1
|
Hasan A, Repici A, Capra AP, Mannino D, Bova V, Catalfamo A, Campolo M, Paterniti I, Esposito E, Ardizzone A. CCR1 antagonist as a potential modulator of inflammatory, autophagic, and apoptotic markers in spinal cord injury. Neuropharmacology 2025; 264:110239. [PMID: 39608704 DOI: 10.1016/j.neuropharm.2024.110239] [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: 10/22/2024] [Revised: 11/15/2024] [Accepted: 11/23/2024] [Indexed: 11/30/2024]
Abstract
Spinal cord injury (SCI) leads to severe and lasting impairments in motor and sensory functions. The intense inflammatory response following SCI is a significant challenge, and autophagy has emerged as a key factor in the recovery process. The C-C chemokine receptor type 1 (CCR1), a G-protein coupled receptor, plays a crucial role in managing the chemokine response under stress. BX471, a selective and potent CCR1 antagonist, has been explored in various disease contexts for its therapeutic potential. In this study, we assessed the effects of BX471 in a mouse model of SCI. The treatment was administered at doses of 3 and 10 mg/kg, 1 h and 6 h after the injury occurred. Results showed that BX471 significantly improved tissue structure by positively influencing autophagy and reducing inflammation. Inflammatory markers, including CCR1 ligands RANTES, MIP-1α, TNF-α, and IL-1β, were measured using Western blot analysis. Additionally, histological evaluations revealed that BX471 effectively decreased infiltration and reduced astrocyte and microglial activation, supporting the idea that enhancing autophagy through CCR1 inhibition could promote neuronal survival. The highest efficacy was observed at the 10 mg/kg dose, leading to optimal out-comes across the assessments. These findings suggest that CCR1 blockade with BX471 may offer a promising therapeutic strategy for SCI, addressing a critical gap in the current pharmacological treatment options.
Collapse
Affiliation(s)
- Ahmed Hasan
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, 31, 98166, Messina, Italy; School of Advanced Studies, Center of Neuroscience, University of Camerino, 62032, Camerino, Italy
| | - Alberto Repici
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, 31, 98166, Messina, Italy
| | - Anna Paola Capra
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, 31, 98166, Messina, Italy
| | - Deborah Mannino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, 31, 98166, Messina, Italy
| | - Valentina Bova
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, 31, 98166, Messina, Italy
| | - Antonio Catalfamo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, 31, 98166, Messina, Italy
| | - Michela Campolo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, 31, 98166, Messina, Italy
| | - Irene Paterniti
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, 31, 98166, Messina, Italy
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, 31, 98166, Messina, Italy.
| | - Alessio Ardizzone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, 31, 98166, Messina, Italy
| |
Collapse
|
2
|
Dong C, Sun Q, Zheng F, Ji J, Xu X, Yao X, Gan D. Heavy moxibustion at Sanyin point ameliorates neurogenic bladder dysfunction in spinal cord injury rats through the PI3 K/mTOR pathway. J Spinal Cord Med 2024:1-11. [PMID: 39660978 DOI: 10.1080/10790268.2024.2420431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2024] Open
Abstract
OBJECTIVE The present study aims to investigate the effect and mechanism of heavy moxibustion (100 moxa-cone) at Sanyin point (the common point of Yin and kidney) on the function of neurogenic bladder (NB) dysfunction in rats with spinal cord injury (SCI). METHODS Twenty-four male Sprague-Dawley rats were divided into four groups (n = 6): control, NB, NB + Moxibustion, and NB + Moxibustion + YS-49 (PI3 K agonist). The rats in control groups accepted a cut open of the skin, fascia, and muscle. The NB model was established using spinal cord transection. Fourteen days later, animals received heavy moxibustion at Sanyin point for three weeks or/and intraperitoneal administration of YS-49 (a PI3 K agonist). Basso, Beattie, and Bresnahan (BBB) scale, urodynamic parameters, bladder size, and weight were measured. The hematoxylin-eosin staining method was used to observe the histology of the bladder mucosa. Moreover, NB dysfunction after SCI could be restored by autophagy activation and autophagy is mediated by the PI3 K/Akt/mTOR pathway. Therefore, the expressions of autophagy factor (LC3 II/I and p62), PI3 K, and p-mTOR in the bladder mucosa were evaluated by western blotting. RESULTS Heavy moxibustion treatment relieved the development of NB dysfunction in rats with SCI, with an increase in the bladder voiding efficiency and a decrease in afferent activity during storage in the moxibustion group compared with the NB group. The expression levels of LC3 II/I were markedly elevated by moxibustion, accompanied by a decrease in the levels of p62. YS-49 addition increased the PI3 K and p-mTOR expression which were down-regulated by moxibustion. Importantly, YS-49 reversed the effects of moxibustion on autophagy and bladder function. CONCLUSION Heavy moxibustion at Sanyin point exerted its effect on healing-impaired NB dysfunction in rats with SCI, possibly activating autophagy through the PI3 K/mTOR pathway.
Collapse
Affiliation(s)
- Chunxuan Dong
- Department of Rehabilitation, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, People's Republic of China
| | - Qibin Sun
- Department of Workstation of Sun Qibin, The Famous Old TCM Doctor in China, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, People's Republic of China
| | - Fangjiang Zheng
- Department of Publicity, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, People's Republic of China
| | - Jing Ji
- Department of Rehabilitation, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, People's Republic of China
| | - Xiumei Xu
- Department of Rehabilitation, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, People's Republic of China
| | - Xingzhang Yao
- Department of Orthopedic Reconstruction, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, People's Republic of China
| | - Decheng Gan
- Department of Traditional Chinese Medicine, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, People's Republic of China
| |
Collapse
|
3
|
Levett JJ, Georgiopoulos M, Martel S, Mugheiry WA, Stavropoulos NA, Vega-Arroyo M, Santaguida C, Weber MH, Golan JD, Jarzem P, Ouellet JA, Klironomos G, Demetriades AK. Pharmacological Treatment of Degenerative Cervical Myelopathy: A Critical Review of Current Evidence. Neurospine 2024; 21:375-400. [PMID: 38955515 PMCID: PMC11224758 DOI: 10.14245/ns.2448140.070] [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: 02/07/2024] [Revised: 03/27/2024] [Accepted: 04/01/2024] [Indexed: 07/04/2024] Open
Abstract
Degenerative cervical myelopathy (DCM) is the leading cause of spinal cord dysfunction in adults, representing substantial morbidity and significant financial and resource burdens. Typically, patients with progressive DCM will eventually receive surgical treatment. Nonetheless, despite advancements in pharmacotherapeutics, evidence for pharmacological therapy remains limited. Health professionals from various fields would find interest in pharmacological agents that could benefit patients with mild DCM or enhance surgical outcomes. This review aims to consolidate all clinical and experimental evidence on the pharmacological treatment of DCM. We conducted a comprehensive narrative review that presents all pharmacological agents that have been investigated for DCM treatment in both humans and animal models. Riluzole exhibits effectiveness solely in rat models, but not in treating mild DCM in humans. Cerebrolysin emerges as a potential neuroprotective agent for myelopathy in animals but had contradictory results in clinical trials. Limaprost alfadex demonstrates motor function improvement in animal models and exhibits promising outcomes in a small clinical trial. Glucocorticoids not only fail to provide clinical benefits but may also lead to adverse events. Cilostazol, anti-Fas ligand antibody, and Jingshu Keli display promise in animal studies, while erythropoietin, granulocyte colony-stimulating factor and limaprost alfadex exhibit potential in both animal and human research. Existing evidence mainly rests on weak clinical data and animal experimentation. Current pharmacological efforts target ion channels, stem cell differentiation, inflammatory, vascular, and apoptotic pathways. The inherent nature and pathogenesis of DCM offer substantial prospects for developing neurodegenerative or neuroprotective therapies capable of altering disease progression, potentially delaying surgical intervention, and optimizing outcomes for those undergoing surgical decompression.
Collapse
Affiliation(s)
- Jordan J Levett
- Faculty of Medicine, University of Montreal, Montreal, QC, Canada
| | - Miltiadis Georgiopoulos
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Spinal Surgery Unit, Swansea Bay University Health Board, Swansea, UK
| | - Simon Martel
- Division of Orthopaedic Surgery, McGill University, Montreal, QC, Canada
| | - Wissam Al Mugheiry
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Nikolaos A. Stavropoulos
- First Department of Orthopaedic Surgery NKUA, “ATTIKON” University General Hospital, Athens, Greece
| | - Miguel Vega-Arroyo
- Winnipeg Spine Program, University of Manitoba, Winnipeg, MB, Canada
- Neurosurgery Department, Sanford Brain & Spine Center, Fargo, ND, USA
| | - Carlo Santaguida
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Michael H. Weber
- Division of Orthopaedic Surgery, McGill University, Montreal, QC, Canada
| | - Jeff D. Golan
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Peter Jarzem
- Division of Orthopaedic Surgery, McGill University, Montreal, QC, Canada
| | - Jean A. Ouellet
- Division of Orthopaedic Surgery, McGill University, Montreal, QC, Canada
| | - Georgios Klironomos
- Department of Neurosurgery, Zucker School of Medicine at Hofstra/Northwell, Bay Shore, NY, USA
| | - Andreas K. Demetriades
- Edinburgh Spinal Surgery Outcomes Study Group, Department of Neurosurgery, Royal Infirmary, Edinburgh, UK
| |
Collapse
|
4
|
Wang X, Zhang W, Zeng S, Wang L, Wang B. Collagenase Type I and Probucol-Loaded Nanoparticles Penetrate the Extracellular Matrix to Target Hepatic Stellate Cells for Hepatic Fibrosis Therapy. Acta Biomater 2024; 175:262-278. [PMID: 38141933 DOI: 10.1016/j.actbio.2023.12.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 12/12/2023] [Accepted: 12/15/2023] [Indexed: 12/25/2023]
Abstract
Hepatic fibrosis is a common pathological process in chronic liver diseases, characterized by excessive reactive oxygen species (ROS), activated hepatic stellate cells (HSCs), and massive synthesis of extracellular matrix (ECM), which are important factors in the development of liver cirrhosis, liver failure, and liver cancer. During the development of hepatic fibrosis, ECM collagen produced by activated HSCs significantly hinders medication delivery to targeted cells and reduces the efficiency of pharmacological therapy. In this study, we designed a multifunctional hyaluronic acid polymeric nanoparticle (HA@PRB/COL NPs) based on autophagy inhibitor probucol (PRB) and collagenase type I (COL) modification, which could enhance ECM degradation and accurately target HSCs through specificity binding CD44 receptor in hepatic fibrosis therapy. Upon encountering excessive collagen I-deposition formed barrier, HA@PRB/COL NPs performed the nanodrill-like function to effectively degrade pericellular collagen I, leading to greater ECM penetration and prominent HSCs internalization capacity of delivered PRB. In mouse hepatic fibrosis model, HA@PRB/COL NPs were efficiently delivered to HSCs through binding CD44 receptor to achieve efficient accumulation in fibrotic liver. Further, we showed that HA@PRB/COL NPs executed the optimal anti-fibrotic activity by inhibiting autophagy and activation of HSCs. In conclusion, our novel dual-functional co-delivery system with degrading fibrotic ECM collagen and targeting activated HSCs exhibits great potentials in the treatment of hepatic fibrosis in clinic. STATEMENT OF SIGNIFICANCE: The excess release of extracellular matrix (ECM) such as collagen in hepatic fibrosis hinders medication delivery and decreases the efficiency of pharmacological drugs. We aimed to develop a nano-delivery carrier system with protein hydrolyzed surfaces and further encapsulated an autophagy inhibitor (PRB) to enhance fibrosis-related ECM degradation-penetration and hepatic stellate cells (HSCs) targeting in hepatic fibrosis niche (HA@PRB/COL NPs). The COL of HA@PRB/COL NPs successfully worked as a scavenger to promote the digestion of the ECM collagen I barrier for deeper penetration into fibroid liver tissue. It also accurately targeted HSCs through specifically binding to the CD44 receptor and subsequently released PRB to inhibit autolysosome and ROS generation, thus preventing HSCs activation. Our HA@PRB/COL NPs system provided a promising therapeutic strategy for hepatic fibrosis in a clinic setting.
Collapse
Affiliation(s)
- Xiaowei Wang
- Department of Biobank, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Wenjun Zhang
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Sheng Zeng
- Clinical Stem Cell Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| | - Liudi Wang
- Clinical Stem Cell Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| | - Bin Wang
- Clinical Stem Cell Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China.
| |
Collapse
|
5
|
Xu W, Sun Y, Zhao S, Zhao J, Zhang J. Identification and validation of autophagy-related genes in primary open-angle glaucoma. BMC Med Genomics 2023; 16:287. [PMID: 37968618 PMCID: PMC10648356 DOI: 10.1186/s12920-023-01722-5] [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: 04/24/2023] [Accepted: 10/31/2023] [Indexed: 11/17/2023] Open
Abstract
BACKGROUND As the most common type of glaucoma, the etiology of primary open-angle glaucoma (POAG) has not been unified. Autophagy may affect the occurrence and development of POAG, while the specific mechanism and target need to be further explored. METHODS The GSE27276 dataset from the Gene Expression Omnibus (GEO) database and the autophagy gene set from the GeneCards database were selected to screen differentially expressed autophagy-related genes (DEARGs) of POAG. Hub DEARGs were selected by constructing protein-protein interaction (PPI) networks and utilizing GSE138125 dataset. Subsequently, immune cell infiltration analysis, genome-wide association study (GWAS) analysis, gene set enrichment analysis (GSEA) and other analyses were performed on the hub genes. Eventually, animal experiments were performed to verify the mRNA levels of the hub genes by quantitative real time polymerase chain reaction (qRT-PCR). RESULTS A total of 67 DEARGs and 2 hub DEARGs, HSPA8 and RPL15, were selected. The hub genes were closely related to the level of immune cell infiltration. GWAS analysis confirmed that the causative regions of the 2 hub genes in glaucoma were on chromosome 11 and chromosome 3, respectively. GSEA illustrated that pathways enriched for highly expressed HSPA8 and RPL15 contained immunity, autophagy, gene expression and energy metabolism-related pathways. qRT-PCR confirmed that the expression of Hspa8 and Rpl15 in the rat POAG model was consistent with the results of bioinformatics analysis. CONCLUSIONS This study indicated that HSPA8 and RPL15 may affect the progression of POAG by regulating autophagy and provided new ideas for the pathogenesis and treatment of POAG.
