deprivation only SH-SY5Y cells. Calycosin pretreatment reduced infarct volume and improved neurological outcome in rats subjected to MCAO. Administration of calycosin increased the ratio of pERK/ERK expression, which was down-regulated in ischemia-reperfusion group. Down-regulation of pERK/ERK significantly attenuated the neuroprotective effect induced by calycosin pretreatment in vitro and in vivo. We concluded calycosin treatment could induce a neuroprotective effect against ischemia, which was related to the regulation of the ERK1/2 pathway.
Collapse
Affiliation(s)
- Shaohua You
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China; Department of Pain Medicine, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Yanfeng Wang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Yongxin Guo
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Chao Guo
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, China
| | - Fuyang Cao
- Department of Anesthesiology, The Sixth Medical Center of Chinese PLA General Hospital, Beijing 100048, China
| | - Wenzhu Shi
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Lujia Yang
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Weidong Mi
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China.
| | - Li Tong
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
14
|
Song QX, Sun Y, Deng K, Mei JY, Chermansky CJ, Damaser MS. Potential role of oxidative stress in the pathogenesis of diabetic bladder dysfunction. Nat Rev Urol 2022; 19:581-596. [PMID: 35974244 DOI: 10.1038/s41585-022-00621-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/22/2022] [Indexed: 11/09/2022]
Abstract
Diabetes mellitus is a chronic metabolic disease, posing a considerable threat to global public health. Treating systemic comorbidities has been one of the greatest clinical challenges in the management of diabetes. Diabetic bladder dysfunction, characterized by detrusor overactivity during the early stage of the disease and detrusor underactivity during the late stage, is a common urological complication of diabetes. Oxidative stress is thought to trigger hyperglycaemia-dependent tissue damage in multiple organs; thus, a growing body of literature has suggested a possible link between functional changes in urothelium, muscle and the corresponding innervations. Improved understanding of the mechanisms of oxidative stress could lead to the development of novel therapeutics to restore the redox equilibrium and scavenge excessive free radicals to normalize bladder function in patients with diabetes.
Collapse
Affiliation(s)
- Qi-Xiang Song
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Sun
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Kangli Deng
- Department of Urology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jin-Yi Mei
- Department of Urology, Changhai Hospital, Naval Medical University, Shanghai, China
| | | | - Margot S Damaser
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA. .,Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA. .,Glickman Urology and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
15
|
Yang Q, Wang W. The Nuclear Translocation of Heme Oxygenase-1 in Human Diseases. Front Cell Dev Biol 2022; 10:890186. [PMID: 35846361 PMCID: PMC9277552 DOI: 10.3389/fcell.2022.890186] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 06/10/2022] [Indexed: 12/30/2022] Open
Abstract
Heme oxygenase-1 (HO-1) is a rate-limiting enzyme in the degradation of heme to generate carbon monoxide (CO), free iron and biliverdin, which could then be converted to bilirubin by biliverdin reductase. HO-1 exhibits cytoprotective effects of anti-apoptosis, anti-oxidation, and anti-inflammation via these byproducts generated during the above process. In the last few years, despite the canonical function of HO-1 and possible biological significance of its byproducts, a noncanonical function, through which HO-1 exhibits functions in diseases independent of its enzyme activity, also has been reported. In this review, the noncanonical functions of HO-1 and its translocation in other subcellular compartments are summarized. More importantly, we emphasize the critical role of HO-1 nuclear translocation in human diseases. Intriguingly, this translocation was linked to tumorigenesis and tumor progression in lung, prostate, head, and neck squamous cell carcinomas and chronic myeloid leukemia. Given the importance of HO-1 nuclear translocation in human diseases, nuclear HO-1 as a novel target might be attractive for the prevention and treatment of human diseases.
