1
|
Schmidt MJ, Naghdloo A, Prabakar RK, Kamal M, Cadaneanu R, Garraway IP, Lewis M, Aparicio A, Zurita-Saavedra A, Corn P, Kuhn P, Pienta KJ, Amend SR, Hicks J. Polyploid cancer cells reveal signatures of chemotherapy resistance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.19.608632. [PMID: 39229204 PMCID: PMC11370377 DOI: 10.1101/2024.08.19.608632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Therapeutic resistance in cancer significantly contributes to mortality, with many patients eventually experiencing recurrence after initial treatment responses. Recent studies have identified therapy-resistant large polyploid cancer cells in patient tissues, particularly in late-stage prostate cancer, linking them to advanced disease and relapse. Here, we analyzed bone marrow aspirates from 44 advanced prostate cancer patients and found the presence of circulating tumor cells with increased genomic content (CTC-IGC) was significantly associated with poorer progression-free survival. Single cell copy number profiling of CTC-IGC displayed clonal origins with typical CTCs, suggesting complete polyploidization. Induced polyploid cancer cells from PC3 and MDA-MB-231 cell lines treated with docetaxel or cisplatin were examined through single cell DNA sequencing, RNA sequencing, and protein immunofluorescence. Novel RNA and protein markers, including HOMER1, TNFRSF9, and LRP1, were identified as linked to chemotherapy resistance. These markers were also present in a subset of patient CTCs and associated with recurrence in public gene expression data. This study highlights the prognostic significance of large polyploid tumor cells, their role in chemotherapy resistance, and their expression of markers tied to cancer relapse, offering new potential avenues for therapeutic development.
Collapse
Affiliation(s)
- Michael J. Schmidt
- Convergent Science Institute in Cancer, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, California, USA
| | - Amin Naghdloo
- Convergent Science Institute in Cancer, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, California, USA
| | - Rishvanth K. Prabakar
- Convergent Science Institute in Cancer, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, California, USA
- Currently at: Simons Center for Quantitative Biology, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Mohamed Kamal
- Convergent Science Institute in Cancer, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, California, USA
- Department of Zoology, Faculty of Science, Benha University, Benha, Egypt
| | - Radu Cadaneanu
- Department of Urology, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA and VA Greater Los Angeles, University of California, Los Angeles, Los Angeles, California, USA
| | - Isla P. Garraway
- Department of Urology, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA and VA Greater Los Angeles, University of California, Los Angeles, Los Angeles, California, USA
| | - Michael Lewis
- VA Greater Los Angeles Medical Center, Los Angeles, CA, USA
- Departments of Medicine and Pathology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Center for Cancer Research and Cellular Therapeutics, Clark, Atlanta, GA, USA
| | - Ana Aparicio
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Amado Zurita-Saavedra
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paul Corn
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Peter Kuhn
- Convergent Science Institute in Cancer, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, California, USA
| | - Kenneth J. Pienta
- Cancer Ecology Center, The Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sarah R. Amend
- Cancer Ecology Center, The Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - James Hicks
- Convergent Science Institute in Cancer, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
2
|
Dou Y, Fei X, He X, Huan Y, Wei J, Wu X, Lyu W, Fei Z, Li X, Fei F. Homer1a reduces inflammatory response after retinal ischemia/reperfusion injury. Neural Regen Res 2024; 19:1608-1617. [PMID: 38051906 PMCID: PMC10883521 DOI: 10.4103/1673-5374.386490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 08/25/2023] [Indexed: 12/07/2023] Open
Abstract
Abstract
JOURNAL/nrgr/04.03/01300535-202407000-00042/figure1/v/2023-11-20T171125Z/r/image-tiff
Elevated intraocular pressure (IOP) is one of the causes of retinal ischemia/reperfusion injury, which results in NLRP3 inflammasome activation and leads to visual damage. Homer1a is reported to play a protective role in neuroinflammation in the cerebrum. However, the effects of Homer1a on NLRP3 inflammasomes in retinal ischemia/reperfusion injury caused by elevated IOP remain unknown. In our study, animal models were constructed using C57BL/6J and Homer1flox/
–/Homer1a+/
–/Nestin-Cre+/
– mice with elevated IOP-induced retinal ischemia/reperfusion injury. For in vitro experiments, the oxygen-glucose deprivation/reperfusion injury model was constructed with Müller cells. We found that Homer1a overexpression ameliorated the decreases in retinal thickness and Müller cell viability after ischemia/reperfusion injury. Furthermore, Homer1a knockdown promoted NF-κB P65Ser536 activation via caspase-8, NF-κB P65 nuclear translocation, NLRP3 inflammasome formation, and the production and processing of interleukin-1β and interleukin-18. The opposite results were observed with Homer1a overexpression. Finally, the combined administration of Homer1a protein and JSH-23 significantly inhibited the reduction in retinal thickness in Homer1flox/
–/Homer1a+/
–/Nestin-Cre+/
– mice and apoptosis in Müller cells after ischemia/reperfusion injury. Taken together, these studies demonstrate that Homer1a exerts protective effects on retinal tissue and Müller cells via the caspase-8/NF-κB P65/NLRP3 pathway after I/R injury.