Collapse
Affiliation(s)
- Wanjing Xu
- Ophthalmology Department of QingPu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, China.
| | - Yuhao Sun
- Otolaryngology Department of QingPu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Shuang Zhao
- Graduate School of Shandong First Medical University, Jinan, China
| | - Jun Zhao
- Ophthalmology Department of Linyi People's Hospital, Linyi, China
| | - Juanmei Zhang
- Ophthalmology Department of Linyi People's Hospital, Linyi, China
| |
Collapse
|
6
|
Guo S, Xu Z, Feng Q, Zhang H, Yu D, Li B, Hu K, Gao X, Zhang Q, Yi H, Wu X, Song D, Zhu H, Cai H, Peng Y, Zhu W, Shi J. Molecular mechanism by which RRM2-inhibitor (cholagogue osalmid) plus bafilomycin A1 cause autophagic cell death in multiple myeloma. Arch Biochem Biophys 2023; 747:109771. [PMID: 37776936 DOI: 10.1016/j.abb.2023.109771] [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: 08/18/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/02/2023]
Abstract
Despite significant improvement in the prognosis of multiple myeloma (MM), the disease remains incurable; thus, more effective therapies are required. Ribonucleoside-diphosphate reductase subunit M2 (RRM2) is significantly associated with drug resistance, rapid relapse, and poor prognosis. Previously, we found that 4-hydroxysalicylanilide (osalmid), a specific inhibitor of RRM2, exhibits anti-MM activity in vitro, in vivo, and in human patients; however, the mechanism remains unclear. Osalmid inhibits the translocation of RRM2 to the nucleus and stimulates autophagosome synthesis but inhibits subsequent autophagosome-lysosome fusion. We confirm that RRM2 binds to receptor-interacting protein kinase 3 (RIPK3) and reduces RIPK3, inhibiting autophagosome-lysosome fusion. Interestingly, the combination of osalmid and bafilomycin A1 (an autophagy inhibitor) depletes RIPK3 and aggravates p62 and autophagosome accumulation, leading to autophagic cell death. Combination therapy demonstrates synergistic cytotoxicity both in vitro and in vivo. Therefore, we propose that combining osalmid and bafilomycin A1(BafA1) may have clinical benefits against MM.
Collapse
Affiliation(s)
- Shushan Guo
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Zhijian Xu
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Qilin Feng
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Hui Zhang
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Dandan Yu
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Bo Li
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Ke Hu
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Xuejie Gao
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Qikai Zhang
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Hongfei Yi
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Xiaosong Wu
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Dongliang Song
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Huabin Zhu
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Haiyan Cai
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yu Peng
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Weiliang Zhu
- State Key Laboratory of Drug Research, Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Jumei Shi
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| |
Collapse
|
7
|
Pluta R. The Dual Role of Autophagy in Postischemic Brain Neurodegeneration of Alzheimer's Disease Proteinopathy. Int J Mol Sci 2023; 24:13793. [PMID: 37762096 PMCID: PMC10530906 DOI: 10.3390/ijms241813793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/02/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Autophagy is a self-defense and self-degrading intracellular system involved in the recycling and elimination of the payload of cytoplasmic redundant components, aggregated or misfolded proteins and intracellular pathogens to maintain cell homeostasis and physiological function. Autophagy is activated in response to metabolic stress or starvation to maintain homeostasis in cells by updating organelles and dysfunctional proteins. In neurodegenerative diseases, such as cerebral ischemia, autophagy is disturbed, e.g., as a result of the pathological accumulation of proteins associated with Alzheimer's disease and their structural changes. Postischemic brain neurodegeneration, such as Alzheimer's disease, is characterized by the accumulation of amyloid and tau protein. After cerebral ischemia, autophagy was found to be activated in neuronal, glial and vascular cells. Some studies have shown the protective properties of autophagy in postischemic brain, while other studies have shown completely opposite properties. Thus, autophagy is now presented as a double-edged sword with possible therapeutic potential in brain ischemia. The exact role and regulatory pathways of autophagy that are involved in cerebral ischemia have not been conclusively elucidated. This review aims to provide a comprehensive look at the advances in the study of autophagy behavior in neuronal, glial and vascular cells for ischemic brain injury. In addition, the importance of autophagy in neurodegeneration after cerebral ischemia has been highlighted. The review also presents the possibility of modulating the autophagy machinery through various compounds on the development of neurodegeneration after cerebral ischemia.
Collapse
Affiliation(s)
- Ryszard Pluta
- Department of Pathophysiology, Medical University of Lublin, 20-090 Lublin, Poland
| |
Collapse
|
8
|
Qiao L, Tang Q, An Z, Qi J. Minocycline relieves neuropathic pain in rats with spinal cord injury via activation of autophagy and suppression of PI3K/Akt/mTOR pathway. J Pharmacol Sci 2023; 153:12-21. [PMID: 37524449 DOI: 10.1016/j.jphs.2023.06.002] [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: 09/29/2022] [Revised: 04/06/2023] [Accepted: 06/20/2023] [Indexed: 08/02/2023] Open
Abstract
OBJECTIVE In this study, we studied whether minocycline hydrochloride improved neuropathic pain induced by spinal cord injury (SCI) in rats through PI3K/Akt pathway. METHODS The SCI was induced by compressed at level of T9-T11 of spinal cord in Sprague Dawley male rats. Animals were given different concentrations of minocycline (3 mg/kg, 30 mg/kg, 90 mg/kg) at the first and 24 h after SCI, then subsequently every 7, 12, 16, 20, 25 days via peroral route. The locomotor function was assessed by Basso Mouse Scale (BMS). The changes of spinal cord tissues were observed by HE. The inflammatory cytokines in spinal cord, IL-6, IL-1β and TNF-α, were measured by ELISA. The LC3B levels of spinal cord were observed by immunofluorescence. The autophagy related proteins and PI3K/AKT pathway related proteins were analysed by Western blot. Furthermore, the PI3K/AKT pathway inhibitor LY294002, and activator IGF-1 were used to confirm the mechanism of minocycline. RESULTS Contrasted to sham group, the inflammatory response in spinal cord was enhanced after SCI. Compared with the SCI rats, minocycline treatment significantly improved the locomotor activity, pathological injury of spinal cord, suppressed the levels of inflammatory factors. In addition, minocycline treatment upregulated autophagy response in damaged spinal cord through increasing LC3B, Beclin-1 and decreasing P62. The results of mechanism study showed that minocycline treatment clearly suppressed phosphorylation of PI3K, Akt and mTOR proteins expression. CONCLUSION Minocycline could improve neuropathic pain induced by SCI through activating autophagy and inhibiting PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Leyan Qiao
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China
| | - Qian Tang
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China
| | - Zhongzhe An
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China.
| | - Jun Qi
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, China.
| |
Collapse
|
9
|
Li J, Cao Y, Li LN, Chu X, Wang YS, Cai JJ, Zhao J, Ma S, Li G, Fan ZK. Neuroprotective Effects of Oxymatrine via Triggering Autophagy and Inhibiting Apoptosis Following Spinal Cord Injury in Rats. Mol Neurobiol 2023; 60:4450-4471. [PMID: 37115405 DOI: 10.1007/s12035-023-03364-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 04/20/2023] [Indexed: 04/29/2023]
Abstract
Spinal cord injury (SCI) is a devastating neurological disorder characterized by high morbidity and disability. However, there is still a lack of effective treatments for it. The identification of drugs that promote autophagy and inhibit apoptosis in neurons is critical for improving patient outcomes following SCI. Previous studies have shown that increasing the activity of silent information regulator 1 (SIRT1) and downstream protein AMP-activated protein kinase (AMPK) in rat models of SCI is highly neuroprotective. Oxymatrine (OMT), a quinolizidine alkaloid, has exhibited neuroprotective effects in various central nervous system (CNS) diseases. However, its explicit effect and molecular mechanism in SCI are still unclear. Herein, we aimed to investigate the therapeutic effects of OMT and explore the potential role of autophagy regulation following SCI in rats. A modified compressive device (weight 35 g, time 5 min) was applied to induce moderate SCI in all groups except the sham group. After treatment with drugs or vehicle (saline), our results indicated that OMT treatment significantly reduced the lesion size, promoted survival of motor neurons, and subsequently attenuated motor dysfunction following SCI in rats. OMT significantly enhanced autophagy activity, inhibited apoptosis in neurons, and increased SIRT1 and p-AMPK expression levels. Interestingly, these effects of OMT on SCI were partially prevented by co-treatment with SIRT1 inhibitor EX527. Furthermore, combining OMT with the potent autophagy inhibitor chloroquine (CQ) could effectively abolish its promotion of autophagic flux. Taken together, these data revealed that OMT exerts a neuroprotective role in functional recovery against SCI in rats, and these effects are potentially associated with OMT-induced activation of autophagy via the SIRT1/AMPK signaling pathway.
Collapse
Affiliation(s)
- Jian Li
- Department of Orthopedics, First Affiliated Hospital, Jinzhou Medical University, Jinzhou, 121000, China
| | - Yang Cao
- Department of Orthopedics, First Affiliated Hospital, Jinzhou Medical University, Jinzhou, 121000, China
| | - Lin-Na Li
- Departments of Endocrinology, First Affiliated Hospital, Jinzhou Medical University, Jinzhou, 121000, China
| | - Xin Chu
- Department of Orthopedics, First Affiliated Hospital, Jinzhou Medical University, Jinzhou, 121000, China
| | - Yan-Song Wang
- Department of Orthopedics, First Affiliated Hospital, Jinzhou Medical University, Jinzhou, 121000, China
| | - Jia-Jun Cai
- Department of Orthopedics, First Affiliated Hospital, Jinzhou Medical University, Jinzhou, 121000, China
| | - Jin Zhao
- Department of Orthopedics, First Affiliated Hospital, Jinzhou Medical University, Jinzhou, 121000, China
| | - Song Ma
- Department of Orthopedics, First Affiliated Hospital, Jinzhou Medical University, Jinzhou, 121000, China
| | - Gang Li
- Department of Orthopedics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Qingdao, 266035, China.
| | - Zhong-Kai Fan
- Department of Orthopedics, First Affiliated Hospital, Jinzhou Medical University, Jinzhou, 121000, China.
| |
Collapse
|
10
|
Pu PM, Li ZY, Dai YX, Sun YL, Wang YJ, Cui XJ, Yao M. Analysis of gene expression profiles and experimental validations of a rat chronic cervical cord compression model. Neurochem Int 2023:105564. [PMID: 37286109 DOI: 10.1016/j.neuint.2023.105564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/31/2023] [Accepted: 06/04/2023] [Indexed: 06/09/2023]
Abstract
Cervical spondylotic myelopathy (CSM) is a severe non-traumatic spinal cord injury (SCI) wherein the spinal canal and cervical cord are compressed due to the degeneration of cervical tissues. To explore the mechanism of CSM, the ideal model of chronic cervical cord compression in rats was constructed by embedding a polyvinyl alcohol polyacrylamide hydrogel in lamina space. Then, the RNA sequencing technology was used to screen the differentially expressed genes (DEGs) and enriched pathways among intact and compressed spinal cords. A total of 444 DEGs were filtered out based on the value of log2(Compression/Sham); these were associated with IL-17, PI3K-AKT, TGF-β, and Hippo signaling pathways according to the GSEA, KEGG, and GO analyses. Transmission electron microscopy indicated the changes in mitochondrial morphology. Western blot and immunofluorescent staining revealed neuronal apoptosis, astrogliosis and microglial neuroinflammation in the lesion area. Specifically, the expression of apoptotic indicators, such as Bax and cleaved caspase-3, and inflammatory cytokines, such as IL-1β, IL-6, and TNF-α, were upregulated. The activation of IL-17 signaling pathways was observed in microglia instead of neurons or astrocytes, the activation of TGF-β and inhibition of Hippo signaling pathways were detected in astrocytes instead of neurons or microglia, and the inhibition of PI3K-AKT signaling pathway was discovered in neurons rather than microglia of astrocytes in the lesion area. In conclusion, this study indicated that neuronal apoptosis was accompanied by inhibiting of the PI3K-AKT pathway. Then, the activation of microglia IL-17 pathway and NLRP3 inflammasome effectuated the neuroinflammation, and astrogliosis was ascribed to the activation of TGF-β and the inhibition of the Hippo pathway in the chronic cervical cord of compression. Therefore, therapeutic methods targeting these pathways in nerve cells could be promising CSM treatments.
Collapse
Affiliation(s)
- Pei-Min Pu
- Spine Disease Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai, 200032, China; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Zhuo-Yao Li
- Spine Disease Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai, 200032, China; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Yu-Xiang Dai
- Spine Disease Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai, 200032, China; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Yue-Li Sun
- Spine Disease Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai, 200032, China; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Yong-Jun Wang
- Spine Disease Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai, 200032, China; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Xue-Jun Cui
- Spine Disease Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai, 200032, China; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Min Yao
- Spine Disease Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai, 200032, China; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
11
|
Sheng Q, Sun Y, Zhai R, Fan X, Ying Y, Liu Z, Kong X. Murine cytomegalovirus localization and uveitic cell infiltration might both contribute to trabecular meshwork impairment in Posner-Schlossman syndrome: Evidence from an open-angle rat model. Exp Eye Res 2023; 231:109477. [PMID: 37137438 DOI: 10.1016/j.exer.2023.109477] [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: 01/31/2023] [Revised: 03/22/2023] [Accepted: 04/16/2023] [Indexed: 05/05/2023]
Abstract
As a special type of glaucoma, Posner-Schlossman syndrome (PSS) is characterized by elevated intraocular pressure (IOP) and anterior uveitis. Cytomegalovirus (CMV) anterior chamber infection has now been considered the leading cause of PSS. We used murine CMV (MCMV) intracameral injection to establish a rat model manifested in IOP elevation and mild anterior uveitis, much like PSS; viral localization and gene expression at various time points and inflammatory cell infiltration derived from innate and adaptive immunity were investigated, as well as pathogenetic changes of the trabecular meshwork (TM). The IOP and uveitic manifestations peaked at 24 h post-infection (p.i.) and returned to normal after 96 h; the iridocorneal angle remained open consistently. At 24 h p.i., leucocytes gathered at the chamber angle. Maximum transcription of MCMV immediate early 1 (IE1) was reached at 24 h in the cornea and 48 h in the iris and ciliary body. MCMV localized in aqueous humor outflow facilities and the iris from 24 h to 28 d p.i. and was detected by in situ hybridization, though it did not transcribe after 7 d p.i. TM and iris pigment epithelial cells harboring viral inclusion bodies and autophagosomes were present at 28 d p.i. These findings shed light on how and where innate and adaptive immunity reacted after MCMV was found and transcribed in a highly ordered cascade, as well as pathogenetic changes in TM as a result of virus and uveitis behaviors.
