1
|
Chen L, Zeng Z, Luo H, Xiao H, Zeng Y. The effects of CypA on apoptosis: potential target for the treatment of diseases. Appl Microbiol Biotechnol 2024; 108:28. [PMID: 38159118 DOI: 10.1007/s00253-023-12860-2] [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: 05/26/2023] [Revised: 10/20/2023] [Accepted: 10/25/2023] [Indexed: 01/03/2024]
Abstract
Cyclophilin A (CypA), the first member of cyclophilins, is distributed extensively in eukaryotic and prokaryotic cells, primarily localized in the cytoplasm. In addition to acting as an intracellular receptor for cyclosporin A (CSA), CypA plays a crucial role in diseases such as aging and tumorigenesis. Apoptosis, a form of programmed cell death, is able to balance the rate of cell viability and death. In this review, we focus on the effects of CypA on apoptosis and the relationship between specific mechanisms of CypA promoting or inhibiting apoptosis and diseases, including tumorigenesis, cardiovascular diseases, organ injury, and microbial infections. Notably, the process of CypA promoting or inhibiting apoptosis is closely related to disease development. Finally, future prospects for the association of CypA and apoptosis are discussed, and a comprehensive understanding of the effects of CypA on apoptosis in relation to diseases is expected to provide new insights into the design of CypA as a therapeutic target for diseases. KEY POINTS: • Understand the effect of CypA on apoptosis. • CypA affects apoptosis through specific pathways. • The effect of CypA on apoptosis is associated with a variety of disease processes.
Collapse
Affiliation(s)
- Li Chen
- Institute of Pathogenic Biology, Basic Medicine School, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang City, Hunan Province, 421001, People's Republic of China
| | - Zhuo Zeng
- Institute of Pathogenic Biology, Basic Medicine School, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang City, Hunan Province, 421001, People's Republic of China
| | - Haodang Luo
- Institute of Pathogenic Biology, Basic Medicine School, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang City, Hunan Province, 421001, People's Republic of China
| | - Hua Xiao
- Institute of Pathogenic Biology, Basic Medicine School, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang City, Hunan Province, 421001, People's Republic of China
| | - Yanhua Zeng
- Institute of Pathogenic Biology, Basic Medicine School, Hengyang Medical College, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang City, Hunan Province, 421001, People's Republic of China.
| |
Collapse
|
2
|
Haider T, Khan S, Bibi T, Zahra SA, Ali H, Din FU, Shah FA, Youn I, Seo EK. Daidzein ameliorates experimental traumatic brain injury-induced neurological symptoms by suppressing oxidative stress and apoptosis. J Biochem Mol Toxicol 2024; 38:e70019. [PMID: 39425453 DOI: 10.1002/jbt.70019] [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: 05/09/2024] [Revised: 08/08/2024] [Accepted: 10/02/2024] [Indexed: 10/21/2024]
Abstract
Traumatic brain injury (TBI) causes deficits in neurological function, induces pathological changes, and increases oxidative stress. The current investigation aimed to determine Daidzein's neuroprotective potential in experimental TBI. Initially, the HT-22 cell line exposed to H2O2 underwent in vitro examination, and the results showed that Daidzein had a neuroprotective effect evident from enhanced cell viability and decreased NO generation. Using three different Daidzein doses-1 mg/kg, 5 mg/kg, and 10 mg/kg-in the in vivo experiment, the potential of Daidzein was evaluated against TBI. The neurological severity score (NSS), kondziela's screen test, and elevated plus maze showed improvements after treatment with Daidzein manifested by decreased score, enhanced motor coordination, and anti-anxiety effects. Additionally, Daidzein improved mechanical allodynia and restored the breakdown of the blood-brain barrier. The FTIR spectral analysis showed restoration of the biochemical compositional changes. Furthermore, H & E and Toluidine blue staining revealed an improvement in the histopathological alterations. The RT-qPCR revealed an increase in mRNA expression level of Nrf2, HO-1, and Bcl-2 and the downregulation of Keap-1, Bax and Cleaved caspase-3 expressions. Thus, exhibiting its antioxidant and antiapoptotic potential. The RT-qPCR also manifested a decrease in mRNA expression of GFAP and Iba-1. Further immunohistochemistry results indicated Daidzein's antioxidant and antiapoptotic properties by upregulating Nrf2 and downregulating cleaved caspase-3. Daidzein also lowered the apoptosis index and improved neuronal survival evidenced by flow cytometric analysis. In addition to this, Daidzein notably increased the antioxidant enzyme levels and decreased the oxidative stress markers. The current study's findings point to the neuroprotective potential of the phytoestrogen Daidzein as it lessened neurological abnormalities, decreased oxidative stress, and lowered proapoptotic protein expression.