Collapse
Affiliation(s)
- Qing Yang
- Department of Breast Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wenqian Wang
- Department of Plastic Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- *Correspondence: Wenqian Wang,
| |
Collapse
|
16
|
Yin YL, Liu YH, Zhu ML, Wang HH, Qiu Y, Wan GR, Li P. Floralozone improves cognitive impairment in vascular dementia rats via regulation of TRPM2 and NMDAR signaling pathway. Physiol Behav 2022; 249:113777. [PMID: 35276121 DOI: 10.1016/j.physbeh.2022.113777] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 02/23/2022] [Accepted: 03/07/2022] [Indexed: 12/11/2022]
Abstract
Vascular dementia (VD) is the second largest type of dementia after Alzheimer's disease. At present, the pathogenesis is complex and there is no effective treatment. Floralozone has been shown to reduce atherosclerosis in rats caused by a high-fat diet. However, whether it plays a role in VD remains elusive. In the present study, the protective activities and relevant mechanisms of Floralozone were evaluated in rats with cognitive impairment, which were induced by bilateral occlusion of the common carotid arteries (BCCAO) in rats. Cognitive function, pathological changes and oxidative stress condition in the brains of VD rats were assessed using Neurobehavioral tests, Morris water maze tests, hematoxylin-eosin staining, Neu N staining, TUNEL staining, Golgi staining, Western blot assay and antioxidant assays (MDA, SOD, GSH), respectively. The results indicated that VD model was established successfully and BCCAO caused a decline in spatial learning and memory and hippocampal histopathological abnormalities of rats. Floralozone (50, 100, 150 mg/kg) dose-dependently alleviated the pathological changes, decreased oxidative stress injury, which eventually reduced cognitive impairment in BCCAO rats. The same results were shown in further experiments with neurobehavioral tests. At the molecular biological level, Floralozone decreased the protein level of transient receptor potential melastatin-related 2 (TRPM2) in VD and normal rats, and increased the protein level of NR2B in hippocampus of N-methyl-D-aspartate receptor (NMDAR). Notably, Floralozone could markedly improved learning and memory function of BCCAO rats in Morris water maze (MWM) and improved neuronal cell loss, synaptic structural plasticity. In conclusion, Floralozone has therapeutic potential for VD, increased synaptic structural plasticity and alleviating neuronal cell apoptosis, which may be related to the TRPM2/NMDAR pathway.
Collapse
Affiliation(s)
- Ya-Ling Yin
- School of Basic Medical Sciences, Department of Physiology and Pathophysiology, Sino-UK Joint Laboratory of Brain Function and Injury and Department of Physiology and Neurobiology, Xinxiang Medical University,Xinxiang, China, 453003; College of Pharmacy, Henan international joint laboratory of cardiovascular remodeling and drug intervention, Xinxiang key laboratory of vascular remodeling intervention and molecular targeted therapy drug development, Xinxiang Medical University,Xinxiang, China, 453003.
| | - Yan-Hua Liu
- College of Pharmacy, Henan international joint laboratory of cardiovascular remodeling and drug intervention, Xinxiang key laboratory of vascular remodeling intervention and molecular targeted therapy drug development, Xinxiang Medical University,Xinxiang, China, 453003.
| | - Mo-Li Zhu
- College of Pharmacy, Henan international joint laboratory of cardiovascular remodeling and drug intervention, Xinxiang key laboratory of vascular remodeling intervention and molecular targeted therapy drug development, Xinxiang Medical University,Xinxiang, China, 453003.
| | - Huan-Huan Wang
- College of Pharmacy, Henan international joint laboratory of cardiovascular remodeling and drug intervention, Xinxiang key laboratory of vascular remodeling intervention and molecular targeted therapy drug development, Xinxiang Medical University,Xinxiang, China, 453003.
| | - Yue Qiu
- College of Pharmacy, Henan international joint laboratory of cardiovascular remodeling and drug intervention, Xinxiang key laboratory of vascular remodeling intervention and molecular targeted therapy drug development, Xinxiang Medical University,Xinxiang, China, 453003.
| | - Guang-Rui Wan
- College of Pharmacy, Henan international joint laboratory of cardiovascular remodeling and drug intervention, Xinxiang key laboratory of vascular remodeling intervention and molecular targeted therapy drug development, Xinxiang Medical University,Xinxiang, China, 453003.
| | - Peng Li
- School of Basic Medical Sciences, Department of Physiology and Pathophysiology, Sino-UK Joint Laboratory of Brain Function and Injury and Department of Physiology and Neurobiology, Xinxiang Medical University,Xinxiang, China, 453003; College of Pharmacy, Henan international joint laboratory of cardiovascular remodeling and drug intervention, Xinxiang key laboratory of vascular remodeling intervention and molecular targeted therapy drug development, Xinxiang Medical University,Xinxiang, China, 453003.
| |
Collapse
|
17
|
|
18
|
Zhu T, Zhu M, Qiu Y, Wu Z, Huang N, Wan G, Xu J, Song P, Wang S, Yin Y, Li P. Puerarin Alleviates Vascular Cognitive Impairment in Vascular Dementia Rats. Front Behav Neurosci 2021; 15:717008. [PMID: 34720898 PMCID: PMC8554240 DOI: 10.3389/fnbeh.2021.717008] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 09/21/2021] [Indexed: 11/13/2022] Open
Abstract
Cerebral ischemia triggers vascular dementia (VD), which is characterized by memory loss, cognitive deficits, and vascular injury in the brain. Puerarin (Pur) represents the major isoflavone glycoside of Radix Puerariae, with verified neuroprotective activity and cardiovascular protective effects. However, whether Pur ameliorates cognitive impairment and vascular injury in rats with permanent occlusion of bilateral common carotid arteries (BCCAO) remains unknown. This work aimed to assess Pur's effects on BCCAO-induced VD and to dissect the underlying mechanisms, especially examining the function of transient receptor potential melastatin-related 2 (TRPM2) in alleviating cognitive deficits and vascular injuries. Rats with BCCAO developed VD. Pur (50, 100, and 150 mg/kg) dose-dependently attenuated the pathological changes, increased synaptic structural plasticity in the dorsal CA1 hippocampal region and decreased oxidative stress, which eventually reduced cognitive impairment and vascular injury in BCCAO rats. Notably, Pur-improved neuronal cell loss, synaptic structural plasticity, and endothelial vasorelaxation function might be mediated by the reactive oxygen species (ROS)-dependent TRPM2/NMDAR pathway, evidenced by decreased levels of ROS, malondialdehyde (MDA), Bax, Bax/Bcl2, and TRPM2, and increased levels of superoxide dismutase (SOD), Bcl2, and NR2A. In conclusion, Pur has therapeutic potential for VD, alleviating neuronal cell apoptosis and vascular injury, which may be related to the ROS-dependent TRPM2/NMDAR pathway.