Collapse
Affiliation(s)
- Qilian Sheng
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China; National Health Commission Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, 83 Fenyang Rd., 200031, Shanghai, China
| | - Yanan Sun
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China; National Health Commission Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, 83 Fenyang Rd., 200031, Shanghai, China
| | - Ruyi Zhai
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China; National Health Commission Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, 83 Fenyang Rd., 200031, Shanghai, China
| | - Xintong Fan
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China; National Health Commission Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, 83 Fenyang Rd., 200031, Shanghai, China
| | - Yue Ying
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China; National Health Commission Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, 83 Fenyang Rd., 200031, Shanghai, China
| | - Zhijun Liu
- Department of Medical Microbiology, Weifang Medical University, Weifang, 261053, China.
| | - Xiangmei Kong
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China; National Health Commission Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, 83 Fenyang Rd., 200031, Shanghai, China.
| |
Collapse
|
12
|
Smith SS, Young AMH, Davies BM, Takahashi H, Allinson KSJ, Kotter MRN. Evidence of impaired macroautophagy in human degenerative cervical myelopathy. Sci Rep 2022; 12:11873. [PMID: 35831377 PMCID: PMC9279443 DOI: 10.1038/s41598-022-15158-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 06/20/2022] [Indexed: 11/23/2022] Open
Abstract
Degenerative cervical myelopathy (DCM) is a common progressive disease of the spinal cord which can cause tetraplegia. Despite its prevalence, few studies have investigated the pathophysiology of DCM. Macroautophagy is a cellular process which degrades intracellular contents and its disruption is thought to contribute to many neurodegenerative diseases. The present study tests the hypothesis that macroautophagy is impaired in DCM. To address this, we utilised a collection of post-mortem cervical spinal cord samples and investigated seven DCM cases and five human controls. Immunohistochemical staining was used to visualise proteins involved in autophagy. This demonstrated significantly reduced numbers of LC3 puncta in cases versus controls (p = 0.0424). Consistent with reduced autophagy, we identified large aggregates of p62 in four of seven cases and no controls. Tau was increased in two of five cases compared to controls. BCL-2 was significantly increased in cases versus controls (p = 0.0133) and may explain this reduction in autophagy. Increased BCL-2 (p = 0.0369) and p62 bodies (p = 0.055) were seen in more severe cases of DCM. This is the first evidence that autophagy is impaired in DCM; the impairment appears greater in more severe cases. Further research is necessary to investigate whether macroautophagy has potential as a therapeutic target in DCM.
Collapse
Affiliation(s)
- Sam S Smith
- School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Adam M H Young
- Academic Neurosurgery Unit, Department of Clinical Neurosurgery, University of Cambridge, Cambridge, UK
| | - Benjamin M Davies
- Academic Neurosurgery Unit, Department of Clinical Neurosurgery, University of Cambridge, Cambridge, UK
| | - Hitoshi Takahashi
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Kieren S J Allinson
- Cambridge University Hospitals NHS Foundation Trust and the Cambridge Brain Bank, Cambridge, UK
| | - Mark R N Kotter
- Academic Neurosurgery Unit, Department of Clinical Neurosurgery, University of Cambridge, Cambridge, UK.
- WT MRC Cambridge Stem Cell Institute, Anne McLaren Laboratory, University of Cambridge, Cambridge, UK.
| |
Collapse
|
13
|
Abbaszadeh F, Jorjani M, Joghataei MT, Mehrabi S. Astaxanthin Modulates Autophagy, Apoptosis, and Neuronal Oxidative Stress in a Rat Model of Compression Spinal Cord Injury. Neurochem Res 2022; 47:2043-2051. [PMID: 35435619 DOI: 10.1007/s11064-022-03593-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 03/28/2022] [Accepted: 03/28/2022] [Indexed: 10/18/2022]
Abstract
The effects of astaxanthin (AST) were evaluated on oxidative mediators, neuronal apoptosis, and autophagy in functional motor recovery after spinal cord injury (SCI). Rats were divided into three groups of sham, SCI + DMSO (dimethyl sulfoxide), and SCI + AST. Rats in the sham group only underwent a laminectomy at thoracic 8-9. While, the SCI + DMSO and SCI + AST groups had a compression SCI with an aneurysm clip. Then, this groups received an intrathecal (i.t.) injection of 5% DMSO and AST (10 μl of 0.005 mg/kg), respectively. The rat motor functions were assessed weekly until the 28th day using a combined behavioral score (CBS). Total antioxidant capacity (TAC), malondialdehyde (MDA), superoxide dismutase (SOD), and glutathione peroxidase (GPx) were measured in spinal tissue to evaluate oxidative stress-related parameters. Besides, autophagy-related proteins (P62, LC3B, and Beclin1) and apoptosis-associated proteins (Bax and Bcl2) were determined using western blotting on the 1st and 7th days after surgery. Hematoxylin-eosin and Fluoro-Jade B staining were performed to detect the histological alterations and neuronal degeneration. As the result, treatment with AST potentially attenuated rat CBS scores (p < 0.001) towards a better motor performance. AST significantly reduced the spinal level of oxidative stress by increasing TAC, SOD, and GPx, while decreasing MDA (p < 0.001). Furthermore, AST treatment remarkably upregulated expression of LC3B (p < 0.001), and Beclin1 (p < 0.05) in the spinal cord, but downregulated P62 (p < 0.05) and the Bax/Bcl2 ratio (p < 0.001). Consequently, AST reduced SCI-induced histological alterations and neuronal degeneration (p < 0.001). In conclusion, AST can improve motor function after SCI by reducing oxidative stress/apoptosis and increasing neuronal autophagy.
Collapse
Affiliation(s)
- Fatemeh Abbaszadeh
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Science, Tehran, Iran
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoumeh Jorjani
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohammad Taghi Joghataei
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Science, Tehran, Iran.
| | - Soraya Mehrabi
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Science, Tehran, Iran
| |
Collapse
|
14
|
Improper Proteostasis: Can It Serve as Biomarkers for Neurodegenerative Diseases? Mol Neurobiol 2022; 59:3382-3401. [PMID: 35305242 DOI: 10.1007/s12035-022-02775-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 02/19/2022] [Indexed: 10/18/2022]
Abstract
Cells synthesize new proteins after multiple molecular decisions. Damage of existing proteins, accumulation of abnormal proteins, and basic requirement of new proteins trigger protein quality control (PQC)-based alternative strategies to cope against proteostasis imbalance. Accumulation of misfolded proteins is linked with various neurodegenerative disorders. However, how deregulated components of this quality control system and their lack of general mechanism-based long-term changes can serve as biomarkers for neurodegeneration remains largely unexplored. Here, our article summarizes the chief findings, which may facilitate the search of novel and relevant proteostasis mechanism-based biomarkers associated with neuronal disorders. Understanding the abnormalities of PQC coupled molecules as possible biomarkers can help to determine neuronal fate and their contribution to the aetiology of several nervous system disorders.
Collapse
|
15
|
Hou W, Hao Y, Sun L, Zhao Y, Zheng X, Song L. The dual roles of autophagy and the GPCRs-mediating autophagy signaling pathway after cerebral ischemic stroke. Mol Brain 2022; 15:14. [PMID: 35109896 PMCID: PMC8812204 DOI: 10.1186/s13041-022-00899-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 01/20/2022] [Indexed: 12/17/2022] Open
Abstract
Ischemic stroke, caused by a lack of blood supply in brain tissues, is the third leading cause of human death and disability worldwide, and usually results in sensory and motor dysfunction, cognitive impairment, and in severe cases, even death. Autophagy is a highly conserved lysosome-dependent process in which eukaryotic cells removal misfolded proteins and damaged organelles in cytoplasm, which is critical for energy metabolism, organelle renewal, and maintenance of intracellular homeostasis. Increasing evidence suggests that autophagy plays important roles in pathophysiological mechanisms under ischemic conditions. However, there are still controversies about whether autophagy plays a neuroprotective or damaging role after ischemia. G-protein-coupled receptors (GPCRs), one of the largest protein receptor superfamilies in mammals, play crucial roles in various physiological and pathological processes. Statistics show that GPCRs are the targets of about one-fifth of drugs known in the world, predicting potential values as targets for drug research. Studies have demonstrated that nutritional deprivation can directly or indirectly activate GPCRs, mediating a series of downstream biological processes, including autophagy. It can be concluded that there are interactions between autophagy and GPCRs signaling pathway, which provides research evidence for regulating GPCRs-mediated autophagy. This review aims to systematically discuss the underlying mechanism and dual roles of autophagy in cerebral ischemia, and describe the GPCRs-mediated autophagy, hoping to probe promising therapeutic targets for ischemic stroke through in-depth exploration of the GPCRs-mediated autophagy signaling pathway.
Collapse
Affiliation(s)
- Weichen Hou
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China
| | - Yulei Hao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China
| | - Li Sun
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China
| | - Yang Zhao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China
| | - Xiangyu Zheng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China.
| | - Lei Song
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China.
| |
Collapse
|
16
|
Liao HY, Wang ZQ, Ran R, Zhou KS, Ma CW, Zhang HH. Biological Functions and Therapeutic Potential of Autophagy in Spinal Cord Injury. Front Cell Dev Biol 2022; 9:761273. [PMID: 34988074 PMCID: PMC8721099 DOI: 10.3389/fcell.2021.761273] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 11/24/2021] [Indexed: 12/12/2022] Open
Abstract
Autophagy is an evolutionarily conserved lysosomal degradation pathway that maintains metabolism and homeostasis by eliminating protein aggregates and damaged organelles. Many studies have reported that autophagy plays an important role in spinal cord injury (SCI). However, the spatiotemporal patterns of autophagy activation after traumatic SCI are contradictory. Most studies show that the activation of autophagy and inhibition of apoptosis have neuroprotective effects on traumatic SCI. However, reports demonstrate that autophagy is strongly associated with distal neuronal death and the impaired functional recovery following traumatic SCI. This article introduces SCI pathophysiology, the physiology and mechanism of autophagy, and our current review on its role in traumatic SCI. We also discuss the interaction between autophagy and apoptosis and the therapeutic effect of activating or inhibiting autophagy in promoting functional recovery. Thus, we aim to provide a theoretical basis for the biological therapy of SCI.
Collapse
Affiliation(s)
- Hai-Yang Liao
- Lanzhou University Second Hospital, Lanzhou, China.,Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
| | - Zhi-Qiang Wang
- Lanzhou University Second Hospital, Lanzhou, China.,Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
| | - Rui Ran
- Lanzhou University Second Hospital, Lanzhou, China.,Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
| | - Kai-Sheng Zhou
- Lanzhou University Second Hospital, Lanzhou, China.,Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
| | - Chun-Wei Ma
- Lanzhou University Second Hospital, Lanzhou, China.,Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
| | - Hai-Hong Zhang
- Lanzhou University Second Hospital, Lanzhou, China.,Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
17
|
Nie BX, Zhao G, Yuan XF, Yu LX, Zhang J, Yuan Y, Liu Y, Hu J, Song E, Zhou YC, Shu J. Inhibition of CDK1 attenuates neuronal apoptosis and autophagy and confers neuroprotection after chronic spinal cord injury in vivo. J Chem Neuroanat 2021; 119:102053. [PMID: 34839004 DOI: 10.1016/j.jchemneu.2021.102053] [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: 08/15/2021] [Revised: 11/18/2021] [Accepted: 11/21/2021] [Indexed: 10/19/2022]
Abstract
Chronic spinal cord injury (CSCI) results from progressive compression of the spinal cord over time. A variety of factors cause CSCI, and its exact pathogenesis is unknown. Cyclin-dependent kinase 1 (CDK1) is closely related to the apoptosis pathway, but no CSCI-related studies on CDK1 have been conducted. In this study, the role of CDK1 in CSCI was explored in a rat model. The CSCI model was established by screw compression using the cervical anterior approach for twelve weeks. The neurological function of the rats was evaluated using the neurological severity scores (NSS) and motor evoked potentials (MEPs). Pathological changes in spinal cord tissue were observed by hematoxylin-eosin (HE) staining, and Nissl staining was performed to assess the survival of motor neurons in the anterior horn of the spinal cord. Changes in autophagy and apoptosis in anterior horn of spinal cord tissue were detected using transmission electron microscopy and the terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay, respectively. The expression levels of glial fibrillary acidic protein (GFAP), ionized calcium-binding adaptor (IBA) and choline acetyltransferase (CHAT) in the anterior horn were determined using immunohistochemistry assays to investigate astrocytes, microglia and motor neurons, respectively, in the anterior horn. Western blot assays were used to detect the expression levels of CDK1, Bcl-2, Bax, Caspase 3, LC3 and Beclin1. Changes in the expression of CDK1, LC3 and Beclin1 were also observed using immunohistochemistry. The results indicated that CSCI resulted in neuronal injury and a decrease in the NSS. In the CSCI model group, anterior horn astrocytes and microglia were activated, and motor neurons were decreased. Neuronal apoptosis was promoted, and the number of autophagic vacuoles was elevated. Rats treated with the CDK1 shRNA lentivirus exhibited better NSS, more surviving motor neurons, and fewer apoptotic neurons than the model rats. The occurrence of autophagy and the expression of proapoptotic and autophagy-related proteins were lower in the CDK1 shRNA group than the model group. In conclusion, CDK1 downregulation suppressed the activation of anterior horn astrocytes and microglia, promoted motor neuron repair, and inhibited neurons apoptosis and autophagy to promote the recovery of motor function after spinal cord injury.