Collapse
Affiliation(s)
- Yanan Dou
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, Xi'an, Shaanxi Province, China
| | - Xiaowei Fei
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, Xi'an, Shaanxi Province, China
| | - Xin He
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, Xi'an, Shaanxi Province, China
| | - Yu Huan
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, Xi'an, Shaanxi Province, China
| | - Jialiang Wei
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, Xi'an, Shaanxi Province, China
| | - Xiuquan Wu
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, Xi'an, Shaanxi Province, China
| | - Weihao Lyu
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, Xi'an, Shaanxi Province, China
| | - Zhou Fei
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, Xi'an, Shaanxi Province, China
| | - Xia Li
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, Xi'an, Shaanxi Province, China
| | - Fei Fei
- Department of Ophthalmology, Xijing Hospital, Air Force Military Medical University, Xi'an, Shaanxi Province, China
| |
Collapse
|
3
|
Fei X, Wang L, Dou YN, Fei F, Zhang Y, Lv W, He X, Wu X, Chao W, Chen H, Wei J, Gao D, Fei Z. Extracellular vesicle encapsulated Homer1a as novel nanotherapeutics against intracerebral hemorrhage in a mouse model. J Neuroinflammation 2024; 21:85. [PMID: 38582897 PMCID: PMC10999083 DOI: 10.1186/s12974-024-03088-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 04/02/2024] [Indexed: 04/08/2024] Open
Abstract
Homer1a and A2 astrocytes are involved in the regulation of inflammation induced by intracerebral hemorrhage (ICH). However, there is no anticipated treatment strategy based on the anti-inflammatory effect of Homer1a and A2 astrocytes. Here, we successfully induced A2 astrocytes in vitro, and then we report an efficient method to prepare Homer1a+ EVs derived from A2 astrocytes which making it more stable, safe, and targetable to injured neurons. Homer1a+ EVs promotes the conversion of A1 to A2 astrocytes in ICH mice. Homer1a+ EVs inhibits activation and nuclear translocation of NF-κB, thereby regulating transcription of IL-17A in neurons. Homer1a+ EVs inhibits the RAGE/NF-κB/IL-17 signaling pathway and the binding ability of IL-17A: IL17-AR and RAGE: DIAPH1. In addition, Homer1a+ EVs ameliorates the pathology, behavior, and survival rate in GFAPCreHomer1fl/-Homer1a± and NestinCreRAGEfl/fl ICH mice. Our study provides a novel insight and potential for the clinical translation of Homer1a+ EVs in the treatment of ICH.
Collapse
Affiliation(s)
- Xiaowei Fei
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China
| | - Li Wang
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China
| | - Ya-Nan Dou
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China
| | - Fei Fei
- Department of Ophthalmology, Xijing Hospital, Air Force Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Yanyu Zhang
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China
| | - Weihao Lv
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China
| | - Xin He
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China
| | - Xiuquan Wu
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China
| | - Wangshu Chao
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China
| | - Hongqing Chen
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China
| | - Jialiang Wei
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China.
| | - Dakuan Gao
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China.
| | - Zhou Fei
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, , Shaanxi, 710032, China.
| |
Collapse
|
4
|
Zhao WJ, Fan CL, Hu XM, Ban XX, Wan H, He Y, Zhang Q, Xiong K. Regulated Cell Death of Retinal Ganglion Cells in Glaucoma: Molecular Insights and Therapeutic Potentials. Cell Mol Neurobiol 2023; 43:3161-3178. [PMID: 37338781 DOI: 10.1007/s10571-023-01373-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 06/05/2023] [Indexed: 06/21/2023]
Abstract
Glaucoma is a group of diseases characterized by the degeneration of retinal ganglion cells (RGCs) and progressive, irreversible vision loss. High intraocular pressure (IOP) heightens the likelihood of glaucoma and correlates with RGC loss. While the current glaucoma therapy prioritizes lower the IOP; however, RGC, and visual loss may persist even when the IOP is well-controlled. As such, discovering and creating IOP-independent neuroprotective strategies for safeguard RGCs is crucial for glaucoma management. Investigating and clarifying the mechanism behind RGC death to counteract its effects is a promising direction for glaucoma control. Empirical studies of glaucoma reveal the role of multiple regulated cell death (RCD) pathways in RGC death. This review delineates the RCD of RGCs following IOP elevation and optic nerve damage and discusses the substantial benefits of mitigating RCD in RGCs in preserving visual function.
Collapse
Affiliation(s)
- Wen-Juan Zhao
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Hunan Province, No. 172, Tongzipo Road, Yuelu District, Changsha City, 410013, China
| | - Chun-Ling Fan
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Hunan Province, No. 172, Tongzipo Road, Yuelu District, Changsha City, 410013, China
| | - Xi-Min Hu
- Department of Dermatology, Xiangya Hospital, Central South University, Hunan Province, No. 172, Tongzipo Road, Yuelu District, Changsha City, 410013, China
| | - Xiao-Xia Ban
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Hunan Province, No. 172, Tongzipo Road, Yuelu District, Changsha City, 410013, China
| | - Hao Wan
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Hunan Province, No. 172, Tongzipo Road, Yuelu District, Changsha City, 410013, China
| | - Ye He
- Changsha Aier Eye Hospital, Hunan Province, No. 188, Furong Road, Furong District, Changsha City, 410015, China
| | - Qi Zhang
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Hunan Province, No. 172, Tongzipo Road, Yuelu District, Changsha City, 410013, China.
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, 571199, China.
| | - Kun Xiong
- Department of Human Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Hunan Province, No. 172, Tongzipo Road, Yuelu District, Changsha City, 410013, China.
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, 571199, China.
- Hunan Key Laboratory of Ophthalmology, Changsha, 410013, China.