Collapse
Affiliation(s)
- Tiantian Zhu
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China.,Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Moli Zhu
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China.,Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Yue Qiu
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China.,Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Zeqing Wu
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China.,Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Ning Huang
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China.,Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Guangrui Wan
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China.,Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Jian Xu
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China.,Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Ping Song
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China.,Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Shuangxi Wang
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China.,Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Yaling Yin
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.,School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Peng Li
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China.,Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.,Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| |
Collapse
|
19
|
Colardo M, Martella N, Pensabene D, Siteni S, Di Bartolomeo S, Pallottini V, Segatto M. Neurotrophins as Key Regulators of Cell Metabolism: Implications for Cholesterol Homeostasis. Int J Mol Sci 2021; 22:5692. [PMID: 34073639 PMCID: PMC8198482 DOI: 10.3390/ijms22115692] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 12/14/2022] Open
Abstract
Neurotrophins constitute a family of growth factors initially characterized as predominant mediators of nervous system development, neuronal survival, regeneration and plasticity. Their biological activity is promoted by the binding of two different types of receptors, leading to the generation of multiple and variegated signaling cascades in the target cells. Increasing evidence indicates that neurotrophins are also emerging as crucial regulators of metabolic processes in both neuronal and non-neuronal cells. In this context, it has been reported that neurotrophins affect redox balance, autophagy, glucose homeostasis and energy expenditure. Additionally, the trophic support provided by these secreted factors may involve the regulation of cholesterol metabolism. In this review, we examine the neurotrophins' signaling pathways and their effects on metabolism by critically discussing the most up-to-date information. In particular, we gather experimental evidence demonstrating the impact of these growth factors on cholesterol metabolism.
Collapse
Affiliation(s)
- Mayra Colardo
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, 86090 Pesche, Italy; (M.C.); (N.M.); (D.P.); (S.D.B.)
| | - Noemi Martella
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, 86090 Pesche, Italy; (M.C.); (N.M.); (D.P.); (S.D.B.)
| | - Daniele Pensabene
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, 86090 Pesche, Italy; (M.C.); (N.M.); (D.P.); (S.D.B.)
| | - Silvia Siteni
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Sabrina Di Bartolomeo
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, 86090 Pesche, Italy; (M.C.); (N.M.); (D.P.); (S.D.B.)
| | - Valentina Pallottini
- Department of Science, University Roma Tre, Viale Marconi 446, 00146 Rome, Italy;
- Neuroendocrinology Metabolism and Neuropharmacology Unit, IRCSS Fondazione Santa Lucia, Via del Fosso Fiorano 64, 00143 Rome, Italy
| | - Marco Segatto
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, 86090 Pesche, Italy; (M.C.); (N.M.); (D.P.); (S.D.B.)
| |
Collapse
|
20
|
Increased Retinal Ganglion Cell Survival by Exogenous IL-2 Depends on IL-10, Dopamine D1 Receptors, and Classical IL-2/IL-2R Signaling Pathways. Neurochem Res 2021; 46:1701-1716. [PMID: 33792824 DOI: 10.1007/s11064-021-03313-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 03/01/2021] [Accepted: 03/24/2021] [Indexed: 10/21/2022]
Abstract
Interleukin-2 (IL-2) is a classical pro-inflammatory cytokine known to display neuroprotective roles in the central nervous system including the retina. In the present study, we investigate the molecular targets involved in the neurotrophic effect of IL-2 on retinal ganglion cells (RGC) after optic nerve axotomy. Analysis of retrograde labeling of RGC showed that common cell survival mediators, as Trk receptors, Src, PI3K, PKC, and intracellular calcium do not mediate the neurotrophic effect of IL-2 on RGC. No involvement of MAPK p38 was also observed. However, other MAPKs as MEK and JNK appear to be mediating this IL-2 effect. Our data also indicate that JAK2/3 are important intracellular proteins for the IL-2 effect. Interestingly, we demonstrate that the IL-2 effect depends on dopamine D1 receptors (D1R), the cAMP/PKA pathway, interleukin-10 (IL-10), and NF-κB, suggesting that RGC survival induced by IL-2 encompasses a molecular network of major complexity. In addition, treatment of retinal cells with recombinant IL-10 or 6-Cl-pb (D1R full agonist) was able to increase RGC survival similar to IL-2. Taken together, our results suggest that after optic nerve axotomy, the increase in RGC survival triggered by IL-2 is mediated by IL-10 and D1R along with the intracellular pathways of MAPKs, JAK/STAT, and cAMP/PKA.