Collapse
Affiliation(s)
- Bang-Xu Nie
- Traumatology Surgery, Second Affiliated Hospital of Kunming Medical University, Kunming 650106, Yunnan, China
| | - Gang Zhao
- Traumatology Surgery, Second Affiliated Hospital of Kunming Medical University, Kunming 650106, Yunnan, China
| | - Xiao-Feng Yuan
- Department of Orthopedics, Affiliated Calmette Hospital of Kunming Medical University, Kunming 650224, Yunnan, China
| | - Lin-Xin Yu
- Department of Orthopedics, Affiliated Calmette Hospital of Kunming Medical University, Kunming 650224, Yunnan, China
| | - Jin Zhang
- Department of Orthopedics, Affiliated Calmette Hospital of Kunming Medical University, Kunming 650224, Yunnan, China
| | - Yong Yuan
- Traumatology Surgery, Second Affiliated Hospital of Kunming Medical University, Kunming 650106, Yunnan, China
| | - Yao Liu
- College of Rehabilitation, Kunming Medical University, Kunming 650504, Yunnan, China
| | - Jun Hu
- Department of Orthopedics, Affiliated Calmette Hospital of Kunming Medical University, Kunming 650224, Yunnan, China
| | - En Song
- Department of Sports Medicine, First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Yu-Cheng Zhou
- Department of Orthopedics, Yunnan Provincial Rehabilitation Center for the Disabled Persons, Kunming 650034, Yunnan, China
| | - Jun Shu
- Traumatology Surgery, Second Affiliated Hospital of Kunming Medical University, Kunming 650106, Yunnan, China.
| |
Collapse
|
18
|
Lu Q, Zhou Y, Xu M, Liang X, Jing H, Wang X, Li N. Sequential delivery for hepatic fibrosis treatment based on carvedilol loaded star-like nanozyme. J Control Release 2021; 341:247-260. [PMID: 34826531 DOI: 10.1016/j.jconrel.2021.11.033] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 11/17/2021] [Accepted: 11/19/2021] [Indexed: 11/30/2022]
Abstract
Hepatic fibrosis, characterized by excessive reactive oxygen species (ROS) generation, hepatic stellate cells (HSCs) activation, and enormous extracellular matrix (ECM) production, can further cause liver cirrhosis, liver failure and liver cancer. However, the combination of limited solubility, low targeting, uncontrolled release and the sophisticated physiological barriers are tremendous challenges for therapeutic effect. In this study, we engineered a sequential delivery strategy based on autophagy inhibitor carvedilol (CAR) loaded and hyaluronic acid (HA) modified star-like Au nanozyme (Au NS@CAR-HA) for targeted HSCs suppression. In hepatic fibrosis acidic environment, CAR-HA can be firstly detached from Au NS@CAR-HA. Then, CAR would be released from CAR-HA conjugation by chemical bond breakage which triggered by intracellular acid potential, thus could suppressing autolysosome generation by up-regulation of autosome and lysosome pH value to inhibit HSCs activation. Meanwhile, Au NS exhibited enhanced ROS scavenging efficiency of hydrogen peroxides and superoxide, which was helpful to restrain the activity of peroxisome proliferators-activated receptors β (PPARβ) and c-Jun N-terminal kinase (JNK), thereby reducing HSCs proliferation to enhance HSCs inactivation efficacy. In conclusion, Au NS@CAR-HA can attenuate hepatic fibrosis via regulating the proliferation and activation of hepatic stellate cells, which provides a new strategy for hepatic fibrosis treatment.
Collapse
Affiliation(s)
- Qiang Lu
- Tianjin Key Laboratory of Drug Delivery & High Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, PR China
| | - Yue Zhou
- Tianjin Key Laboratory of Drug Delivery & High Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, PR China
| | - Min Xu
- Tianjin Key Laboratory of Drug Delivery & High Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, PR China
| | - Xiaoyang Liang
- Tianjin Key Laboratory of Drug Delivery & High Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, PR China
| | - Huaqing Jing
- Tianjin Key Laboratory of Drug Delivery & High Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, PR China
| | - Xinxing Wang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, PR China.
| | - Nan Li
- Tianjin Key Laboratory of Drug Delivery & High Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, PR China.
| |
Collapse
|
19
|
Claude-Taupin A, Codogno P, Dupont N. Links between autophagy and tissue mechanics. J Cell Sci 2021; 134:271984. [PMID: 34472605 DOI: 10.1242/jcs.258589] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Physical constraints, such as compression, shear stress, stretching and tension, play major roles during development, tissue homeostasis, immune responses and pathologies. Cells and organelles also face mechanical forces during migration and extravasation, and investigations into how mechanical forces are translated into a wide panel of biological responses, including changes in cell morphology, membrane transport, metabolism, energy production and gene expression, is a flourishing field. Recent studies demonstrate the role of macroautophagy in the integration of physical constraints. The aim of this Review is to summarize and discuss our knowledge of the role of macroautophagy in controlling a large panel of cell responses, from morphological and metabolic changes, to inflammation and senescence, for the integration of mechanical forces. Moreover, wherever possible, we also discuss the cell surface molecules and structures that sense mechanical forces upstream of macroautophagy.
Collapse
Affiliation(s)
- Aurore Claude-Taupin
- Institut Necker-Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, Université de Paris, 75015 Paris, France
| | - Patrice Codogno
- Institut Necker-Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, Université de Paris, 75015 Paris, France
| | - Nicolas Dupont
- Institut Necker-Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, Université de Paris, 75015 Paris, France
| |
Collapse
|
20
|
Beckers A, Vanhunsel S, Van Dyck A, Bergmans S, Masin L, Moons L. Injury-induced Autophagy Delays Axonal Regeneration after Optic Nerve Damage in Adult Zebrafish. Neuroscience 2021; 470:52-69. [PMID: 34280491 DOI: 10.1016/j.neuroscience.2021.07.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/18/2021] [Accepted: 07/09/2021] [Indexed: 12/15/2022]
Abstract
Optic neuropathies comprise a group of disorders in which the axons of retinal ganglion cells (RGCs), the retinal projection neurons conveying visual information to the brain, are damaged. This results in visual impairment or even blindness, which is irreversible as adult mammals lack the capacity to repair or replace injured or lost neurons. Despite intensive research, no efficient treatment to induce axonal regeneration in the central nervous system (CNS) is available yet. Autophagy, the cellular recycling response, was shown repeatedly to be elevated in animal models of optic nerve injury, and both beneficial and detrimental effects have been reported. In this study, we subjected spontaneously regenerating adult zebrafish to optic nerve damage (ONC) and revealed that autophagy is enhanced after optic nerve damage in zebrafish, both in RGC axons and somas, as well as in macroglial cells of the retina, the optic nerve and the visual target areas in the brain. Interestingly, the pattern of the autophagic response in the axons followed the spatiotemporal window of axonal regrowth, which suggests that autophagy is ongoing at the growth cones. Pharmacological inhibition of the recycling pathway resulted in accelerated RGC target reinnervation, possibly linked to increased mechanistic target of rapamycin (mTOR) activity, known to stimulate axonal regrowth. Taken together, these intriguing findings underline that further research is warranted to decipher if modulation of autophagy could be an effective therapeutic method to induce CNS regeneration.
Collapse
Affiliation(s)
- An Beckers
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, 3000 Leuven, Belgium
| | - Sophie Vanhunsel
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, 3000 Leuven, Belgium
| | - Annelies Van Dyck
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, 3000 Leuven, Belgium
| | - Steven Bergmans
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, 3000 Leuven, Belgium
| | - Luca Masin
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, 3000 Leuven, Belgium
| | - Lieve Moons
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Section, Department of Biology, KU Leuven, 3000 Leuven, Belgium; Leuven Brain Institute, 3000 Leuven, Belgium.
| |
Collapse
|
21
|
Trehalose Augments Neuron Survival and Improves Recovery from Spinal Cord Injury via mTOR-Independent Activation of Autophagy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8898996. [PMID: 34336117 PMCID: PMC8289614 DOI: 10.1155/2021/8898996] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 06/08/2021] [Indexed: 01/11/2023]
Abstract
Spinal cord injury (SCI) is a major cause of irreversible nerve injury and leads to serious tissue loss and neurological dysfunction. Thorough investigation of cellular mechanisms, such as autophagy, is crucial for developing novel and effective therapeutics. We administered trehalose, an mTOR-independent autophagy agonist, in SCI rats suffering from moderate compression injury to elucidate the relationship between autophagy and SCI and evaluate trehalose's therapeutic potential. 60 rats were divided into 4 groups and were treated with either control vehicle, trehalose, chloroquine, or trehalose + chloroquine 2 weeks prior to administration of moderate spinal cord crush injury. 20 additional sham rats were treated with control vehicle. H&E staining, Nissl staining, western blot, and immunofluorescence studies were conducted to examine nerve morphology and quantify autophagy and mitochondrial-dependent apoptosis at various time points after surgery. Functional recovery was assessed over a period of 4 weeks after surgery. Trehalose promotes autophagosome recruitment via an mTOR-independent pathway, enhances autophagy flux in neurons, inhibits apoptosis via the intrinsic mitochondria-dependent pathway, reduces lesion cavity expansion, decreases neuron loss, and ultimately improves functional recovery following SCI (all p < 0.05). Furthermore, these effects were diminished upon administration of chloroquine, an autophagy flux inhibitor, indicating that trehalose's beneficial effects were due largely to activation of autophagy. This study presents new evidence that autophagy plays a critical neuroprotective and neuroregenerative role in SCI, and that mTOR-independent activation of autophagy with trehalose leads to improved outcomes. Thus, trehalose has great translational potential as a novel therapeutic agent after SCI.
Collapse
|
22
|
Gu Y, Chen D, Zhou L, Zhao X, Lin J, Lin B, Lin T, Chen Z, Chen Z, Wang Z, Liu W. Lysine-specific demethylase 1 inhibition enhances autophagy and attenuates early-stage post-spinal cord injury apoptosis. Cell Death Discov 2021; 7:69. [PMID: 33824301 PMCID: PMC8024310 DOI: 10.1038/s41420-021-00455-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 03/04/2021] [Accepted: 03/18/2021] [Indexed: 11/23/2022] Open
Abstract
Neuron death in spinal cords is caused primarily by apoptosis after spinal cord injury (SCI). Autophagy can act as a cellular response to maintain neuron homeostasis that can reduce apoptosis. Although more studies have shown that an epigenetic enzyme called Lysine-specific demethylase 1 (LSD1) can negatively regulate autophagy during cancer research, existing research does not focus on impacts related to LSD1 in nerve injury diseases. This study was designed to determine whether inhibiting LSD1 could enhance autophagy against apoptosis and provide effective neuroprotection in vitro and vivo after SCI. The results showed that LSD1 inhibition treatment significantly reduced spinal cord damage in SCI rat models and was characterized by upregulated autophagy and downregulated apoptosis. Further research demonstrated that using both pharmacological inhibition and gene knockdown could enhance autophagy and reduce apoptosis for in vitro simulation of SCI-caused damage models. Additionally, 3-methyladenine (3-MA) could partially eliminate the effect of autophagy enhancement and apoptosis suppression. These findings demonstrated that LSD1 inhibition could protect against SCI by activating autophagy and hindering apoptosis, suggesting a potential candidate for SCI therapy.
Collapse
Affiliation(s)
- Yang Gu
- Department of Orthopedic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Dehui Chen
- Department of Orthopedic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Linquan Zhou
- Department of Orthopedic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Xin Zhao
- School of Health, Fujian Medical University, Fuzhou, 350108, Fujian, China
| | - Jiemin Lin
- School of Health, Fujian Medical University, Fuzhou, 350108, Fujian, China
| | - Bin Lin
- Department of Orthopedic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Taotao Lin
- Department of Orthopedic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Zhi Chen
- Department of Orthopedic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Zhaohong Chen
- Wound Repair Department, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China.,Fujian Provincial Key Laboratory of Burn and Trauma, Fuzhou, 350001, Fujian, China
| | - Zhenyu Wang
- Department of Orthopedic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China.
| | - Wenge Liu
- Department of Orthopedic Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China.
| |
Collapse
|
23
|
Tu J, Vargas Castillo J, Das A, Diwan AD. Degenerative Cervical Myelopathy: Insights into Its Pathobiology and Molecular Mechanisms. J Clin Med 2021; 10:jcm10061214. [PMID: 33804008 PMCID: PMC8001572 DOI: 10.3390/jcm10061214] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/07/2021] [Accepted: 03/09/2021] [Indexed: 12/12/2022] Open
Abstract
Degenerative cervical myelopathy (DCM), earlier referred to as cervical spondylotic myelopathy (CSM), is the most common and serious neurological disorder in the elderly population caused by chronic progressive compression or irritation of the spinal cord in the neck. The clinical features of DCM include localised neck pain and functional impairment of motor function in the arms, fingers and hands. If left untreated, this can lead to significant and permanent nerve damage including paralysis and death. Despite recent advancements in understanding the DCM pathology, prognosis remains poor and little is known about the molecular mechanisms underlying its pathogenesis. Moreover, there is scant evidence for the best treatment suitable for DCM patients. Decompressive surgery remains the most effective long-term treatment for this pathology, although the decision of when to perform such a procedure remains challenging. Given the fact that the aged population in the world is continuously increasing, DCM is posing a formidable challenge that needs urgent attention. Here, in this comprehensive review, we discuss the current knowledge of DCM pathology, including epidemiology, diagnosis, natural history, pathophysiology, risk factors, molecular features and treatment options. In addition to describing different scoring and classification systems used by clinicians in diagnosing DCM, we also highlight how advanced imaging techniques are being used to study the disease process. Last but not the least, we discuss several molecular underpinnings of DCM aetiology, including the cells involved and the pathways and molecules that are hallmarks of this disease.