| |
Collapse
|
5
|
Saadh MJ, Castillo-Acobo RY, Baher H, Narayanan J, Palacios Garay JP, Yamaguchi MNV, Arias-Gonzáles JL, Cotrina-Aliaga JC, Akram SV, Lakshmaiya N, Amin AH, Mohany M, Al-Rejaie SS, Ahsan M, Bahrami A, Akhavan-Sigari R. The protective role of sulforaphane and Homer1a in retinal ischemia-reperfusion injury: Unraveling the neuroprotective interplay. Life Sci 2023; 329:121968. [PMID: 37487941 DOI: 10.1016/j.lfs.2023.121968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/10/2023] [Accepted: 07/18/2023] [Indexed: 07/26/2023]
Abstract
AIMS Retinal ischemia/reperfusion (I/R) injury is a common pathological basis for various ophthalmic diseases. This study aimed to investigate the potential of sulforaphane (SFN) and Homer1a in regulating cell apoptosis induced by retinal I/R injury and to explore the underlying regulatory mechanism between them. MATERIALS AND METHODS In in vivo experiments, C57BL/6J mice and Homer1flox/-/Homer1a+/-/Nestin-Cre+/- mice were used to construct retinal I/R injury models. In vitro experiments utilized the oxygen-glucose deprivation-reperfusion (OGD/R) injury model with primary retinal ganglion cells (RGCs). The effects of Homer1a and SFN on cell apoptosis were observed through pathological analyses, flow cytometry, and visual electrophysiological assessments. KEY FINDINGS We discovered that after OGD/R injury, apoptosis of RGCs and intracellular Ca2+ activity significantly increased. However, these changes were reversed upon the addition of SFN, and similar observations were reproduced in in vivo studies. Furthermore, both in vivo and in vitro studies confirmed the upregulation of Homer1a after I/R, which could be further enhanced by the administration of SFN. Moreover, upregulation of Homer1a resulted in a reduction in cell apoptosis and pro-apoptotic proteins, while downregulation of Homer1a had the opposite effect. Flash visual evoked potential, oscillatory potentials, and escape latency measurements in mice supported these findings. Furthermore, the addition of SFN strengthened the neuroprotective effects in the OGD/R + H+ group but weakened them in Homer1flox/-/Homer1a+/-/Nestin-Cre+/- mice. SIGNIFICANCE These results indicate that Homer1a plays a significant role in the therapeutic potential of sulforaphane for retinal I/R injury, thereby providing a theoretical basis for clinical treatment.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan; Applied Science Research Center, Applied Science Private University, Amman 11152, Jordan
| | | | - Hala Baher
- Department of Radiology and Ultrasonography Techniques, College of Medical Techniques, Al-Farahidi University, Baghdad, Iraq
| | | | | | | | - José Luis Arias-Gonzáles
- Department of Social Sciences, Faculty of Social Studies, University of British Columbia, BC, Canada
| | | | - Shaik Vaseem Akram
- Uttaranchal Institute of Technology, Division of research and Innovation, Uttaranchal University, Dehradun, India
| | - Natrayan Lakshmaiya
- Department of Mechanical Engineering, Saveetha School of Engineering, SRM Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Ali H Amin
- Zoology Department, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
| | - Mohamed Mohany
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 55760, Riyadh 11451, Saudi Arabia
| | - Salim S Al-Rejaie
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 55760, Riyadh 11451, Saudi Arabia
| | - Muhammad Ahsan
- Department of Measurements and Control Systems, Silesian University of Technology, Gliwice, 44-100, Poland; Joint Doctoral School, Silesian University of Technology, Akademicka 2A, Gliwice, 44-100, Poland.
| | - Abolfazl Bahrami
- Department of Cell Biology, Tuebingen University, Tuebingen, Germany; Biomedical Center for Systems Biology Science Munich, Ludwig-Maximilians-University, Munich, Germany.
| | - Reza Akhavan-Sigari
- Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw, Poland; Department of Neurosurgery, University Medical Center Tuebingen, Germany
| |
Collapse
|
6
|
Diering GH. Remembering and forgetting in sleep: Selective synaptic plasticity during sleep driven by scaling factors Homer1a and Arc. Neurobiol Stress 2022; 22:100512. [PMID: 36632309 PMCID: PMC9826981 DOI: 10.1016/j.ynstr.2022.100512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 12/01/2022] [Accepted: 12/29/2022] [Indexed: 01/02/2023] Open
Abstract
Sleep is a conserved and essential process that supports learning and memory. Synapses are a major target of sleep function and a locus of sleep need. Evidence in the literature suggests that the need for sleep has a cellular or microcircuit level basis, and that sleep need can accumulate within localized brain regions as a function of waking activity. Activation of sleep promoting kinases and accumulation of synaptic phosphorylation was recently shown to be part of the molecular basis for the localized sleep need. A prominent hypothesis in the field suggests that some benefits of sleep are mediated by a broad but selective weakening, or scaling-down, of synaptic strength during sleep in order to offset increased excitability from synaptic potentiation during wake. The literature also shows that synapses can be strengthened during sleep, raising the question of what molecular mechanisms may allow for selection of synaptic plasticity types during sleep. Here I describe mechanisms of action of the scaling factors Arc and Homer1a in selective plasticity and links with sleep need. Arc and Homer1a are induced in neurons in response to waking neuronal activity and accumulate with time spent awake. I suggest that during sleep, Arc and Homer1a drive broad weakening of synapses through homeostatic scaling-down, but in a manner that is sensitive to the plasticity history of individual synapses, based on patterned phosphorylation of synaptic proteins. Therefore, Arc and Homer1a may offer insights into the intricate links between a cellular basis of sleep need and memory consolidation during sleep.
Collapse
Affiliation(s)
- Graham H. Diering
- Department of Cell Biology and Physiology and the UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA,Carolina Institute for Developmental Disabilities, USA,111 Mason Farm Road, 5200 Medical and Biomolecular Research Building, Chapel Hill, NC, 27599-7545, USA.
| |
Collapse
|
7
|
Boccuni I, Fairless R. Retinal Glutamate Neurotransmission: From Physiology to Pathophysiological Mechanisms of Retinal Ganglion Cell Degeneration. Life (Basel) 2022; 12:638. [PMID: 35629305 PMCID: PMC9147752 DOI: 10.3390/life12050638] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/22/2022] [Accepted: 04/22/2022] [Indexed: 12/12/2022] Open
Abstract
Glutamate neurotransmission and metabolism are finely modulated by the retinal network, where the efficient processing of visual information is shaped by the differential distribution and composition of glutamate receptors and transporters. However, disturbances in glutamate homeostasis can result in glutamate excitotoxicity, a major initiating factor of common neurodegenerative diseases. Within the retina, glutamate excitotoxicity can impair visual transmission by initiating degeneration of neuronal populations, including retinal ganglion cells (RGCs). The vulnerability of RGCs is observed not just as a result of retinal diseases but has also been ascribed to other common neurodegenerative and peripheral diseases. In this review, we describe the vulnerability of RGCs to glutamate excitotoxicity and the contribution of different glutamate receptors and transporters to this. In particular, we focus on the N-methyl-d-aspartate (NMDA) receptor as the major effector of glutamate-induced mechanisms of neurodegeneration, including impairment of calcium homeostasis, changes in gene expression and signalling, and mitochondrial dysfunction, as well as the role of endoplasmic reticular stress. Due to recent developments in the search for modulators of NMDA receptor signalling, novel neuroprotective strategies may be on the horizon.