Collapse
|
21
|
Xue J, Liu Y, Zhang S, Ding L, Shen B, Shao Y, Wei Z. Caffeine improves bladder function in diabetic rats via a neuroprotective effect. Exp Ther Med 2021; 21:501. [PMID: 33791010 PMCID: PMC8005692 DOI: 10.3892/etm.2021.9932] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 02/18/2021] [Indexed: 12/16/2022] Open
Abstract
Diabetic cystopathy (DCP) is one of the most common complications of diabetes mellitus (DM). A previous study reported that caffeine may improve bladder dysfunction in rats with DM. The aim of the present study was to investigate the mechanisms behind the capacity for caffeine to improve bladder function in rats with DM. Sprague Dawley rats were divided into four groups: control, caffeine, DM and DM plus caffeine treatment (DM + caffeine). Bladder function was measured by urodynamic analyses. The levels of nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF) and calcitonin gene-related peptide (CGRP) in the bladder tissue were detected by ELISA. Apoptosis in the dorsal root ganglion (DRG) was detected by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assay. The expression levels of B-cell lymphoma-2 (Bcl-2), Bcl-2-associated X protein (Bax), caspase-3, cleaved caspase-3, caspase-9 and cleaved caspase-9 proteins in the DRG were detected by western blotting. Following treatment with caffeine, the urination time and micturition interval of rats with DM were improved, the bladder wet weight was decreased, and the maximum voiding pressure was increased. Relative to that in the DM group, the expression levels of NGF, BDNF and CGRP in the bladder tissue of DM + caffeine rats increased; cellular apoptosis in the DRG of DM + caffeine rates decreased; and the expression levels of Bcl-2, Bax, cleaved caspase-3 and cleaved caspase-9 proteins in the DRG of DM + caffeine rats were restored to a certain extent. In conclusion, caffeine promotes bladder function in rats with DM through a protective effect on DRG.
Collapse
Affiliation(s)
- Jun Xue
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| | - Yadong Liu
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| | - Sichong Zhang
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| | - Liucheng Ding
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| | - Baixin Shen
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| | - Yunpeng Shao
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| | - Zhongqing Wei
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| |
Collapse
|
22
|
Chai GR, Liu S, Yang HW, Chen XL. Quercetin protects against diabetic retinopathy in rats by inducing heme oxygenase-1 expression. Neural Regen Res 2021; 16:1344-1350. [PMID: 33318415 PMCID: PMC8284280 DOI: 10.4103/1673-5374.301027] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Quercetin is a widely-occurring flavonoid that protects against cancer, and improves memory and cardiovascular functions. However, whether quercetin exhibits therapeutic effects in diabetic retinopathy remains unclear. In this study, we established a rat model of streptozocin-induced diabetic retinopathy. Seventy-two hours later, the rats were intraperitoneally administered 150 mg/kg quercetin for 16 successive weeks. Quercetin markedly increased the thickness of the retinal cell layer, increased the number of ganglion cells, and decreased the overexpression of the pro-inflammatory factors interleukin-1β, interleukin-18, interleukin-6 and tumor necrosis factor-α in the retinal tissue as well as the overexpression of high mobility group box-1 and the overactivation of the NLRP3 inflammasome. Furthermore, quercetin inhibited the overexpression of TLR4 and NF-κBp65, reduced the expression of the pro-angiogenic vascular endothelial growth factor and soluble intercellular adhesion molecule-1, and upregulated the neurotrophins brain-derived neurotrophic factor and nerve growth factor. Intraperitoneal injection of the heme oxygenase-1 inhibitor zinc protoporphyrin blocked the protective effect of quercetin. These findings suggest that quercetin exerts therapeutic effects in diabetic retinopathy possibly by inducing heme oxygenase-1 expression. This study was approved by the Animal Ethics Committee of China Medical University, China (approval No. 2016PS229K) on April 8, 2016.