Collapse
Affiliation(s)
- Ji Tu
- Spine Labs, St. George and Sutherland Clinical School, University of New South Wales, Kogarah, NSW 2217, Australia; (J.T.); (A.D.D.)
| | | | - Abhirup Das
- Spine Labs, St. George and Sutherland Clinical School, University of New South Wales, Kogarah, NSW 2217, Australia; (J.T.); (A.D.D.)
- Spine Service, St. George Hospital, Kogarah, NSW 2217, Australia;
- Correspondence:
| | - Ashish D. Diwan
- Spine Labs, St. George and Sutherland Clinical School, University of New South Wales, Kogarah, NSW 2217, Australia; (J.T.); (A.D.D.)
- Spine Service, St. George Hospital, Kogarah, NSW 2217, Australia;
| |
Collapse
|
24
|
Cheng X, Yu Z, Xu J, Quan D, Long H. Pathophysiological Changes and the Role of Notch-1 Activation After Decompression in a Compressive Spinal Cord Injury Rat Model. Front Neurosci 2021; 15:579431. [PMID: 33584186 PMCID: PMC7876297 DOI: 10.3389/fnins.2021.579431] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 01/11/2021] [Indexed: 12/13/2022] Open
Abstract
Surgical decompression is the primary treatment for cervical spondylotic myelopathy (CSM) patients with compressive spinal cord injury (CSCI). However, the prognosis of patients with CSCI varies, and the pathophysiological changes following decompression remain poor. This study aimed to investigate the pathophysiological changes and the role of Notch-1 activation after decompression in a rat CSCI model. Surgical decompression was conducted at 1 week post-injury (wpi). DAPT was intraperitoneally injected to down-regulate Notch-1 expression. Basso, Beattie, and Bresnahan scores and an inclined plane test were used to evaluate the motor function recovery. Hematoxylin and eosin staining was performed to assess pathophysiological changes, while hypoxia-inducible factor 1 alpha, vascular endothelial growth factor (VEGF), von Willebrand factor (vWF), matrix metalloproteinase (MMP)-9, MMP-2, Notch-1, and Hes-1 expression in the spinal cord were examined by immunohistochemical analysis or quantitative PCR. The results show that early decompression can partially promote motor function recovery. Improvements in structural and cellular damage and hypoxic levels were also observed in the decompressed spinal cord. Moreover, decompression resulted in increased VEGF and vWF expression, but decreased MMP-9 and MMP-2 expression at 3 wpi. Expression levels of Notch-1 and its downstream gene Hes-1 were increased after decompression, and the inhibition of Notch-1 significantly reduced the decompression-induced motor function recovery. This exploratory study revealed preliminary pathophysiological changes in the compressed and decompressed rat spinal cord. Furthermore, we confirmed that early surgical decompression partially promotes motor function recovery may via activation of the Notch-1 signaling pathway after CSCI. These results could provide new insights for the development of drug therapy to enhance recovery following surgery.
Collapse
Affiliation(s)
- Xing Cheng
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhengran Yu
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jinghui Xu
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Daping Quan
- PCFM Lab, GD HPPC Lab, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, China
| | - Houqing Long
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
25
|
Cai Y, Li X, Pan Z, Zhu Y, Tuo J, Meng Q, Dai G, Yang G, Pan Y. Anthocyanin ameliorates hypoxia and ischemia induced inflammation and apoptosis by increasing autophagic flux in SH-SY5Y cells. Eur J Pharmacol 2020; 883:173360. [DOI: 10.1016/j.ejphar.2020.173360] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 07/05/2020] [Accepted: 07/08/2020] [Indexed: 01/21/2023]
|
26
|
Role of ABT888, a Novel Poly(ADP-Ribose) Polymerase (PARP) Inhibitor in Countering Autophagy and Apoptotic Processes Associated to Spinal Cord Injury. Mol Neurobiol 2020; 57:4394-4407. [PMID: 32729104 DOI: 10.1007/s12035-020-02033-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/22/2020] [Indexed: 02/04/2023]
Abstract
Accidents are the cause of some 50 deaths per 100,000 population each year; some 3% of these are from traumatic spinal cord injury (SCI), a damage that causes temporary or permanent motor deficits, often leading to permanent neurological alterations. The activation of poly(ADP-ribose) polymerase (PARP) as DNA damage response, together with autophagy and apoptosis processes contributes to the secondary injury processes seen after SCI. Thus, in the present study, a mouse compression model of SCI was used to determine whether the treatment with ABT888, as PARP-1/2 inhibitor, could restore the neuronal damage induced by SCI. Mice were orally administered with ABT888 (at a dose of 25 mg/kg) 1 h and 6 h after SCI induction. Histological analysis, myeloperoxidase (MPO) activity, and Basso Mouse scale (BMS) were performed. The expression of autophagy-related proteins and apoptosis-inducing factors was quantified in the cytosolic fraction from spinal cord tissue collected after 24 h after SCI. TUNEL assay was performed in SCI-tissues 24 h after damage. ABT888 treatment significantly reduced histological damage and neutrophilic infiltration, improving motor skills. PARP-1/2 inhibition by ABT888 slowed cell death, decreasing autophagy-activation proteins. These results showed that ABT888, inhibiting PARP-1/2 activity, through a reduction in the apoptosis-autophagy machinery, plays a protective role after SCI, suggesting a new insight into the potential application of ABT888 as novel candidate in SCI therapies.
Collapse
|
27
|
Chen G, Li J, Wang Z, Liu W. Ezetimibe protects against spinal cord injury by regulating autophagy and apoptosis through inactivation of PI3K/AKT/mTOR signaling. Am J Transl Res 2020; 12:2685-2694. [PMID: 32655800 PMCID: PMC7344056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 05/25/2020] [Indexed: 06/11/2023]
Abstract
Spinal cord injury (SCI) is a severe traumatic disease of the central nervous system characterized by high incidence and disability rate. We aimed to investigate the therapeutic potential of Ezetimibe (Eze) in SCI and identify the underlying mechanisms. Acute SCI rat model was established by using the modified weight-drop method. Following administration with Eze, the neurological function was evaluated using the Basso, Beattie, and Bresnahan (BBB) locomotor scale score, and the motor neurons were stained with Nissl staining. The pathological changes of spinal cord tissues were tested using Hematoxylin and eosin staining. The presence of apoptotic cells was examined using Terminal dexynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) staining. Moreover, the expression of main autophagy markers LC3II/I, Beclin1 and p62 and apoptosis-related proteins was tested using western blot analysis. The changes of PI3K/AKT/mTOR signaling-associated proteins were measured. Experimental results showed that Eze treatment obviously improved functional recovery, the neuronal survival and morphological characteristics of spinal cord. Additionally, Eze administration dramatically upregulated the expression of LC3II/I and Beclin1 whereas downregulated that of p62. Concurrently, significantly reduced apoptosis was observed following Eze intervention, accompanied by increased expression of anti-apoptotic protein Bcl-2 and decreased expression of pro-apoptotic proteins Bax, cleaved caspase-3 and cleaved caspase-9. Further results indicated that Eze treatment remarkably suppressed the expression of phospho-PI3K (p-PI3K), p-AKT and p-mTOR. These findings demonstrated that Eze could protect against SCI by activating autophagy and hindering apoptosis through regulating PI3K/AKT/mTOR signaling, suggesting a potential candidate for SCI therapy.
Collapse
Affiliation(s)
- Gang Chen
- Orthopedics Department, Union Hospital Affiliated to Fujian Medical University Fuzhou 350000, Fujian Province, China
| | - Jiandong Li
- Orthopedics Department, Union Hospital Affiliated to Fujian Medical University Fuzhou 350000, Fujian Province, China
| | - Zhenyu Wang
- Orthopedics Department, Union Hospital Affiliated to Fujian Medical University Fuzhou 350000, Fujian Province, China
| | - Wenge Liu
- Orthopedics Department, Union Hospital Affiliated to Fujian Medical University Fuzhou 350000, Fujian Province, China
| |
Collapse
|
28
|
Akter F, Yu X, Qin X, Yao S, Nikrouz P, Syed YA, Kotter M. The Pathophysiology of Degenerative Cervical Myelopathy and the Physiology of Recovery Following Decompression. Front Neurosci 2020; 14:138. [PMID: 32425740 PMCID: PMC7203415 DOI: 10.3389/fnins.2020.00138] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 02/03/2020] [Indexed: 12/15/2022] Open
Abstract
Background: Degenerative cervical myelopathy (DCM), also known as cervical spondylotic myelopathy is the leading cause of spinal cord compression in adults. The mainstay of treatment is surgical decompression, which leads to partial recovery of symptoms, however, long term prognosis of the condition remains poor. Despite advances in treatment methods, the underlying pathobiology is not well-known. A better understanding of the disease is therefore required for the development of treatments to improve outcomes following surgery. Objective: To systematically evaluate the pathophysiology of DCM and the mechanism underlying recovery following decompression. Methods: A total of 13,808 published articles were identified in our systematic search of electronic databases (PUBMED, WEB OF SCIENCE). A total of 51 studies investigating the secondary injury mechanisms of DCM or physiology of recovery in animal models of disease underwent comprehensive review. Results: Forty-seven studies addressed the pathophysiology of DCM. Majority of the studies demonstrated evidence of neuronal loss following spinal cord compression. A number of studies provided further details of structural changes in neurons such as myelin damage and axon degeneration. The mechanisms of injury to cells included direct apoptosis and increased inflammation. Only four papers investigated the pathobiological changes that occur in spinal cords following decompression. One study demonstrated evidence of axonal plasticity following decompressive surgery. Another study demonstrated ischaemic-reperfusion injury following decompression, however this phenomenon was worse when decompression was delayed. Conclusions: In preclinical studies, the pathophysiology of DCM has been poorly studied and a number of questions remain unanswered. The physiological changes seen in the decompressed spinal cord has not been widely investigated and it is paramount that researchers investigate the decompressed spinal cord further to enable the development of therapeutic tools, to enhance recovery following surgery.
Collapse
Affiliation(s)
- Farhana Akter
- Department of Clinical Neuroscience, University of Cambridge, Cambridge, United Kingdom.,Faculty of Arts and Sciences, Harvard University, Cambridge, MA, United States.,Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, United States
| | - Xinming Yu
- Department of Clinical Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Xingping Qin
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, United States
| | - Shun Yao
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Parisa Nikrouz
- Maidstone and Tunbridge Wells Trust, Maidstone, United Kingdom
| | - Yasir Ahmed Syed
- Neuroscience and Mental Health Research Institute (NMHRI), Cathays, United Kingdom.,School of Bioscience, Cardiff University, The Sir Martin Evans Building, Cardiff, United Kingdom
| | - Mark Kotter
- Department of Clinical Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
29
|
Memmert S, Nogueira AVB, Damanaki A, Nokhbehsaim M, Rath-Deschner B, Götz W, Gölz L, Cirelli JA, Till A, Jäger A, Deschner J. Regulation of the autophagy-marker Sequestosome 1 in periodontal cells and tissues by biomechanical loading. J Orofac Orthop 2019; 81:10-21. [PMID: 31591651 DOI: 10.1007/s00056-019-00197-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 08/20/2019] [Indexed: 01/24/2023]
Abstract
PURPOSE Orthodontic treatment is based on the principle of force application to teeth and subsequently to the surrounding tissues and periodontal cells. Sequestosome 1 (SQSTM1) is a well-known marker for autophagy, which is an important cellular mechanism of adaptation to stress. The aim of this study was to analyze whether biomechanical loading conditions regulate SQSTM1 in periodontal cells and tissues, thereby providing further information on the role of autophagy in orthodontic tooth movement. METHODS Periodontal ligament (PDL) fibroblasts were exposed to cyclic tensile strain of low magnitude (3%, CTSL), and the regulation of autophagy-associated targets was determined with an array-based approach. SQSTM1 was selected for further biomechanical loading experiments with dynamic and static tensile strain and assessed via real-time polymerase chain reaction (RT-PCR) and immunoblotting. Signaling pathways involved in SQSTM1 activation were analyzed by using specific inhibitors, including an autophagy inhibitor. Finally, SQSTM1 expression was analyzed in gingival biopsies and histological sections of rats in presence and absence of orthodontic forces. RESULTS Multiple autophagy-associated targets were regulated by CTSL in PDL fibroblasts. All biomechanical loading conditions tested increased the SQSTM1 expression significantly. Stimulatory effects of CTSL on SQSTM1 expression were diminished by inhibition of the c‑Jun N‑terminal kinase (JNK) pathway and of autophagy. Increased SQSTM1 levels after CTSL were confirmed by immunoblotting. Orthodontic force application also led to significantly elevated SQTSM1 levels in the gingiva and PDL of treated animals as compared to control. CONCLUSIONS Our in vitro and in vivo findings provide evidence of a role of SQSTM1 and thereby autophagy in orthodontic tooth movement.