Collapse
Affiliation(s)
- Isabella Boccuni
- Institute for Physiology and Pathophysiology, Heidelberg University, 69120 Heidelberg, Germany
- Department of Neurology, University Clinic Heidelberg, 69120 Heidelberg, Germany;
| | - Richard Fairless
- Department of Neurology, University Clinic Heidelberg, 69120 Heidelberg, Germany;
- Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
8
|
Fei X, Dou YN, Wang L, Wu X, Huan Y, Wu S, He X, Lv W, Wei J, Fei Z. Homer1 promotes the conversion of A1 astrocytes to A2 astrocytes and improves the recovery of transgenic mice after intracerebral hemorrhage. J Neuroinflammation 2022; 19:67. [PMID: 35287697 PMCID: PMC8922810 DOI: 10.1186/s12974-022-02428-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/03/2022] [Indexed: 12/24/2022] Open
Abstract
Background Inflammation induced by intracerebral hemorrhage (ICH) is one of the main causes of the high mortality and poor prognosis of patients with ICH. A1 astrocytes are closely associated with neuroinflammation and neurotoxicity, whereas A2 astrocytes are neuroprotective. Homer scaffolding protein 1 (Homer1) plays a protective role in ischemic encephalopathy and neurodegenerative diseases. However, the role of Homer1 in ICH-induced inflammation and the effect of Homer1 on the phenotypic conversion of astrocytes remain unknown. Methods Femoral artery autologous blood from C57BL/6 mice was used to create an ICH model. We use the A1 phenotype marker C3 and A2 phenotype marker S100A10 to detect astrocyte conversion after ICH. Homer1 overexpression/knock-down mice were constructed by adeno-associated virus (AAV) infection to explore the role of Homer1 and its mechanism of action after ICH. Finally, Homer1 protein and selumetinib were injected into in situ hemorrhage sites in the brains of Homer1flox/flox/Nestin-Cre+/− mice to study the efficacy of Homer1 in the treatment of ICH by using a mouse cytokine array to explore the potential mechanism. Results The expression of Homer1 peaked on the third day after ICH and colocalized with astrocytes. Homer1 promotes A1 phenotypic conversion in astrocytes in vivo and in vitro. Overexpression of Homer1 inhibits the activation of MAPK signaling, whereas Homer1 knock-down increases the expression of pathway-related proteins. The Homer1 protein and selumetinib, a non-ATP competitive MEK1/2 inhibitor, improved the outcome in ICH in Homer1flox/flox/Nestin-Cre+/− mice. The efficacy of Homer1 in the treatment of ICH is associated with reduced expression of the inflammatory factor TNFSF10 and increased expression of the anti-inflammatory factors activin A, persephin, and TWEAK. Conclusions Homer1 plays an important role in inhibiting inflammation after ICH by suppressing the A1 phenotype conversion in astrocytes. In situ injection of Homer1 protein may be a novel and effective method for the treatment of inflammation after ICH.
Collapse
Affiliation(s)
- Xiaowei Fei
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, Xi'an, 710032, Shaanxi, China
| | - Ya-Nan Dou
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, Xi'an, 710032, Shaanxi, China
| | - Li Wang
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, Xi'an, 710032, Shaanxi, China
| | - Xiuquan Wu
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, Xi'an, 710032, Shaanxi, China
| | - Yu Huan
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, Xi'an, 710032, Shaanxi, China
| | - Shuang Wu
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, Xi'an, 710032, Shaanxi, China
| | - Xin He
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, Xi'an, 710032, Shaanxi, China
| | - Weihao Lv
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, Xi'an, 710032, Shaanxi, China
| | - Jialiang Wei
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, Xi'an, 710032, Shaanxi, China.
| | - Zhou Fei
- Department of Neurosurgery, Xijing Hospital, Air Force Military Medical University, No. 127, Changle West Road, Xincheng District, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
9
|
Wu XQ, Su N, Fei Z, Fei F. Homer signaling pathways as effective therapeutic targets for ischemic and traumatic brain injuries and retinal lesions. Neural Regen Res 2021; 17:1454-1461. [PMID: 34916418 PMCID: PMC8771115 DOI: 10.4103/1673-5374.330588] [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] [Indexed: 11/26/2022] Open
Abstract
Ischemic and traumatic insults to the central nervous system account for most serious acute and fatal brain injuries and are usually characterized by primary and secondary damage. Secondary damage presents the greatest challenge for medical staff; however, there are currently few effective therapeutic targets for secondary damage. Homer proteins are postsynaptic scaffolding proteins that have been implicated in ischemic and traumatic insults to the central nervous system. Homer signaling can exert either positive or negative effects during such insults, depending on the specific subtype of Homer protein. Homer 1b/c couples with other proteins to form postsynaptic densities, which form the basis of synaptic transmission, while Homer1a expression can be induced by harmful external factors. Homer 1c is used as a unique biomarker to reveal alterations in synaptic connectivity before and during the early stages of apoptosis in retinal ganglion cells, mediated or affected by extracellular or intracellular signaling or cytoskeletal processes. This review summarizes the structural features, related signaling pathways, and diverse roles of Homer proteins in physiological and pathological processes. Upregulating Homer1a or downregulating Homer1b/c may play a neuroprotective role in secondary brain injuries. Homer also plays an important role in the formation of photoreceptor synapses. These findings confirm the neuroprotective effects of Homer, and support the future design of therapeutic drug targets or gene therapies for ischemic and traumatic brain injuries and retinal disorders based on Homer proteins.
Collapse
Affiliation(s)
- Xiu-Quan Wu
- Department of Neurosurgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Ning Su
- Department of Radiation Oncology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Zhou Fei
- Department of Neurosurgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Fei Fei
- Department of Ophthalmology, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi Province, China
| |
Collapse
|
10
|
Wu X, Dou YN, Fei Z, Fei F. Parkin Prevents Glutamate Excitotoxicity Through Inhibiting NLRP3 Inflammasome in Retinal Ganglion Cells. Neuroscience 2021; 478:1-10. [PMID: 34600073 DOI: 10.1016/j.neuroscience.2021.09.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 09/18/2021] [Accepted: 09/22/2021] [Indexed: 12/26/2022]
Abstract
Glutamate excitotoxicity is one of the important pathophysiological culprits in retinal ganglion cells (RGCs) damage after acute optic nerve injury such as traumatic optic neuropathies and glaucoma. It is necessary to elucidate the mechanism of glutamate injury to RGCs in order to find the relevant neuroprotector. In this study, it was observed that the expression of Parkin increased and peaked at 24 h after glutamate injury to RGCs. Moreover, upregulating Parkin attenuated glutamate induced apoptosis, mitochondrial dysfunction and oxidative stress. And, it was found that Parkin could exert neuroprotective effects on RGCs by inhibiting nucleotide-binding domain leucine-rich repeat containing family pyrin domain containing 3 (NLRP3) inflammasome. Moreover, the genetic and pharmacological downregulation of NLRP3 improved survival of RGCs against glutamate excitotoxicity. In the end, knockdown of Parkin exacerbated glutamate induced RGCs damage via triggering NLRP3 inflammasome activation. Taken together, these results shed light on the promising molecular targets for the prevention and treatment of acute optic nerve injury.