Collapse
Affiliation(s)
- Guang-Rui Chai
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Shu Liu
- Department of Geratology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Hong-Wei Yang
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Xiao-Long Chen
- Department of Ophthalmology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
23
|
Aisa MC, Barbati A, Cappuccini B, De Rosa F, Gerli S, Clerici G, Kaptilnyy VA, Ishenko AI, Di Renzo GC. Urinary Nerve Growth Factor in full-term, preterm and intra uterine growth restriction neonates: Association with brain growth at 30-40 days of postnatal period and with neuro-development outcome at two years. A pilot study. Neurosci Lett 2020; 741:135459. [PMID: 33223047 DOI: 10.1016/j.neulet.2020.135459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 10/19/2020] [Accepted: 10/21/2020] [Indexed: 10/23/2022]
Abstract
Nerve Growth Factor (NGF) and Brain Derived Neurotrophic Factor (BDNF) are crucial for the peripheral and central nervous system development, respectively, and differential brain and blood levels in Intra Uterine Growth Restriction (IUGR) and prematurity have been found. As reduced growth of brain regions, measured at 30-40 days of postnatal period, has been demonstrated in preterm and IUGR neonates who showed impaired neuro-development at two years of age, in this study, the levels of NGF and BDNF were evaluated in the urine samples of 30-40 day-old subjects who were full-term, preterm and IUGR and showed a normal or an abnormal neuro-development at follow up after two years. Neurotrophins were measured concurrently with volumes of whole brain, thalamus, frontal cortex and cerebellum. Values were then correlated with later neuro-developmental outcome. Biochemical parameters and cerebral volumes were assessed using colorimetric ELISA kits and three-dimensional ultra-sonography (3DUS), respectively. Neuro-development was estimated using the Griffiths-II test. Urinary NGF and brain volumes significantly correlated and were lower in preterm and IUGR subjects characterized by poor neuro-development. No differences were seen in the case of BDNF. The present investigation demonstrates, for the first time, the strong and direct association of NGF with brain growth at the initial phase of the postnatal period and with neuro-developmental outcome in later life. Remarkably, urinary NGF may be suggested as an early prognostic indicator of high long-term risk of motor and cognitive impairment in IUGR and preterm neonates.
Collapse
Affiliation(s)
- Maria Cristina Aisa
- Department of Surgical and Biomedical Sciences, Section of Obstetrics and Gynecology, University of Perugia, Perugia, Italy; GeBiSa, Research Foundation, Perugia, Italy; Centro Europeo per la Medicina e la Ricerca (CEMER), Perugia, Italy.
| | - Antonella Barbati
- Department of Surgical and Biomedical Sciences, Section of Obstetrics and Gynecology, University of Perugia, Perugia, Italy
| | | | | | - Sandro Gerli
- Department of Surgical and Biomedical Sciences, Section of Obstetrics and Gynecology, University of Perugia, Perugia, Italy; GeBiSa, Research Foundation, Perugia, Italy; Department of Obstetrics and Gynecology No. 1 of the Institute of Clinical Medicine, I. M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Graziano Clerici
- Centro Europeo per la Medicina e la Ricerca (CEMER), Perugia, Italy; Department of Obstetrics and Gynecology No. 1 of the Institute of Clinical Medicine, I. M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Vitaly Alexandrovich Kaptilnyy
- Department of Obstetrics and Gynecology No. 1 of the Institute of Clinical Medicine, I. M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Anatoly Ivanovich Ishenko
- Department of Obstetrics and Gynecology No. 1 of the Institute of Clinical Medicine, I. M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Gian Carlo Di Renzo
- Department of Surgical and Biomedical Sciences, Section of Obstetrics and Gynecology, University of Perugia, Perugia, Italy; GeBiSa, Research Foundation, Perugia, Italy; Second Department of Obstetrics and Gynecology, I. M. Sechenov First State Medical University, 119992 Moscow, Russia; Centre of Perinatal and Reproductive Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
24
|
Dai C, Xiong J, Wang Y, Shen J, Velkov T, Xiao X. Nerve Growth Factor Confers Neuroprotection against Colistin-Induced Peripheral Neurotoxicity. ACS Infect Dis 2020; 6:1451-1459. [PMID: 32422040 DOI: 10.1021/acsinfecdis.0c00107] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Neurotoxicity is an unwanted side effect for patients when receiving parenteral colistin therapy. The development of effective neuroprotective agents that can be coadministered during colistin therapy remains a priority area in antimicrobial chemotherapy. The present study aimed to investigate the protective effect of nerve growth factor (NGF) against colistin-induced peripheral neurotoxicity using a murine model. C57BL/6 mice were randomly divided into the following 6 groups: (i) untreated control, (ii) NGF alone (36 μg/kg/day administered intraperitoneally), (iii) colistin alone (18 mg/kg/day administered intraperitoneally), and (iv-vi) colistin (18 mg/kg/day) plus NGF (9, 18, and 36 μg/kg/day). After treatment for 7 days, neurobehavioral and electrophysiology changes, histopathological assessments of sciatic nerve damage, and oxidative stress biomarkers were examined. The mRNA expression levels of Nrf2, HO-1, Akt, Bax, and caspase-3 and -9 were assessed using quantitative RT-PCR. The results showed that, across all the groups wherein NGF was coadministered with colistin, a marked attenuation of colistin-induced sciatic nerve damage and improved sensory and motor function were observed. In comparison to the colistin only treatment group, animals that received NGF displayed upregulated Nrf2 and HO-1 mRNA expression levels and downregulated Bax and caspase-3 and -9 mRNA expression levels. In summary, our study reveals that NGF coadministration protects against colistin-induced peripheral neurotoxicity via the activation of Akt and Nrf2/HO-1 pathways and inhibition of oxidative stress. This study highlights the potential clinical application of NGF as a neuroprotective agent for coadministration during colistin therapy.