Collapse
Affiliation(s)
- S Memmert
- Department of Orthodontics, Center of Dento-Maxillo-Facial Medicine, University of Bonn, Welschnonnenstr. 17, 53111, Bonn, Germany. .,Section of Experimental Dento-Maxillo-Facial Medicine, Center of Dento-Maxillo-Facial Medicine, University of Bonn, Bonn, Germany.
| | - A V B Nogueira
- Department of Periodontology and Operative Dentistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - A Damanaki
- Department of Periodontology and Operative Dentistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - M Nokhbehsaim
- Section of Experimental Dento-Maxillo-Facial Medicine, Center of Dento-Maxillo-Facial Medicine, University of Bonn, Bonn, Germany
| | - B Rath-Deschner
- Department of Orthodontics, Center of Dento-Maxillo-Facial Medicine, University of Bonn, Welschnonnenstr. 17, 53111, Bonn, Germany
| | - W Götz
- Department of Orthodontics, Center of Dento-Maxillo-Facial Medicine, University of Bonn, Welschnonnenstr. 17, 53111, Bonn, Germany
| | - L Gölz
- Department of Orthodontics and Orofacial Orthopedics, University of Erlangen, Erlangen, Germany.,Institute of Human Genetics, University Hospital of Bonn, Bonn, Germany
| | - J A Cirelli
- Department of Diagnosis and Surgery, School of Dentistry at Araraquara, University Estadual Paulista-UNESP, Araraquara, Brazil
| | - A Till
- Institute of Reconstructive Neurobiology, Life and Brain Center, University of Bonn, Bonn, Germany
| | - A Jäger
- Department of Orthodontics, Center of Dento-Maxillo-Facial Medicine, University of Bonn, Welschnonnenstr. 17, 53111, Bonn, Germany
| | - J Deschner
- Department of Periodontology and Operative Dentistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
30
|
Wu J, Lipinski MM. Autophagy in Neurotrauma: Good, Bad, or Dysregulated. Cells 2019; 8:E693. [PMID: 31295858 PMCID: PMC6678153 DOI: 10.3390/cells8070693] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/06/2019] [Accepted: 07/09/2019] [Indexed: 12/12/2022] Open
Abstract
Autophagy is a physiological process that helps maintain a balance between the manufacture of cellular components and breakdown of damaged organelles and other toxic cellular constituents. Changes in autophagic markers are readily detectable in the spinal cord and brain following neurotrauma, including traumatic spinal cord and brain injury (SCI/TBI). However, the role of autophagy in neurotrauma remains less clear. Whether autophagy is good or bad is under debate, with strong support for both a beneficial and detrimental role for autophagy in experimental models of neurotrauma. Emerging data suggest that autophagic flux, a measure of autophagic degradation activity, is impaired in injured central nervous systems (CNS), and interventions that stimulate autophagic flux may provide neuroprotection in SCI/TBI models. Recent data demonstrating that neurotrauma can cause lysosomal membrane damage resulting in pathological autophagosome accumulation in the spinal cord and brain further supports the idea that the impairment of the autophagy-lysosome pathway may be a part of secondary injury processes of SCI/TBI. Here, we review experimental work on the complex and varied responses of autophagy in terms of both the beneficial and detrimental effects in SCI and TBI models. We also discuss the existing and developing therapeutic options aimed at reducing the disruption of autophagy to protect the CNS after injuries.
Collapse
Affiliation(s)
- Junfang Wu
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA.
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
- Center to Advance Chronic Pain Research, University of Maryland, Baltimore, MD 21201, USA.
| | - Marta M Lipinski
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
31
|
Wang G, Xue Y, Wang Y, Dong F, Shen M, Zong R, Liu Z, Li C. The role of autophagy in the pathogenesis of exposure keratitis. J Cell Mol Med 2019; 23:4217-4228. [PMID: 30973208 PMCID: PMC6533494 DOI: 10.1111/jcmm.14310] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 03/03/2019] [Accepted: 03/08/2019] [Indexed: 12/13/2022] Open
Abstract
Incomplete tear film spreading and eyelid closure can cause defective renewal of the ocular surface and air exposure-induced epithelial keratopathy (EK). In this study, we characterized the role of autophagy in mediating the ocular surface changes leading to EK. Human corneal epithelial cells (HCECs) and C57BL/6 mice were employed as EK models, respectively. Transmission electron microscopy (TEM) evaluated changes in HCECs after air exposure. Each of these models was treated with either an autophagy inhibitor [chloroquine (CQ) or 3-methyladenine (3-MA)] or activator [Rapamycin (Rapa)]. Immunohistochemistry assessed autophagy-related proteins, LC3 and p62 expression levels. Western blotting confirmed the expression levels of the autophagy-related proteins [Beclin1 and mammalian target of rapamycin (mTOR)], the endoplasmic reticulum (ER) stress-related proteins (PERK, eIF2α and CHOP) and the PI3K/Akt/mTOR signalling pathway-related proteins. Real-time quantitative PCR (qRT-PCR) determined IL-1β, IL-6 and MMP9 gene expression levels. The TUNEL assay detected apoptotic cells. TEM identified autophagic vacuoles in both EK models. Increased LC3 puncta formation and decreased p62 immunofluorescent staining and Western blotting confirmed autophagy induction. CQ treatment increased TUNEL positive staining in HCECs, while Rapa had an opposite effect. Similarly, CQ injection enhanced air exposure-induced apoptosis and inflammation in the mouse corneal epithelium, which was inhibited by Rapa treatment. Furthermore, the phosphorylation status of PERK and eIF2α and CHOP expression increased in both EK models indicating that ER stress-induced autophagy promoted cell survival. Taken together, air exposure-induced autophagy is indispensable for the maintenance of corneal epithelial physiology and cell survival.
Collapse
Affiliation(s)
- Guoliang Wang
- Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, China.,Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen, China
| | - Yuhua Xue
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Yanzi Wang
- Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, China.,Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen, China
| | - Fei Dong
- Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, China.,Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen, China
| | - Mei Shen
- Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, China.,Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen, China
| | - Rongrong Zong
- Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, China.,Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen, China
| | - Zuguo Liu
- Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, China.,Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen, China
| | - Cheng Li
- Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, China.,Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen, China
| |
Collapse
|
32
|
Wang Y, Shao Y, Gao Y, Wan G, Wan D, Zhu H, Qiu Y, Ye X. Catalpol prevents denervated muscular atrophy related to the inhibition of autophagy and reduces BAX/BCL2 ratio via mTOR pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 13:243-253. [PMID: 30643390 PMCID: PMC6319426 DOI: 10.2147/dddt.s188968] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Aim To investigate the effects of catalpol on muscular atrophy induced by sciatic nerve crush injury (SNCI). Methods Seventy male Kunming mice were randomized into five groups (n=10): model, sham, catalpol (Cat), rapamycin (Rapa), and catalpol+rapamycin (Rapa+Cat). The ratio of gastrocnemius muscle wet weight (right/left, R/L) between the operated leg (right) and the normal leg (left) was calculated, and acetylcholinesterase (AChE) immunohistochemistry assays were performed to observe the change of motor end plate (MEP), along with the sizes of denervated and innervated muscle fibers. The expression levels of LC3II, TUNEL, BAX/BCL-2, LC3II/LC3I and P62, Beclin1, mTOR, and p-mTOR (ser2448) proteins in muscle were examined by fluorescence immunohistochemistry or Western blotting. Results Results show that catalpol improved the results of the grid walking tests by reducing the percentage of foot slips, which increased the gastrocnemius muscle wet weight (R/L), enhanced AChE expression at the MEP, and enlarged the section area of the muscle. The expression of LC3II and TUNEL was significantly inhibited by catalpol. The BAX/BCL-2 ratio was significantly increased in muscles of denervated and control groups. Lower LC3II/LC3I and BAX/BCL-2 ratios in denervated muscles were also detected after catalpol treatment. Conclusion These results indicated that apoptosis and autophagy play a role in the regulation of denervation-induced muscle atrophy after SNCI, and catalpol alleviates muscle atrophy through the regulation of muscle apoptosis and autophagy via the mTOR signaling pathway.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Chinese Medicine, College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China, ;
| | - Yali Shao
- Department of Chinese Medicine, College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China, ;
| | - Yuqing Gao
- Department of Chinese Medicine, College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China, ;
| | - Guoran Wan
- Department of Clinic Medicine, Chongqing Three Gorges Medical College, Chongqing 404120, China
| | - Dong Wan
- Department of Emergency, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Huifeng Zhu
- Department of Chinese Medicine, College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China, ;
| | - Yan Qiu
- Department of Chinese Medicine, College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China, ;
| | - Xiyue Ye
- Department of Chinese Medicine, College of Pharmaceutical Sciences and Traditional Chinese Medicine, Southwest University, Chongqing Engineering Research Center for Pharmacological Evaluation, Chongqing 400715, China, ;
| |
Collapse
|
33
|
Saraswat Ohri S, Bankston AN, Mullins SA, Liu Y, Andres KR, Beare JE, Howard RM, Burke DA, Riegler AS, Smith AE, Hetman M, Whittemore SR. Blocking Autophagy in Oligodendrocytes Limits Functional Recovery after Spinal Cord Injury. J Neurosci 2018; 38:5900-5912. [PMID: 29793971 PMCID: PMC6021994 DOI: 10.1523/jneurosci.0679-17.2018] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Revised: 05/15/2018] [Accepted: 05/17/2018] [Indexed: 01/21/2023] Open
Abstract
Autophagy mechanisms are well documented in neurons after spinal cord injury (SCI), but the direct functional role of autophagy in oligodendrocyte (OL) survival in SCI pathogenesis remains unknown. Autophagy is an evolutionary conserved lysosomal-mediated catabolic pathway that ensures degradation of dysfunctional cellular components to maintain homeostasis in response to various forms of stress, including nutrient deprivation, hypoxia, reactive oxygen species, DNA damage, and endoplasmic reticulum (ER) stress. Using pharmacological gain and loss of function and genetic approaches, we investigated the contribution of autophagy in OL survival and its role in the pathogenesis of thoracic contusive SCI in female mice. Although upregulation of Atg5 (an essential autophagy gene) occurs after SCI, autophagy flux is impaired. Purified myelin fractions of contused 8 d post-SCI samples show enriched protein levels of LC3B, ATG5, and BECLIN 1. Data show that, while the nonspecific drugs rapamycin (activates autophagy) and spautin 1 (blocks autophagy) were pharmacologically active on autophagy in vivo, their administration did not alter locomotor recovery after SCI. To directly analyze the role of autophagy, transgenic mice with conditional deletion of Atg5 in OLs were generated. Analysis of hindlimb locomotion demonstrated a significant reduction in locomotor recovery after SCI that correlated with a greater loss in spared white matter. Immunohistochemical analysis demonstrated that deletion of Atg5 from OLs resulted in decreased autophagic flux and was detrimental to OL function after SCI. Thus, our study provides evidence that autophagy is an essential cytoprotective pathway operating in OLs and is required for hindlimb locomotor recovery after thoracic SCI.SIGNIFICANCE STATEMENT This study describes the role of autophagy in oligodendrocyte (OL) survival and pathogenesis after thoracic spinal cord injury (SCI). Modulation of autophagy with available nonselective drugs after thoracic SCI does not affect locomotor recovery despite being pharmacologically active in vivo, indicating significant off-target effects. Using transgenic mice with conditional deletion of Atg5 in OLs, this study definitively identifies autophagy as an essential homeostatic pathway that operates in OLs and exhibits a direct functional role in SCI pathogenesis and recovery. Therefore, this study emphasizes the need to discover novel autophagy-specific drugs that specifically modulate autophagy for further investigation for clinical translation to treat SCI and other CNS pathologies related to OL survival.
Collapse
Affiliation(s)
| | - Andrew N Bankston
- Kentucky Spinal Cord Injury Research Center
- Departments of Neurological Surgery
| | - S Ashley Mullins
- Kentucky Spinal Cord Injury Research Center
- Departments of Neurological Surgery
| | - Yu Liu
- Kentucky Spinal Cord Injury Research Center
- Departments of Neurological Surgery
| | - Kariena R Andres
- Kentucky Spinal Cord Injury Research Center
- Departments of Neurological Surgery
| | - Jason E Beare
- Kentucky Spinal Cord Injury Research Center
- Cardiovascular Innovation Institute, University of Louisville, School of Medicine, Louisville, Kentucky 40292
| | - Russell M Howard
- Kentucky Spinal Cord Injury Research Center
- Departments of Neurological Surgery
| | - Darlene A Burke
- Kentucky Spinal Cord Injury Research Center
- Departments of Neurological Surgery
| | - Amberly S Riegler
- Kentucky Spinal Cord Injury Research Center
- Departments of Neurological Surgery
| | - Allison E Smith
- Kentucky Spinal Cord Injury Research Center
- Departments of Neurological Surgery
| | - Michal Hetman
- Kentucky Spinal Cord Injury Research Center
- Departments of Neurological Surgery
- Pharmacology & Toxicology
- Anatomical Sciences & Neurobiology, and
| | - Scott R Whittemore
- Kentucky Spinal Cord Injury Research Center,
- Departments of Neurological Surgery
- Pharmacology & Toxicology
- Anatomical Sciences & Neurobiology, and
| |
Collapse
|
34
|
Schwarze K, Kribben A, Ritter O, Müller GA, Patschan D. Autophagy activation in circulating proangiogenic cells aggravates AKI in type I diabetes mellitus. Am J Physiol Renal Physiol 2018; 315:F1139-F1148. [PMID: 29897281 DOI: 10.1152/ajprenal.00502.2017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Acute kidney injury (AKI) occurs frequently in hospitals worldwide, but the therapeutic options are limited. Diabetes mellitus (DM) affects more and more people around the globe. The disease worsens the prognosis of AKI even further. In recent years, cell-based therapies have increasingly been applied in experimental AKI. The aim of the study was to utilize two established autophagy inducers for pharmacological preconditioning of so-called proangiogenic cells (PACs) in PAC treatment of diabetic AKI. Insulin-dependent DM was induced in male C57/Bl6N mice by intraperitoneal injections of streptozotocine. Six weeks later, animals underwent bilateral renal ischemia for 45 min, followed by intravenous injections of either native or zVAD (benzyloxycarbonyl-Val-Ala-Asp-fluoro-methylketone)- or Z-Leu-Leu-Leu-al (MG132)-pretreated syngeneic murine PACs. Mice were analyzed 48 h (short term) and 6 wk (long term) later, respectively. DM worsened postischemic AKI, and PAC preconditioning with zVAD and MG132 resulted in a further decline of excretory kidney function. Injection of native PACs reduced fibrosis in nondiabetic mice, but cell preconditioning promoted interstitial matrix accumulation significantly. Both substances aggravated endothelial-to-mesenchymal transition (EndoMT) under diabetic conditions; these effects occurred either exclusively in the short (zVAD) or in the short and long term (MG132). Preconditioned cells stimulated the autophagocytic flux in intrarenal endothelial cells, and all experimental groups displayed increased endothelial abundances of senescence-associated β-galactosidase, a marker of premature cell senescence. Pharmacological autophagy activation may not serve as an effective strategy for improving PAC competence in diabetic AKI in general. On the contrary, several outcome parameters (excretory function, fibrosis, EndoMT) may even be worsened.