Collapse
Affiliation(s)
- Xiuquan Wu
- Department of Neurosurgery, Xijing Hospital, the Fourth Military Medical University, 15 Changle West Road, Xi'an 71032, People's Republic of China
| | - Ya-Nan Dou
- Department of Neurosurgery, Xijing Hospital, the Fourth Military Medical University, 15 Changle West Road, Xi'an 71032, People's Republic of China
| | - Zhou Fei
- Department of Neurosurgery, Xijing Hospital, the Fourth Military Medical University, 15 Changle West Road, Xi'an 71032, People's Republic of China.
| | - Fei Fei
- Department of Ophthalmology, Xijing Hospital, the Fourth Military Medical University, 15 Changle West Road, Xi'an 71032, People's Republic of China.
| |
Collapse
|
11
|
Furuichi T, Muto Y, Sadakata T, Sato Y, Hayashi K, Shiraishi-Yamaguchi Y, Shinoda Y. The physiological role of Homer2a and its novel short isoform, Homer2e, in NMDA receptor-mediated apoptosis in cerebellar granule cells. Mol Brain 2021; 14:90. [PMID: 34118975 PMCID: PMC8199691 DOI: 10.1186/s13041-021-00804-8] [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: 04/21/2021] [Accepted: 06/04/2021] [Indexed: 11/10/2022] Open
Abstract
Homer is a postsynaptic scaffold protein, which has long and short isoforms. The long form of Homer consists of an N-terminal target-binding domain and a C-terminal multimerization domain, linking multiple proteins within a complex. The short form of Homer only has the N-terminal domain and likely acts as a dominant negative regulator. Homer2a, one of the long form isoforms of the Homer family, expresses with a transient peak in the early postnatal stage of mouse cerebellar granule cells (CGCs); however, the functions of Homer2a in CGCs are not fully understood yet. In this study, we investigated the physiological roles of Homer2a in CGCs using recombinant adenovirus vectors. Overexpression of the Homer2a N-terminal domain construct, which was made structurally reminiscent with Homer1a, altered NMDAR1 localization, decreased NMDA currents, and promoted the survival of CGCs. These results suggest that the Homer2a N-terminal domain acts as a dominant negative protein to attenuate NMDAR-mediated excitotoxicity. Moreover, we identified a novel short form N-terminal domain-containing Homer2, named Homer2e, which was induced by apoptotic stimulation such as ischemic brain injury. Our study suggests that the long and short forms of Homer2 are involved in apoptosis of CGCs.
Collapse
Affiliation(s)
- Teiichi Furuichi
- Laboratory for Molecular Neurogenesis, RIKEN Brain Science Institute, Wako, Saitama, 351-0198, Japan.
- JST-CREST, Kawaguchi, Saitama, 332-0012, Japan.
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Chiba, 278-8510, Japan.
| | - Yuko Muto
- Laboratory for Molecular Neurogenesis, RIKEN Brain Science Institute, Wako, Saitama, 351-0198, Japan
| | - Tetsushi Sadakata
- Laboratory for Molecular Neurogenesis, RIKEN Brain Science Institute, Wako, Saitama, 351-0198, Japan
- JST-CREST, Kawaguchi, Saitama, 332-0012, Japan
- Education and Research Support Center, Gunma University Graduate School of Medicine, Maebashi, Gunma, 371-8511, Japan
| | - Yumi Sato
- Laboratory for Molecular Neurogenesis, RIKEN Brain Science Institute, Wako, Saitama, 351-0198, Japan
- Laboratory of Proteome Research, Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, 567-0085, Japan
| | - Kanehiro Hayashi
- Laboratory for Molecular Neurogenesis, RIKEN Brain Science Institute, Wako, Saitama, 351-0198, Japan
- JST-CREST, Kawaguchi, Saitama, 332-0012, Japan
- Department of Anatomy, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Yoko Shiraishi-Yamaguchi
- Laboratory for Molecular Neurogenesis, RIKEN Brain Science Institute, Wako, Saitama, 351-0198, Japan
- Department of Developing Human Resources for R&D Programs, Japan Science and Technology Agency (JST), Chiyoda-ku, Tokyo, 102-8666, Japan
| | - Yo Shinoda
- Laboratory for Molecular Neurogenesis, RIKEN Brain Science Institute, Wako, Saitama, 351-0198, Japan.
- JST-CREST, Kawaguchi, Saitama, 332-0012, Japan.
- Department of Environmental Health, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, 192-0392, Japan.