Collapse
Affiliation(s)
- Chongshan Dai
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, P. R. China
| | - Jianli Xiong
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471023, P. R. China
| | - Yang Wang
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, P. R. China
| | - Jianzhong Shen
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, P. R. China
| | - Tony Velkov
- Department of Pharmacology & Therapeutics, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Xilong Xiao
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, P. R. China
| |
Collapse
|
25
|
Daverey A, Agrawal SK. Curcumin Protects against White Matter Injury through NF-κB and Nrf2 Cross Talk. J Neurotrauma 2020; 37:1255-1265. [PMID: 31914858 DOI: 10.1089/neu.2019.6749] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Inflammation and oxidative stress play a central role in the pathogenesis of white matter injury (WMI). Curcumin (Cur), a polyphenolic compound, exhibits anti-inflammatory and anti-oxidant effects on several conditions. The objective of this study was to investigate neuroprotective effects of Cur on WMI and explore its underlying mechanisms of action. Sprague-Dawley rats were subjected to the removal of white matter from the dorsal column of the spinal cord. Dorsal columns were randomly divided into three groups: Sham (Ringer's solution bubbled with 95% O2 and 5% CO2), hypoxia (Hyp; Ringer's solution bubbled with 95% N2 and 5% CO2 for 1 h), and Cur-treated (Hyp+Cur; Ringer's solution bubbled with 95% N2 and 5% CO2 for 1 h in the presence of 50 μM Cur). For NF-κB inhibition experiments, dorsal columns were incubated with 50 μM BAY 11-7082 (BAY) for 30 min in 95% O2 and 5% CO2 prior to 1-h incubation with 50 μM Cur in 95% N2 and 5% CO2. Our data show that Cur inhibited hypoxia-induced HIF1-α expression and tissue damage by demonstrating the improved morphology of astrocytes and remarkable reduction in vacuolation. Cur also inhibited the hypoxia-induced upregulation of glial fibrillary acidic protein (GFAP) and neurofilament-H (NF-H) after hypoxia and downregulated the expression of pro-inflammatory cytokines such as tumor necrosis factor alpha (TNF-α) and interleukin 1 (IL-1). Terminal dexynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL)-assay analysis showed that Cur effectively attenuated apoptosis in white matter. In addition, we demonstrated that Cur exerted its neuroprotective effect through cross talk between nuclear factor kappa-light-chain-enhancer of activated B (NF-κB) and nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathways. In conclusion, our results indicate that treatment with Cur inhibited the hypoxia, inflammation and apoptosis associated with WMI. Further, the Nrf-2 pathway inhibits NF-κB activation by preventing IkB degradation and increasing HO-1 expression, which in turn reduces reactive oxygen species (ROS) and as a result NF-κB activation is suppressed. Similarly, NF-κB-mediated transcription reduces Nrf2 activation by reducing anti-oxidant response element (ARE) gene and free CREB binding protein by competing with Nrf2 for CBP thus inhibiting the Nrf-2 activation.
Collapse
Affiliation(s)
- Amita Daverey
- Department of Neurosurgery, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Sandeep K Agrawal
- Department of Neurosurgery, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
26
|
Chen H, Zhang J, Dai Y, Xu J. Nerve growth factor inhibits TLR3-induced inflammatory cascades in human corneal epithelial cells. JOURNAL OF INFLAMMATION-LONDON 2019; 16:27. [PMID: 31889912 PMCID: PMC6933932 DOI: 10.1186/s12950-019-0232-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 12/18/2019] [Indexed: 12/14/2022]
Abstract
Background In herpes simplex epithelial keratitis, excessive TLR3-induced cellular responses after virus infection evoke inflammatory cascades that might be destructive to the host cornea. Nerve growth factor (NGF), a pluripotent neurotrophic factor with immune regulatory effect, was proved to be effective in Herpes simplex keratitis (HSK) treatment, although the detailed mechanisms remain unclear. This study aims to investigate the effects of NGF on modulating inflammatory responses triggered by TLR3 activation in human corneal epithelial cells (HCECs) in vitro. Methods HCECs were stimulated with TLR3 agonist, poly(I:C), in the absence or presence of NGF. Cell viability and cytotoxicity were measured by a CCK-8 assay and LDH release assay, respectively. The activation of NF-κB signaling pathway was examined using immunofluorescence staining and western blotting. Levels of proinflammatory cytokines were determined by ELISA or RT-qPCR. ROS generation and 8-OHdG positive cells were examined by a fluorometric analysis. Results It was shown that NGF significantly inhibited the generation of proinflammatory cytokines in HCECs triggered by TLR3 activation (P < 0.05), probably via suppressing NF-κB activation. NGF also impeded the upstream signal to initiate NF-κB activation by scavenging ROS by approximately 50% (P < 0.05). In addition, 8-OHdG positive cells were substantially attenuated by NGF treatment (P < 0.01). Conclusions Taken together, this study indicates that NGF could inhibit TLR3-induced inflammatory cascades in HCECs, suggesting NGF as a potential therapeutic agent for HSK.