Collapse
Affiliation(s)
- K Schwarze
- Clinic of Nephrology and Rheumatology, University Hospital of Göttingen , Göttingen , Germany
| | - A Kribben
- Department of Nephrology, University Hospital Essen, University Duisburg-Essen , Essen , Germany
| | - O Ritter
- Department of Cardiology, Pulmology, Angiology, and Nephrology, Brandenburg Medical School, University Hospital Brandenburg , Brandenburg , Germany
| | - G A Müller
- Clinic of Nephrology and Rheumatology, University Hospital of Göttingen , Göttingen , Germany
| | - D Patschan
- Department of Cardiology, Pulmology, Angiology, and Nephrology, Brandenburg Medical School, University Hospital Brandenburg , Brandenburg , Germany
| |
Collapse
|
35
|
Muñoz-Galdeano T, Reigada D, Del Águila Á, Velez I, Caballero-López MJ, Maza RM, Nieto-Díaz M. Cell Specific Changes of Autophagy in a Mouse Model of Contusive Spinal Cord Injury. Front Cell Neurosci 2018; 12:164. [PMID: 29946241 PMCID: PMC6005838 DOI: 10.3389/fncel.2018.00164] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Accepted: 05/28/2018] [Indexed: 12/15/2022] Open
Abstract
Autophagy is an essential process of cellular waist clearance that becomes altered following spinal cord injury (SCI). Details on these changes, including timing after injury, underlying mechanisms, and affected cells, remain controversial. Here we present a characterization of autophagy in the mice spinal cord before and after a contusive SCI. In the undamaged spinal cord, analysis of LC3 and Beclin 1 autophagic markers reveals important differences in basal autophagy between neurons, oligodendrocytes, and astrocytes and even within cell populations. Following moderate contusion, western blot analyses of LC3 indicates that autophagy increases to a maximum at 7 days post injury (dpi), whereas unaltered Beclin 1 expression and increase of p62 suggests a possible blockage of autophagosome clearance. Immunofluorescence analyses of LC3 and Beclin 1 provide additional details that reveal a complex, cell-specific scenario. Autophagy is first activated (1 dpi) in the severed axons, followed by a later (7 dpi) accumulation of phagophores and/or autophagosomes in the neuronal soma without signs of increased initiation. Oligodendrocytes and reactive astrocytes also accumulate phagophores and autophagosomes at 7 dpi, but whereas the accumulation in astrocytes is associated with an increased autophagy initiation, it seems to result from a blockage of the autophagic flux in oligodendrocytes. Comparison with previous studies highlights the complex and heterogeneous autophagic responses induced by the SCI, leading in many cases to contradictory results and interpretations. Future studies should consider this complexity in the design of therapeutic interventions based on the modulation of autophagy to treat SCI.
Collapse
Affiliation(s)
- Teresa Muñoz-Galdeano
- Laboratory of Molecular Neuroprotection, UDI-HNP, Hospital Nacional de Parapléjicos (SESCAM), Toledo, Spain
| | - David Reigada
- Laboratory of Molecular Neuroprotection, UDI-HNP, Hospital Nacional de Parapléjicos (SESCAM), Toledo, Spain
| | - Ángela Del Águila
- Laboratory of Molecular Neuroprotection, UDI-HNP, Hospital Nacional de Parapléjicos (SESCAM), Toledo, Spain.,Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Irene Velez
- Laboratory of Molecular Neuroprotection, UDI-HNP, Hospital Nacional de Parapléjicos (SESCAM), Toledo, Spain
| | - Marcos J Caballero-López
- Laboratory of Molecular Neuroprotection, UDI-HNP, Hospital Nacional de Parapléjicos (SESCAM), Toledo, Spain
| | - Rodrigo M Maza
- Laboratory of Molecular Neuroprotection, UDI-HNP, Hospital Nacional de Parapléjicos (SESCAM), Toledo, Spain
| | - Manuel Nieto-Díaz
- Laboratory of Molecular Neuroprotection, UDI-HNP, Hospital Nacional de Parapléjicos (SESCAM), Toledo, Spain
| |
Collapse
|
36
|
Wang P, Shao BZ, Deng Z, Chen S, Yue Z, Miao CY. Autophagy in ischemic stroke. Prog Neurobiol 2018; 163-164:98-117. [DOI: 10.1016/j.pneurobio.2018.01.001] [Citation(s) in RCA: 183] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 12/04/2017] [Accepted: 01/10/2018] [Indexed: 02/07/2023]
|
37
|
Inhibition of autophagy potentiates the apoptosis-inducing effects of photodynamic therapy on human colon cancer cells. Photodiagnosis Photodyn Ther 2018; 21:396-403. [DOI: 10.1016/j.pdpdt.2018.01.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 01/08/2018] [Accepted: 01/16/2018] [Indexed: 01/11/2023]
|
38
|
Li Z, Liu F, Zhang L, Cao Y, Shao Y, Wang X, Jiang X, Chen Z. Neuroserpin restores autophagy and promotes functional recovery after acute spinal cord injury in rats. Mol Med Rep 2018; 17:2957-2963. [PMID: 29257287 PMCID: PMC5783514 DOI: 10.3892/mmr.2017.8249] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Accepted: 04/06/2017] [Indexed: 01/07/2023] Open
Abstract
This study is to reveal the characteristics of autophagy and the effect of neuroserpin (NSP) treatment on autophagy during the process of functional recovery following spinal cord injury (SCI). After the clip compress rat model of SCI had been made, autophagy‑associated proteins, including LC3‑II, beclin‑1 and p62, were evaluated at 2, 4, 24, 72 h, and 168 h in the experimental group, and the sham group as control. Transmission electron microscopy (TEM) was further used for autophagy detection at 4 and 72 h. All the male rats were randomly divided into three groups: Sham, vehicle and NSP group. NSP or an equal volume of saline vehicle was administered via intrathecal injection immediately after SCI. Each group was further divided into subgroups for the following experiments: i)Western blot (LC3‑II and p62); ii) Immunofluorescent double staining (LC3/MAP‑2/DAPI); iii) Nissl staining and Basso Beattie Bresnahan (BBB score) for NSP neuroprotection evaluation. Our results revealed both LC3‑II and p62 expression trended upward at 24, 72 and 168 h after SCI. The LC3‑II peaked at 72 h, while p62 peaked at 24 h. Beclin‑1 dropped significantly at 72 and 168 h. TEM results showed that autophagosomes largely accumulated at 72 h after SCI when compared with the sham group. Western blot analysis showed that LC3‑II and p62 were markedly decreased with NSP treatment at 72 h after injury compared with that of the vehicle‑group. Immunofluorescent double labeling indicated that accumulation of autophagosomes was reduced in the NSP group. Further, post‑SCI treatment with NSP improved the BBB scale and increased the number of anterior horn motor neurons. Together, this study demonstrates that autophagic flux is impaired, meanwhile NSP restores autophagic flux and promotes functional recovery after SCI in rats.
Collapse
Affiliation(s)
- Zheng Li
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Fubing Liu
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Liang Zhang
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Yuanwu Cao
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Yunchao Shao
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Xiaofeng Wang
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Xiaoxing Jiang
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Zixian Chen
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|
39
|
Hei C, Liu P, Yang X, Niu J, Li PA. Inhibition of mTOR signaling Confers Protection against Cerebral Ischemic Injury in Acute Hyperglycemic Rats. Int J Biol Sci 2017; 13:878-887. [PMID: 28808420 PMCID: PMC5555105 DOI: 10.7150/ijbs.18976] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 01/28/2017] [Indexed: 01/04/2023] Open
Abstract
Hyperglycemia is known to exacerbate neuronal death resulted from cerebral ischemia. The mechanisms are not fully understood. The mammalian target of rapamycin (mTOR) pathway regulates cell growth, division and apoptosis. Recent studies suggest that activation of mTOR may mediate ischemic brain damage. The objective of the present experiment is to explore whether mTOR mediates ischemic brain damage in acute hyperglycemic animals. Rats were subjected to 10 min of forebrain ischemia under euglycemic, hyperglycemic and rapamycin-treated hyperglycemic conditions. The rat brain samples were collected from the cortex and hippocampi after 3h and 16h of reperfusion. The results showed that hyperglycemia significantly increased neuronal death in the cortex and hippocampus and the exacerbation effect of hyperglycemia was associated with further activation of mTOR under control and/or ischemic conditions. Inhibition of mTOR with rapamycin ameliorated the damage and suppressed hyperglycemia-elevated p-MTOR, p-P70S6K and p-S6. In addition, hyperglycemia per se increased the levels of cytosolic cytochrome c and autophagy marker LC3-II, while rapamycin alleviated these alterations. It is concluded that activation of mTOR signaling may play a detrimental role in mediating the aggravating effect of hyperglycemia on cerebral ischemia.
Collapse
Affiliation(s)
- Changchun Hei
- Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region and Department Human Anatomy, Histology and Embryology, Ningxia Medical University, Yinchuan 750004, China.,Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, 1801 Fayetteville Street, Durham, NC 27707, USA
| | - Ping Liu
- Department of Endocrinology, General Hospital of Ningxia Medical University, Yinchuan 750004, China.,Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, 1801 Fayetteville Street, Durham, NC 27707, USA
| | - Xiao Yang
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, 1801 Fayetteville Street, Durham, NC 27707, USA.,Neuroscience Center, General Hospital of Ningxia Medical University, and Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Yinchuan 750004, China
| | - Jianguo Niu
- Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region and Department Human Anatomy, Histology and Embryology, Ningxia Medical University, Yinchuan 750004, China
| | - P Andy Li
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, 1801 Fayetteville Street, Durham, NC 27707, USA
| |
Collapse
|
40
|
Hirt J, Liton PB. Autophagy and mechanotransduction in outflow pathway cells. Exp Eye Res 2017; 158:146-153. [PMID: 27373974 PMCID: PMC5199638 DOI: 10.1016/j.exer.2016.06.021] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 06/23/2016] [Accepted: 06/27/2016] [Indexed: 12/19/2022]
Abstract
Because of elevations in IOP and other forces, cells in the trabecular meshwork (TM) are constantly subjected to mechanical strain. In order to preserve cellular function and regain homeostasis, cells must sense and adapt to these morphological changes. We and others have already shown that mechanical stress can trigger a broad range of responses in TM cells; however, very little is known about the strategies that TM cells use to respond to this stress, so they can adapt and survive. Autophagy, a lysosomal degradation pathway, has emerged as an important cellular homeostatic mechanism promoting cell survival and adaptation to a number of cytotoxic stresses. Our laboratory has reported the activation of autophagy in TM cells in response to static biaxial strain and high pressure. Moreover, our newest data also suggest the activation of chaperon-assisted selective autophagy, a recently identified tension-induced autophagy essential for mechanotransduction, in TM cells under cyclic mechanical stress. In this review manuscript we will discuss autophagy as part of an integrated response triggered in TM cells in response to strain, exerting a dual role in repair and mechanotransduction, and the potential effects of dysregulated in outflow pathway pathophysiology.
Collapse
Affiliation(s)
- Joshua Hirt
- Duke University, Department of Ophthalmology, Durham, NC, USA
| | - Paloma B Liton
- Duke University, Department of Ophthalmology, Durham, NC, USA.
| |
Collapse
|
41
|
Expression of autophagy in different stages of neurogenic bladder after spinal cord injury in rats. Spinal Cord 2017; 55:834-839. [DOI: 10.1038/sc.2017.37] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 03/06/2017] [Accepted: 03/07/2017] [Indexed: 02/05/2023]
|
42
|
Bao J, Li G, Yuan X, Li PL, Gulbins E. Contribution of p62 to Phenotype Transition of Coronary Arterial Myocytes with Defective Autophagy. Cell Physiol Biochem 2017; 41:555-568. [PMID: 28214847 PMCID: PMC8216328 DOI: 10.1159/000457877] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 12/04/2016] [Indexed: 12/30/2022] Open
Abstract
Background: Autophagy disorder contributes to dedifferentiation of arterial smooth muscle cells, but the mechanisms are poorly understood. Here, we sought to investigate the role of scaffolding adaptor p62/SQSTM1 (p62) in phenotype switching of mouse coronary arterial myocytes (CAMs) induced by CD38 gene deficiency or lysosomal dysfunction which blocks autophagic flux in the cells. Methods: Protein expression was measured by western blot analysis and immunofluorescent staining. Cell cycle and proliferation rate were analyzed by flow cytometry and MTS assay respectively. mRNA abundance was tested by qRT-PCR. Results: CD38 gene deficiency or bafilomycin A1 (baf), a selective lysosomal inhibitor treatment increased proliferation rate and vimentin expression in CAMs which was prevented by p62 gene silencing. Cell percentage in G2/M and G0/G1 phase was decreased and increased by CD38 deficiency or baf treatment, respectively which was accompanied by accrual of cyclin-dependent kinase 1 (CDK1) protein. Although free ubiquitin content was increased, the colocalization of it to CDK1 was markedly decreased in CD38−/− or baf treated CAMs. Furthermore, the changes in both cell cycle and CDK1 ubiquitinylation could be restored by p62 gene silencing. Conclusion: The results suggest in CD38−/− or baf treated CAMs, p62 accumulation promotes phenotype transition and proliferation by accelerating cell cycle progress through G2/M which might relate to the compromised ubiquitinylation and degradation of CDK1.