| |
Collapse
|
12
|
Zehra A, Alshemmari H, Kavil YN, Majid Khan A, Zaffar Hashmi M. Effects of PCB70 and PCB75 on HeLa cell proliferation, membrane integrity and cell signaling pathway. ARAB J CHEM 2021. [DOI: 10.1016/j.arabjc.2020.102985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
13
|
Chen H, Li X, Ma H, Zheng W, Shen X. Reduction in Nesfatin-1 Levels in the Cerebrospinal Fluid and Increased Nigrostriatal Degeneration Following Ventricular Administration of Anti-nesfatin-1 Antibody in Mice. Front Neurosci 2021; 15:621173. [PMID: 33613183 PMCID: PMC7890421 DOI: 10.3389/fnins.2021.621173] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 01/11/2021] [Indexed: 12/19/2022] Open
Abstract
Nesfatin-1 is one of several brain-gut peptides that have a close relationship with the central dopaminergic system. Our previous studies have shown that nesfatin-1 is capable of protecting nigral dopaminergic neurons against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced neurotoxicity. A recent study also revealed a reduced blood level of nesfatin-1 in patients with Parkinson’s disease (PD). The current study was designed to investigate whether reduced nesfatin-1 in cerebrospinal fluid (CSF) induces nigrostriatal system degeneration. An intra-cerebroventricular (ICV) injection technique was used to administer anti-nesfatin-1 antibody directly into the lateral ventricle of the brain. Enzyme-linked immunosorbent assay (ELISA) results showed that ICV injection of anti-nesfatin-1 antibody into the lateral ventricle of the brain once daily for 2 weeks caused a significant reduction in nesfatin-1 levels in the CSF (93.1%). Treatment with anti-nesfatin-1 antibody resulted in a substantial loss (23%) of TH-positive (TH+) dopaminergic neurons in the substantia nigra pars compacta (SNpc), as shown by immunofluorescence staining, a depletion in dopamine and its metabolites in the striatum detected by high-performance liquid chromatography (HPLC), and obvious nuclear shrinkage and mitochondrial lesions in dopaminergic neurons in the SNpc detected by transmission electron microscopy (TEM). Furthermore, the results from our Western blot and ELISA experiments demonstrated that anti-nesfatin-1 antibody injection induced an upregulation of caspase-3 activation, increased the expression of p-ERK, and elevated brain-derived neurotrophic factor (BDNF) levels in the SNpc. Taken together, these observations suggest that reduced nesfatin-1 in the brain may induce nigrostriatal dopaminergic system degeneration; this effect may be mediated via mitochondrial dysfunction-related apoptosis. Our data support a role of nesfatin-1 in maintaining the normal physiological function of the nigrostriatal dopaminergic system.
Collapse
Affiliation(s)
- Huanhuan Chen
- Department of Epidemiology and Health Statistics, Medical School of Qingdao University, Qingdao, China
| | - Xuelian Li
- Department of Epidemiology and Health Statistics, Medical School of Qingdao University, Qingdao, China
| | - Hui Ma
- Department of Epidemiology and Health Statistics, Medical School of Qingdao University, Qingdao, China
| | - Wei Zheng
- School of Health Sciences, Purdue University, West Lafayette, IN, United States
| | - Xiaoli Shen
- Department of Epidemiology and Health Statistics, Medical School of Qingdao University, Qingdao, China.,School of Health Sciences, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
14
|
Fei F, Su N, Li X, Fei Z. Neuroprotection mediated by natural products and their chemical derivatives. Neural Regen Res 2020; 15:2008-2015. [PMID: 32394948 PMCID: PMC7716029 DOI: 10.4103/1673-5374.282240] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Neuronal injuries can lead to various diseases such as neurodegenerative diseases, stroke, trauma, ischemia and, more specifically, glaucoma and optic neuritis. The cellular mechanisms that regulate neuronal death include calcium influx and calcium overload, excitatory amino acid release, oxidative stress, inflammation and microglial activation. Much attention has been paid to the effective prevention and treatment of neuroprotective drugs by natural products. This review summarizes the neuroprotective aspects of natural products, extracted from Panax ginseng, Camellia sinensis, soy and some other plants, and some of their chemical derivatives. Their antioxidative and anti-inflammatory action and their inhibition of apoptosis and microglial activation are assessed. This will provide new directions for the development of novel drugs and strategies to treat neurodegenerative diseases.
Collapse
Affiliation(s)
- Fei Fei
- Department of Ophthalmology, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Ning Su
- Department of Radiation Oncology, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Xia Li
- Department of Neurosurgery, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Zhou Fei
- Department of Neurosurgery, Xijing Hospital, the Fourth Military Medical University, Xi'an, Shaanxi Province, China
| |
Collapse
|
15
|
Nagai N, Iwai Y, Deguchi S, Otake H, Kanai K, Okamoto N, Shimomura Y. Therapeutic Potential of a Combination of Magnesium Hydroxide Nanoparticles and Sericin for Epithelial Corneal Wound Healing. NANOMATERIALS 2019; 9:nano9050768. [PMID: 31109118 PMCID: PMC6567023 DOI: 10.3390/nano9050768] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 05/13/2019] [Accepted: 05/15/2019] [Indexed: 01/31/2023]
Abstract
We previously found the instillation of sericin to be useful as therapy for keratopathy with or without diabetes mellitus. In this study, we investigated whether a combination of solid magnesium hydroxide nanoparticles (MHN) enhances epithelial corneal wound healing by sericin using rabbits, normal rats and type 2 diabetes mellitus rats with debrided corneal epithelium (ex vivo and in vivo studies). Ophthalmic formulations containing sericin and MHN (N-Ser) were prepared using a bead mill method. The mean particle size of the N-Ser was 110.3 nm at the time of preparation, and 148.1 nm one month later. The instillation of N-Ser had no effect on the amount of lacrimal fluid in normal rabbits (in vivo), but the MHN in N-Ser was found to expand the intercellular space in ex vivo rat corneas. In addition, the instillation of N-Ser increased the phosphorylation of Extracellular Signal-regulated Kinase (ERK)1/2, a factor involved in cell adhesion and cell proliferation in the corneal epithelium, in comparison with the instillation of sericin alone. The combination with MHN enhanced epithelial corneal wound healing by sericin in rat debrided corneal epithelium (in vivo). This study provides significant information to prepare potent drugs to cure severe keratopathy, such as diabetic keratopathy.