Collapse
Affiliation(s)
- Huiyu Chen
- Department of Ophthalmology and Visual Science, Eye & ENT Hospital, NHC Key Laboratory of myopia (Fudan University); Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai Medical College of Fudan University, Shanghai, 200031 China
| | - Jing Zhang
- Department of Ophthalmology and Visual Science, Eye & ENT Hospital, NHC Key Laboratory of myopia (Fudan University); Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai Medical College of Fudan University, Shanghai, 200031 China
| | - Yiqin Dai
- Department of Ophthalmology and Visual Science, Eye & ENT Hospital, NHC Key Laboratory of myopia (Fudan University); Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai Medical College of Fudan University, Shanghai, 200031 China
| | - Jianjiang Xu
- Department of Ophthalmology and Visual Science, Eye & ENT Hospital, NHC Key Laboratory of myopia (Fudan University); Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai Medical College of Fudan University, Shanghai, 200031 China
| |
Collapse
|
27
|
Cecilia OM, José Alberto CG, José NP, Ernesto Germán CM, Ana Karen LC, Luis Miguel RP, Ricardo Raúl RR, Adolfo Daniel RC. Oxidative Stress as the Main Target in Diabetic Retinopathy Pathophysiology. J Diabetes Res 2019; 2019:8562408. [PMID: 31511825 PMCID: PMC6710812 DOI: 10.1155/2019/8562408] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 06/17/2019] [Accepted: 07/15/2019] [Indexed: 12/12/2022] Open
Abstract
Diabetic retinopathy (DR) is one of the most common complications of diabetes mellitus (DM) causing vision impairment even at young ages. There are numerous mechanisms involved in its development such as inflammation and cellular degeneration leading to endothelial and neural damage. These mechanisms are interlinked thus worsening the diabetic retinopathy outcome. In this review, we propose oxidative stress as the focus point of this complication onset.
Collapse
Affiliation(s)
- Olvera-Montaño Cecilia
- Institute of Clinical and Experimental Therapeutics, Department of Physiology, Health Sciences University Center, University of Guadalajara, Mexico
| | - Castellanos-González José Alberto
- Department of Ophthalmology, Specialties Hospital of the National Occidental Medical Center, Mexican Institute of Social Security, Mexico
| | - Navarro-Partida José
- Tecnológico de Monterrey Institute, School of Medicine and Health Sciences, Campus Guadalajara, Mexico
| | - Cardona-Muñoz Ernesto Germán
- Institute of Clinical and Experimental Therapeutics, Department of Physiology, Health Sciences University Center, University of Guadalajara, Mexico
| | - López-Contreras Ana Karen
- Institute of Clinical and Experimental Therapeutics, Department of Physiology, Health Sciences University Center, University of Guadalajara, Mexico
| | | | - Robles-Rivera Ricardo Raúl
- Institute of Clinical and Experimental Therapeutics, Department of Physiology, Health Sciences University Center, University of Guadalajara, Mexico
| | - Rodríguez-Carrizalez Adolfo Daniel
- Institute of Clinical and Experimental Therapeutics, Department of Physiology, Health Sciences University Center, University of Guadalajara, Mexico
| |
Collapse
|
28
|
Li D, Liu Y, Xu R, Jia X, Li X, Huo C, Wang X. RETRACTED ARTICLE: Astragalus polysaccharide alleviates H2O2-triggered oxidative injury in human umbilical vein endothelial cells via promoting KLF2. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:2188-2195. [PMID: 31159593 DOI: 10.1080/21691401.2019.1621886] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Dongtao Li
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Yan Liu
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Rong Xu
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Xin Jia
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Xing Li
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Cong Huo
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Xiaoming Wang
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| |
Collapse
|
29
|
Chen J, Du H, Zhang Y, Chen H, Zheng M, Lin P, Lan Q, Yuan Q, Lai Y, Pan X, Chen R, Liu N. Netrin-1 Prevents Rat Primary Cortical Neurons from Apoptosis via the DCC/ERK Pathway. Front Cell Neurosci 2017; 11:387. [PMID: 29321724 PMCID: PMC5733550 DOI: 10.3389/fncel.2017.00387] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 11/21/2017] [Indexed: 12/12/2022] Open
Abstract
In the nervous system, Netrin-1 serves as a neural guide, mediating the neuronal development. However, it remains blurred whether Netrin-1 can protect neurons from apoptosis induced by cerebral stroke. In the current study, the cultured rat primary cortical neurons were transfected with Netrin-1-encoding lentivirus before the oxygen-glucose-deprivation (OGD) treatment. Cell death and apoptosis were evaluated by lactate dehydrogenase (LDH) release and flow cytometry. We found that Netrin-1 attenuated OGD-induced cell death and neuronal apoptosis at 24 h after OGD treatment, and that the overexpression of Netrin-1 activated the ERK signaling pathway. These effects were partly abolished by blocking its receptor deleted in colorectal cancer (DCC) or U0126, an inhibitor of the ERK signaling pathway. Netrin-1 overexpression in neurons elevated the expression of DCC, on mRNA level and protein level. Netrin-1 also reduced DNA damage. Taken together, our findings suggest that Netrin-1 attenuates cell death and neuronal apoptosis via the DCC/ERK signaling pathway in the cultured primary cortical neurons after OGD injury, which may involve the mediation of DNA damage in the neurons.