Collapse
|
43
|
Wei X, Zhou Z, Li L, Gu J, Wang C, Xu F, Dong Q, Zhou X. Intrathecal Injection of 3-Methyladenine Reduces Neuronal Damage and Promotes Functional Recovery via Autophagy Attenuation after Spinal Cord Ischemia/Reperfusion Injury in Rats. Biol Pharm Bull 2017; 39:665-73. [PMID: 27150140 DOI: 10.1248/bpb.b15-00610] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The present study aimed to determine the occurrence of autophagy following ischemia/reperfusion (I/R) injury in the rat spinal cord and whether autophagy inhibition contributes to neural tissue damage and locomotor impairment. A spinal cord I/R model was induced via descending thoracic aorta occlusion for 10 min using systemic hypotension (40 mmHg) in adult male Sprague-Dawley rats. Then, 600 nmol 3-methyladenine (3-MA) or vehicle was intrathecally administered. Ultrastructural spinal cord changes were observed via transmission electron microscopy (TEM) and immunofluorescent double-labeling. Western blots were used to determine the protein expression of microtubule-associated protein light chain 3 (LC3) and Beclin 1. Autophagy was activated after spinal cord I/R injury as demonstrated by significantly increased LC3 and Beclin 1 expression at 3-48 h after injury. Furthermore, TEM images indicated the presence of autophagosomes and autolysosomes in the injured spinal cord. 3-MA significantly decreased LC3 and Beclin 1 expression and the number of LC3-positive cells in spinal cord of I/R versus vehicle groups. Moreover, the 3-MA-treated rats exhibited better neurobehavioral scores compared with control rats. These findings suggest activation of autophagy leading to neuronal cell death in the I/R injured spinal cord. These effects were significantly inhibited by intrathecal 3-MA administration. Thus intrathecal 3-MA administration may represent a novel treatment target following spinal cord I/R injury.
Collapse
Affiliation(s)
- Xing Wei
- Department of Anesthesia, the Second Affiliated Hospital of Soochow University
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Autophagy transduces physical constraints into biological responses. Int J Biochem Cell Biol 2016; 79:419-426. [PMID: 27566364 DOI: 10.1016/j.biocel.2016.08.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 08/18/2016] [Accepted: 08/22/2016] [Indexed: 12/15/2022]
Abstract
Autophagy is a fundamental cell biological process that controls the quality and quantity of the eukaryotic cytoplasm. Dysfunctional autophagy, when defective or excessive, has been linked to human pathologies ranging from neurodegenerative and infectious diseases to cancer and inflammatory diseases. Autophagy takes place at basal levels in all eukaryotic cells. The process is stimulated during metabolic, genotoxic, infectious, and hypoxic stress conditions and acts an adaptive mechanism essential for cell survival. Recent data demonstrate that changes in the mechanical cellular environment influence cell fate through the modulation of the autophagic pathway. Mechanical stimuli, such as applied forces, combine with biochemical signals to control development and physiological functions of different organs and can also contribute to the progression of various human diseases. Here we review recent findings regarding the regulation of autophagy upon three types of mechanical stress, compression, shear stress, and stretching, and discuss the potential implications of mechanical stress-induced autophagy in physiology and physiopathology.
Collapse
|
45
|
Liu P, Yang X, Hei C, Meli Y, Niu J, Sun T, Li PA. Rapamycin Reduced Ischemic Brain Damage in Diabetic Animals Is Associated with Suppressions of mTOR and ERK1/2 Signaling. Int J Biol Sci 2016; 12:1032-40. [PMID: 27489506 PMCID: PMC4971741 DOI: 10.7150/ijbs.15624] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 05/28/2016] [Indexed: 12/17/2022] Open
Abstract
The objectives of the present study are to investigate the activation of mTOR and ERK1/2 signaling after cerebral ischemia in diabetic rats and to examine the neuroprotective effects of rapamycin. Ten minutes transient global cerebral ischemia was induced in straptozotocin-induced diabetic hyperglycemic rats and non-diabetic, euglycemic rats. Brain samples were harvested after 16 h of reperfusion. Rapamycin or vehicle was injected 1 month prior to the induction of ischemia. The results showed that diabetes increased ischemic neuronal cell death and associated with elevations of p-P70S6K and Ras/ERK1/2 and suppression of p-AMPKα. Rapamycin ameliorated diabetes-enhanced ischemic brain damage and suppressed phosphorylation of P70S6K and ERK1/2. It is concluded that diabetes activates mTOR and ERK1/2 signaling pathways in rats subjected to transient cerebral ischemia and inhibition of mTOR by rapamycin reduces ischemic brain damage and suppresses the mTOR and ERK1/2 signaling in diabetic settings.
Collapse
Affiliation(s)
- Ping Liu
- 1. Department of Endocrinology, General Hospital of Ningxia Medical University, Yinchuan 750004, China
- 2. Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, 1801 Fayetteville Street, Durham, NC 27707, USA
| | - Xiao Yang
- 2. Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, 1801 Fayetteville Street, Durham, NC 27707, USA
- 3. Neuroscience Center, General Hospital of Ningcia Medical University, and Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Yinchuan 750004, China
| | - Changchun Hei
- 2. Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, 1801 Fayetteville Street, Durham, NC 27707, USA
- 4. Department of Human Anatomy, Histology and Embryology, Ningxia Medical University, Yinchuan 75004, China
| | - Yvonne Meli
- 2. Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, 1801 Fayetteville Street, Durham, NC 27707, USA
| | - Jianguo Niu
- 4. Department of Human Anatomy, Histology and Embryology, Ningxia Medical University, Yinchuan 75004, China
| | - Tao Sun
- 4. Department of Human Anatomy, Histology and Embryology, Ningxia Medical University, Yinchuan 75004, China
| | - P. Andy Li
- 2. Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, 1801 Fayetteville Street, Durham, NC 27707, USA
| |
Collapse
|
46
|
Xie S, Deng Y, Pan YY, Ren J, Jin M, Wang Y, Wang ZH, Zhu D, Guo XL, Yuan X, Shang J, Liu HG. Chronic intermittent hypoxia induces cardiac hypertrophy by impairing autophagy through the adenosine 5'-monophosphate-activated protein kinase pathway. Arch Biochem Biophys 2016; 606:41-52. [PMID: 27412517 DOI: 10.1016/j.abb.2016.07.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 07/05/2016] [Accepted: 07/08/2016] [Indexed: 12/19/2022]
Abstract
Autophagy is tightly regulated to maintain cardiac homeostasis. Impaired autophagy is closely associated with pathological cardiac hypertrophy. However, the relationship between autophagy and cardiac hypertrophy induced by chronic intermittent hypoxia (CIH) is not known. In the present study, we measured autophagy-related genes and autophagosomes during 10 weeks of CIH in rats, and 6 days in H9C2 cardiomyocytes, and showed that autophagy was impaired. This conclusion was confirmed by the autophagy flux assay. We detected significant hypertrophic changes in myocardium with impaired autophagy. Rapamycin, an autophagy enhancer, attenuated the cardiac hypertrophy induced by CIH. Moreover, silencing autophagy-related gene 5 (ATG5) exerted the opposite effect. The role of adenosine monophosphate-activated protein kinase (AMPK) in regulating autophagy under CIH was confirmed using AICAR to upregulate this enzyme and restore autophagy flux. Restoring autophagy by AICAR or rapamycin significantly reversed the hypertrophic changes in cardiomyocytes. To investigate the mechanism of autophagy impairment, we compared phospho (p)-AMPK, p-Akt, cathepsin D, and NFAT3 levels, along with calcineurin activity, between sham and CIH groups. CIH activated calcineurin, and inhibited AMPK and AMPK-mediated autophagy in an Akt- and NFAT3-independent manner. Collectively, these data demonstrated that impaired autophagy induced by CIH through the AMPK pathway contributed to cardiac hypertrophy.
Collapse
Affiliation(s)
- Sheng Xie
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, China
| | - Yan Deng
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, China
| | - Yue-Ying Pan
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, China
| | - Jie Ren
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, China
| | - Meng Jin
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, China
| | - Yu Wang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, China
| | - Zhi-Hua Wang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, China
| | - Die Zhu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, China
| | - Xue-Ling Guo
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, China
| | - Xiao Yuan
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, China
| | - Jin Shang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, China
| | - Hui-Guo Liu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Huazhong University of Science and Technology, China.
| |
Collapse
|
47
|
Chakrabarti M, Klionsky DJ, Ray SK. miR-30e Blocks Autophagy and Acts Synergistically with Proanthocyanidin for Inhibition of AVEN and BIRC6 to Increase Apoptosis in Glioblastoma Stem Cells and Glioblastoma SNB19 Cells. PLoS One 2016; 11:e0158537. [PMID: 27388765 PMCID: PMC4936720 DOI: 10.1371/journal.pone.0158537] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 06/17/2016] [Indexed: 11/18/2022] Open
Abstract
Glioblastoma is the most common and malignant brain tumor in humans. It is a heterogeneous tumor harboring glioblastoma stem cells (GSC) and other glioblastoma cells that survive and sustain tumor growth in a hypoxic environment via induction of autophagy and resistance to apoptosis. So, a therapeutic strategy to inhibit autophagy and promote apoptosis could greatly help control growth of glioblastoma. We created hypoxia using sodium sulfite (SS) for induction of substantiated autophagy in human GSC and glioblastoma SNB19 cells. Induction of autophagy was confirmed by acridine orange (AO) staining and significant increase in Beclin-1 in autophagic cells. microRNA database (miRDB) search suggested that miR-30e could suppress the autophagy marker Beclin-1 and also inhibit the caspase activation inhibitors (AVEN and BIRC6). Pro-apoptotic effect of proanthocyanidin (PAC) has not yet been explored in glioblastoma cells. Combination of 50 nM miR-30e and 150 μM PAC acted synergistically for inhibition of viability in both cells. This combination therapy most effectively altered expression of molecules for inhibition of autophagy and induced extrinsic and intrinsic pathways of apoptosis through suppression of AVEN and BIRC6. Collectively, combination of miR-30e and PAC is a promising therapeutic strategy to inhibit autophagy and increase apoptosis in GSC and SNB19 cells.
Collapse
Affiliation(s)
- Mrinmay Chakrabarti
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, United States of America
| | - Daniel J. Klionsky
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Swapan K. Ray
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, United States of America
- * E-mail:
| |
Collapse
|
48
|
Li H, Qiu S, Li X, Li M, Peng Y. Autophagy biomarkers in CSF correlates with infarct size, clinical severity and neurological outcome in AIS patients. J Transl Med 2015; 13:359. [PMID: 26576535 PMCID: PMC4650838 DOI: 10.1186/s12967-015-0726-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 11/03/2015] [Indexed: 12/12/2022] Open
Abstract
Background Autophagy is demonstrated to be involved in acute ischemic stroke(AIS), which, however, is confined to cells and/or animals levels. The aim of this study was to determine two autophagy biomarkers, Beclin1 and LC3B, in cerebrospinal fluid (CSF) and serum of patients with AIS, and to evaluate a possible correlation between levels of Beclin1 and LC3B and severity of neurological deficit and clinical outcome of stroke patients. Methods Levels of Beclin1 and LC3B were quantified by ELISA in CSF and serum collected from 37 AIS patients and 21 controls. The clinical severity at stroke onset was determined by the National Institute of Health Stroke Scale (NIHSS) and the neurological outcome was determined by the Modified Rankin Scale (mRs) and the improvement in NIHSS between stroke onset and 3 months later. Associations between autophagy biomarkers and infarct volume, NIHSS and mRs were assessed using Pearson analysis. Results The levels of Beclin1 and LC3B were increased both in CSF and serum of AIS patients relative to controls. In CSF, they were positively correlated with infarct volume and NIHSS scores, and negatively correlated with mRs scores, but no significant association was observed in serum. Moreover, AIS patients with higher levels of Beclin1 and LC3B in CSF had significantly higher improvement in NIHSS. Conclusion CSF and serum levels of autophagy biomarkers are altered in AIS patients. CSF levels of autophagy biomarkers are associated with infarct volume, clinical severity of and neurological outcome.
Collapse
Affiliation(s)
- Honghong Li
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 West Yanjiang Road, Guangzhou, 510120, China. .,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Shuwei Qiu
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 West Yanjiang Road, Guangzhou, 510120, China. .,Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Xiangpen Li
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 West Yanjiang Road, Guangzhou, 510120, China.
| | - Mei Li
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 West Yanjiang Road, Guangzhou, 510120, China.
| | - Ying Peng
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 West Yanjiang Road, Guangzhou, 510120, China. .,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
49
|
Xie S, Deng Y, Pan YY, Wang ZH, Ren J, Guo XL, Yuan X, Shang J, Liu HG. Melatonin protects against chronic intermittent hypoxia-induced cardiac hypertrophy by modulating autophagy through the 5′ adenosine monophosphate-activated protein kinase pathway. Biochem Biophys Res Commun 2015; 464:975-981. [DOI: 10.1016/j.bbrc.2015.06.149] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 06/22/2015] [Indexed: 12/19/2022]
|
50
|
Lipinski MM, Wu J, Faden AI, Sarkar C. Function and Mechanisms of Autophagy in Brain and Spinal Cord Trauma. Antioxid Redox Signal 2015; 23:565-77. [PMID: 25808205 PMCID: PMC4545370 DOI: 10.1089/ars.2015.6306] [Citation(s) in RCA: 152] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
SIGNIFICANCE Traumatic brain injury (TBI) and spinal cord injury (SCI) are major causes of death and long-term disability worldwide. Despite important pathophysiological differences between these disorders, in many respects, mechanisms of injury are similar. During both TBI and SCI, some cells are directly mechanically injured, but more die as a result of injury-induced biochemical changes (secondary injury). Autophagy, a lysosome-dependent cellular degradation pathway with neuroprotective properties, has been implicated both clinically and experimentally in the delayed response to TBI and SCI. However, until recently, its mechanisms and function remained unknown, reflecting in part the difficulty of isolating autophagic processes from ongoing cell death and other cellular events. RECENT ADVANCES Emerging data suggest that depending on the location and severity of traumatic injury, autophagy flux--defined as the progress of cargo through the autophagy system and leading to its degradation--may be either increased or decreased after central nervous system trauma. CRITICAL ISSUES While increased autophagy flux may be protective after mild injury, after more severe trauma inhibition of autophagy flux may contribute to neuronal cell death, indicating disruption of autophagy as a part of the secondary injury mechanism. FUTURE DIRECTIONS Augmentation and/or restoration of autophagy flux may provide a potential therapeutic target for treatment of TBI and SCI. Development of those treatments will require thorough characterization of changes in autophagy flux, its mechanisms and function over time after injury.
Collapse
Affiliation(s)
- Marta M Lipinski
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine , Baltimore, Maryland
| | - Junfang Wu
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine , Baltimore, Maryland
| | - Alan I Faden
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine , Baltimore, Maryland
| | - Chinmoy Sarkar
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine , Baltimore, Maryland
| |
Collapse
|