Collapse
Affiliation(s)
- Noriaki Nagai
- Faculty of Pharmacy, Kindai University, 3-4-1 Kowakae, Higashi-Osaka, Osaka 577-8502, Japan.
| | - Yoshie Iwai
- Faculty of Pharmacy, Kindai University, 3-4-1 Kowakae, Higashi-Osaka, Osaka 577-8502, Japan.
| | - Saori Deguchi
- Faculty of Pharmacy, Kindai University, 3-4-1 Kowakae, Higashi-Osaka, Osaka 577-8502, Japan.
| | - Hiroko Otake
- Faculty of Pharmacy, Kindai University, 3-4-1 Kowakae, Higashi-Osaka, Osaka 577-8502, Japan.
| | - Kazutaka Kanai
- Department of Small Animal Internal Medicine, School of Veterinary Medicine, University of Kitasato, Towada, Aomori 034-8628, Japan.
| | - Norio Okamoto
- Okamoto Eye Clinic, 5-11-12-312 Izumicho, Suita, Osaka 564-0041, Japan.
| | - Yoshikazu Shimomura
- Department of Ophthalmology, Fuchu Hospital, 1-10-17 Hikocho, Izumi, Osaka 594-0076, Japan.
| |
Collapse
|
16
|
Wei J, Wu X, Luo P, Yue K, Yu Y, Pu J, Zhang L, Dai S, Han D, Fei Z. Homer1a Attenuates Endoplasmic Reticulum Stress-Induced Mitochondrial Stress After Ischemic Reperfusion Injury by Inhibiting the PERK Pathway. Front Cell Neurosci 2019; 13:101. [PMID: 30930751 PMCID: PMC6428733 DOI: 10.3389/fncel.2019.00101] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 02/27/2019] [Indexed: 12/17/2022] Open
Abstract
Homer1a is the short form of a scaffold protein that plays a protective role in many forms of stress. However, the role of Homer1a in cerebral ischemia/reperfusion (I/R) injury and its potential mechanism is still unknown. In this study, we found that Homer1a was upregulated by oxygen and glucose deprivation (OGD) and that overexpression of Homer1a alleviated OGD-induced lactate dehydrogenase (LDH) release and cell death in cultured cortical neurons. After OGD treatment, the overexpression of Homer1a preserved mitochondrial function, as evidenced by less cytochrome c release, less reactive oxygen species (ROS) production, less ATP and mitochondrial membrane potential (MMP) loss, less caspase-9 activation, and inhibition of endoplasmic reticulum (ER) stress confirmed by the decreased expression of phosphate-PKR-like ER Kinase (p-PERK)/PERK and phosphate- inositol-requiring enzyme 1 (p-IRE1)/IRE1 and immunofluorescence (IF) staining. In addition, mitochondrial protection of Homer1a was blocked by the ER stress activator Tunicamycin (TM) with a re-escalated ROS level, increasing ATP and MMP loss. Furthermore, Homer1a overexpression-induced mitochondrial stress attenuation was significantly reversed by activating the PERK pathway with TM and p-IRE1 inhibitor 3,5-dibromosalicylaldehyde (DBSA), as evidenced by increased cytochrome c release, increased ATP loss and a higher ROS level. However, activating the IRE1 pathway with TM and p-PERK inhibitor GSK2656157 showed little change in cytochrome c release and exhibited a moderate upgrade of ATP loss and ROS production in neurons. In summary, these findings demonstrated that Homer1a protects against OGD-induced injury by preserving mitochondrial function through inhibiting the PERK pathway. Our finding may reveal a promising target of protecting neurons from cerebral I/R injury.
Collapse
Affiliation(s)
- Jialiang Wei
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,Department of Health Services, Fourth Military Medical University, Xi'an, China
| | - Xiuquan Wu
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Peng Luo
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Kangyi Yue
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yang Yu
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jingnan Pu
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Lei Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Shuhui Dai
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Donghui Han
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhou Fei
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
17
|
ID2 protects retinal pigment epithelium cells from oxidative damage through p-ERK1/2/ID2/NRF2. Arch Biochem Biophys 2018; 650:1-13. [PMID: 29753724 DOI: 10.1016/j.abb.2018.05.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 05/07/2018] [Accepted: 05/09/2018] [Indexed: 12/19/2022]
Abstract
Age-related macular degeneration (AMD) is the leading cause of blindness during aging. The degeneration of retinal pigment epithelium (RPE) is the main pathologic characteristic of AMD. ID2 is a member of the Inhibitor of DNA binding proteins (ID) family and is involved in regulation of cell proliferation and differentiation. However, currently the role of ID2 in oxidative injury response in RPE cells remains unknown. Here we showed that oxidative stress increased ID2 expression in RPE cells. Knockdown of ID2 promoted cell apoptosis and increased ROS level in RPE cells that were subjected to oxidative damage. In addition, over-expression of ID2 attenuated the oxidative damage response in RPE cells. Mechanistically, ID2 protected RPE cells from oxidative damage through activating NRF2. Furthermore, phosphorylation of ERK1/2 positively regulated the protective function of ID2. Finally, we confirmed that the oxidative damage increased Id2 expression and over-expression of Id2 elevated Nrf2 expression in primary mouse RPE cells. Therefore, ID2 protects RPE cells from oxidative damage through the p-ERK1/2/ID2/NRF2 pathway. Our study contributes to a better understanding of the mechanisms underlying oxidative stress in AMD and may present a new strategy for AMD treatment.
Collapse
|
18
|
Instillation of Sericin Enhances Corneal Wound Healing through the ERK Pathway in Rat Debrided Corneal Epithelium. Int J Mol Sci 2018; 19:ijms19041123. [PMID: 29642540 PMCID: PMC5979510 DOI: 10.3390/ijms19041123] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 04/04/2018] [Accepted: 04/06/2018] [Indexed: 11/25/2022] Open
Abstract
Sericin is a major constituent of silk produced by silkworms. We previously found that the instillation of sericin enhanced the proliferation of corneal epithelial cells, and acted to promote corneal wound healing in both normal and diabetic model rats. However, the mechanisms by which sericin promotes the proliferation of corneal cells have not been established. In this study, we investigated the effects of sericin on Akt and ERK activation in a human corneal epithelial cell line (HCE-T cells) and rat debrided corneal epithelium. Although Akt phosphorylation was not detected following the treatment of HCE-T cells with sericin, ERK1/2 phosphorylation was enhanced. The growth of HCE-T cells treated with sericin was significantly increased, with the cell growth of sericin-treated HCE-T cells being 1.7-fold higher in comparison with vehicle-treated HCE-T cells. On the other hand, both of an ERK inhibitor U0126 (non-specific specific inhibitor) and SCH772984 (specific inhibitor) attenuated the enhanced cell growth by sericin, and the growth level in the case of co-treatment with sericin and ERK1/2 inhibitor was similar to that of cells treated with ERK1/2 inhibitor alone. In an in vivo study using rat debrided corneal epithelium, the corneal wound healing rate was enhanced by the instillation of sericin, and this enhancement was also attenuated by the instillation of U0126. In addition, the corneal wound healing rate in rats co-instilled with sericin and U0126 was similar to that following the instillation of U0126 alone. In conclusion, we found that the instillation of sericin enhanced cell proliferation via the activation of the MAPK/ERK pathway, resulting in the promotion of corneal wound healing in rat eyes. These findings provide significant information for designing further studies to develop potent corneal wound-healing drugs.