Collapse
Affiliation(s)
- Tehreem Haider
- Pharmacological Sciences Research Lab, Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Salman Khan
- Pharmacological Sciences Research Lab, Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Tehmina Bibi
- Pharmacological Sciences Research Lab, Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Sana Ali Zahra
- Pharmacological Sciences Research Lab, Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Hussain Ali
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Fakhar Ud Din
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Fawad Ali Shah
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Isoo Youn
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Korea
| | - Eun Kyoung Seo
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Korea
| |
Collapse
|
3
|
Suryavanshi P, Baule S, Glykys J. Trauma in Neonatal Acute Brain Slices Alters Calcium and Network Dynamics and Causes Calpain-Mediated Cell Death. eNeuro 2024; 11:ENEURO.0007-24.2024. [PMID: 38886064 PMCID: PMC11232372 DOI: 10.1523/eneuro.0007-24.2024] [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: 01/02/2024] [Revised: 05/07/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024] Open
Abstract
Preparing acute brain slices produces trauma that mimics severe penetrating brain injury. In neonatal acute brain slices, the spatiotemporal characteristics of trauma-induced calcium dynamics in neurons and its effect on network activity are relatively unknown. Using multiphoton laser scanning microscopy of the somatosensory neocortex in acute neonatal mouse brain slices (P8-12), we simultaneously imaged neuronal Ca2+ dynamics (GCaMP6s) and cytotoxicity (propidium iodide or PI) to determine the relationship between cytotoxic Ca2+ loaded neurons (GCaMP-filled) and cell viability at different depths and incubation times. PI+ cells and GCaMP-filled neurons were abundant at the surface of the slices, with an exponential decrease with depth. Regions with high PI+ cells correlated with elevated neuronal and neuropil Ca2+ The number of PI+ cells and GCaMP-filled neurons increased with prolonged incubation. GCaMP-filled neurons did not participate in stimulus-evoked or seizure-evoked network activity. Significantly, the superficial tissue, with a higher degree of trauma-induced injury, showed attenuated seizure-related neuronal Ca2+ responses. Calpain inhibition prevented the increase in PI+ cells and GCaMP-filled neurons in the deep tissue and during prolonged incubation times. Isoform-specific pharmacological inhibition implicated calpain-2 as a significant contributor to trauma-induced injury in acute slices. Our results show a calpain-mediated spatiotemporal relationship between cell death and aberrant neuronal Ca2+ load in acute neonatal brain slices. Also, we demonstrate that neurons in acute brain slices exhibit altered physiology depending on the degree of trauma-induced injury. Blocking calpains may be a therapeutic option to prevent acute neuronal death during traumatic brain injury in the young brain.