Collapse
Affiliation(s)
- Jianhao Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China.,Institute of Cerebral Vascular Disease of Fujian Province, Fuzhou, China
| | - Houwei Du
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China.,Institute of Cerebral Vascular Disease of Fujian Province, Fuzhou, China
| | - Yixian Zhang
- Institute of Cerebral Vascular Disease of Fujian Province, Fuzhou, China.,Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China
| | - Hongbin Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China.,Institute of Cerebral Vascular Disease of Fujian Province, Fuzhou, China
| | - Mouwei Zheng
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China.,Institute of Cerebral Vascular Disease of Fujian Province, Fuzhou, China
| | - Peiqiang Lin
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China.,Institute of Cerebral Vascular Disease of Fujian Province, Fuzhou, China
| | - Quan Lan
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China.,Institute of Cerebral Vascular Disease of Fujian Province, Fuzhou, China
| | - Qilin Yuan
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China.,Institute of Cerebral Vascular Disease of Fujian Province, Fuzhou, China
| | - Yongxing Lai
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China.,Institute of Cerebral Vascular Disease of Fujian Province, Fuzhou, China
| | - Xiaodong Pan
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China.,Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Ronghua Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China.,Institute of Cerebral Vascular Disease of Fujian Province, Fuzhou, China
| | - Nan Liu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China.,Institute of Cerebral Vascular Disease of Fujian Province, Fuzhou, China.,Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
30
|
P21 Waf1/Cip1 depletion promotes dexamethasone-induced apoptosis in osteoblastic MC3T3-E1 cells by inhibiting the Nrf2/HO-1 pathway. Arch Toxicol 2017; 92:679-692. [PMID: 28940008 DOI: 10.1007/s00204-017-2070-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 09/18/2017] [Indexed: 12/22/2022]
Abstract
Dexamethasone (Dex), a glucocorticoid with strong anti-inflammatory and immunosuppressive activities, has been shown to exhibit marked cytotoxicity and apoptosis in osteoblasts, but the underlying mechanisms have not yet been comprehensively investigated. P21Waf1/Cip1 (p21) plays a critical role in the regulation of cell cycle progression and apoptosis. The present study aims to investigate the role of p21 in Dex-induced apoptosis in osteoblastic MC3T3-E1 cells, and to explore its mechanisms. Results demonstrated that Dex-induced apoptosis decreased the phosphorylation of Akt in a concentration-dependent manner. Moreover, LY294002, an inhibitor of the PI3K/Akt pathway enhanced the Dex-induced apoptosis of osteoblasts. On the contrary, insulin-like growth factor-1 (IGF-1), an activator of PI3K/Akt, attenuated the apoptosis of Dex in MC3T3-E1 cells. The protein level of p21 was downregulated by shortening its half-life, which was associated with inhibition of the PI3K/Akt pathway by Dex. Furthermore, depletion of p21 by siRNA enhanced Dex-induced caspase-3 activation and ROS generation, and promoted apoptosis of MC3T3-E1 cells. In addition, suppression of p21 led to a reduction of Dex-induced upregulation of nuclear Nrf2 and heme oxygenase-1 (HO-1) protein levels. These findings demonstrate that p21 depletion promotes Dex-induced apoptosis of MC3T3-E1 cells by inhibiting the antioxidant Nrf2/HO-1 pathway, which highlights the anti-apoptotic effect of p21 in MC3T3-E1 cells.
Collapse
|