Collapse
|
19
|
Wu X, Luo P, Rao W, Dai S, Zhang L, Ma W, Pu J, Yu Y, Wang J, Fei Z. Homer1a Attenuates Hydrogen Peroxide-Induced Oxidative Damage in HT-22 Cells through AMPK-Dependent Autophagy. Front Neurosci 2018; 12:51. [PMID: 29479301 PMCID: PMC5811507 DOI: 10.3389/fnins.2018.00051] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 01/22/2018] [Indexed: 12/25/2022] Open
Abstract
Neuronal oxidative stress is involved in diverse neurological disorders. Homer1a, as an important member of the Homer family and localized at the postsynaptic density, is known to protect cells against oxidative injury. However, the exact neuroprotective mechanism of Homer1a has not been fully elucidated. Here, we found that Homer1a promoted cell viability and reduced H2O2-induced LDH release. The overexpression of Homer1a enhanced autophagy after H2O2 treatment, which was confirmed by increased expression of LC3II, Beclin-1, and greater autophagosome formation. In addition, we demonstrated that activating autophagy improved cell survival and reduced H2O2-induced oxidative stress and mitochondrial damage. Moreover, the autophagy inhibitor 3-MA partially prevented the protective effects of Homer1a against oxidative challenge. We also found that the upregulation of Homer1a after H2O2 treatment increased the phosphorylation of AMPK. Furthermore, the AMPK inhibitor compound C inhibited Homer1a-induced autophagy and abolished Homer1a-mediated neuroprotection. All the above data suggests that Homer1a confers protection against H2O2-induced oxidative damage via AMPK-dependent autophagy.
Collapse
Affiliation(s)
- Xiuquan Wu
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Peng Luo
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Wei Rao
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,Department of Neurosurgery, PLA Navy General Hospital, Beijing, China
| | - Shuhui Dai
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Lei Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Wenke Ma
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,Department of Neurosurgery, Baoji Center Hospital of Shanxi Province, Baoji, China
| | - Jingnan Pu
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yang Yu
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jiu Wang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhou Fei
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
20
|
Yao B, Wang S, Xiao P, Wang Q, Hea Y, Zhang Y. MAPK signaling pathways in eye wounds: Multifunction and cooperation. Exp Cell Res 2017; 359:10-16. [DOI: 10.1016/j.yexcr.2017.06.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 06/29/2017] [Indexed: 12/12/2022]
|
21
|
Qian F, Tang FR. Metabotropic Glutamate Receptors and Interacting Proteins in Epileptogenesis. Curr Neuropharmacol 2017; 14:551-62. [PMID: 27030135 PMCID: PMC4983745 DOI: 10.2174/1570159x14666160331142228] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 12/30/2015] [Accepted: 03/13/2016] [Indexed: 02/07/2023] Open
Abstract
Neurotransmitter and receptor systems are involved in different neurological and neuropsychological disorders such as Parkinson's disease, depression, Alzheimer’s disease and epilepsy. Recent advances in studies of signal transduction pathways or interacting proteins of neurotransmitter receptor systems suggest that different receptor systems may share the common signal transduction pathways or interacting proteins which may be better therapeutic targets for development of drugs to effectively control brain diseases. In this paper, we reviewed metabotropic glutamate receptors (mGluRs) and their related signal transduction pathways or interacting proteins in status epilepticus and temporal lobe epilepsy, and proposed some novel therapeutical drug targets for controlling epilepsy and epileptogenesis.
Collapse
Affiliation(s)
| | - Feng-Ru Tang
- Radiobiology Research Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore.
| |
Collapse
|
22
|
Hartsock MJ, Cho H, Wu L, Chen WJ, Gong J, Duh EJ. A Mouse Model of Retinal Ischemia-Reperfusion Injury Through Elevation of Intraocular Pressure. J Vis Exp 2016. [PMID: 27501124 DOI: 10.3791/54065] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Retinal ischemia-reperfusion (I/R) is a pathophysiological process contributing to cellular damage in multiple ocular conditions, including glaucoma, diabetic retinopathy, and retinal vascular occlusions. Rodent models of I/R injury are providing significant insights into mechanisms and treatment strategies for human I/R injury, especially with regard to neurodegenerative damage in the retinal neurovascular unit. Presented here is a protocol for inducing retinal I/R injury in mice through elevation of intraocular pressure (IOP). In this protocol, the ocular anterior chamber is cannulated with a needle, through which flows the drip of an elevated saline reservoir. Using this drip to raise IOP above systolic arterial blood pressure, a practitioner temporarily halts inner retinal blood flow (ischemia). When circulation is reinstated (reperfusion) by removal of the cannula, severe cellular damage ensues, resulting ultimately in retinal neurodegeneration. Recent studies demonstrate inflammation, vascular permeability, and capillary degeneration as additional elements of this model. Compared to alternative retinal I/R methodologies, such as retinal arterial ligation, retinal I/R injury by elevated IOP offers advantages in its anatomical specificity, experimental tractability, and technical accessibility, presenting itself as a valuable tool for examining neuronal pathogenesis and therapy in the retinal neurovascular unit.
Collapse
Affiliation(s)
- Matthew J Hartsock
- Department of Ophthalmology, School of Medicine, Johns Hopkins University
| | - Hongkwan Cho
- Department of Ophthalmology, School of Medicine, Johns Hopkins University
| | - Lijuan Wu
- Department of Ophthalmology, School of Medicine, Johns Hopkins University
| | - Wan-Ju Chen
- Department of Ophthalmology, School of Medicine, Johns Hopkins University
| | - Junsong Gong
- Department of Ophthalmology, School of Medicine, Johns Hopkins University
| | - Elia J Duh
- Department of Ophthalmology, School of Medicine, Johns Hopkins University;
| |
Collapse
|