Collapse
Affiliation(s)
- Pratyush Suryavanshi
- Department of Pediatrics, The University of Iowa, Iowa City, Iowa 52241
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, Iowa 52241
| | - Samuel Baule
- Department of Pediatrics, The University of Iowa, Iowa City, Iowa 52241
- Departments of Biomedical Engineering, The University of Iowa, Iowa City, Iowa 52241
| | - Joseph Glykys
- Department of Pediatrics, The University of Iowa, Iowa City, Iowa 52241
- Iowa Neuroscience Institute, The University of Iowa, Iowa City, Iowa 52241
- Neurology, The University of Iowa, Iowa City, Iowa 52241
| |
Collapse
|
4
|
Qian Y, Li X, Li G, Liu H, Li Q, Liu X, Zhang Y, He Z, Zhao Y, Fan H. Astrocyte-Derived Exosomal miR-148a-3p Suppresses Neuroinflammation and Restores Neurological Function in Traumatic Brain Injury by Regulating the Microglial Phenotype. eNeuro 2024; 11:ENEURO.0336-23.2024. [PMID: 38272675 PMCID: PMC10860656 DOI: 10.1523/eneuro.0336-23.2024] [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: 08/31/2023] [Revised: 12/29/2023] [Accepted: 01/01/2024] [Indexed: 01/27/2024] Open
Abstract
Interactions between astrocytes and microglia play an important role in the regeneration and repair of traumatic brain injury (TBI), and exosomes are involved in cell-cell interactions. A TBI model was constructed in rats. Brain extract (Ext) was isolated 1 d after TBI. Astrocyte-derived exosomes were obtained by coculturing Ext with primary astrocytes, and the morphology of exosomes was observed by electron microscopy. The isolated exosomes were cocultured with microglia to observe phenotypic changes in M1 and M2 markers. Aberrant RNA expression was detected in necrotic brain tissue and edematous brain tissue. The role of miR-148a-3p in regulating microglial phenotype was explored by knocking down or overexpressing miR-148a-3p. Finally, the effect of miR-148a-3p on TBI was studied in a rat TBI model. Astrocyte-derived exosomes stimulated by Ext promoted the transition of microglia from the M1 phenotype to the M2 phenotype. MiR-148a-3p was highly expressed in TBI. Transfecting miR-148a-3p promoted the transition of microglia from the M1 phenotype to the M2 phenotype and inhibited the lipopolysaccharide-induced inflammatory response in pre-microglia. In a rat TBI model, miR-148a-3p significantly improved the modified neurological severity score and attenuated brain injury, which promoted the transition of microglia from the M1 phenotype to the M2 phenotype. MiR-148a-3p alleviated TBI by inhibiting the nuclear factor κB pathway. Astrocyte-derived exosomal miR-148a-3p regulates the microglial phenotype, inhibits neuroinflammation, and restores neurological function in TBI. These results provide new potential targets for the treatment of TBI.
Collapse
Affiliation(s)
- Yan Qian
- Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, Yunnan 655000, China
| | - Xin Li
- Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, Yunnan 655000, China
| | - Guiliang Li
- Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, Yunnan 655000, China
| | - Huali Liu
- Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, Yunnan 655000, China
| | - Qiaofen Li
- Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, Yunnan 655000, China
| | - Xia Liu
- Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, Yunnan 655000, China
| | - Yang Zhang
- Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, Yunnan 655000, China
| | - Zongying He
- Rehabilitation Medicine, Qujing No.1 Hospital, Qujing, Yunnan 655000, China
| | - Ying Zhao
- Rehabilitation Medicine, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650000, China
| | - Hong Fan
- Rehabilitation Medicine, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650000, China
| |
Collapse
|
5
|
Zhang C, Liu R, Chen M, Xu Y, Jin X, Shen B, Wang J. Autophagy inhibitors 3-MA and BAF may attenuate hippocampal neuronal necroptosis after global cerebral ischemia-reperfusion injury in male rats by inhibiting the interaction of the RIP3/AIF/CypA complex. J Neurosci Res 2024; 102:e25301. [PMID: 38361405 DOI: 10.1002/jnr.25301] [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: 04/23/2023] [Revised: 10/12/2023] [Accepted: 01/17/2024] [Indexed: 02/17/2024]
Abstract
Our previous study found that receptor interacting protein 3 (RIP3) and apoptosis-inducing factor (AIF) were involved in neuronal programmed necrosis during global cerebral ischemia-reperfusion (I/R) injury. Here, we further studied its downstream mechanisms and the role of the autophagy inhibitors 3-methyladenine (3-MA) and bafilomycin A1 (BAF). A 20-min global cerebral I/R injury model was constructed using the 4-vessel occlusion (4-VO) method in male rats. 3-MA and BAF were injected into the lateral ventricle 1 h before ischemia. Spatial and activation changes of proteins were detected by immunofluorescence (IF), and protein interaction was determined by immunoprecipitation (IP). The phosphorylation of H2AX (γ-H2AX) and activation of mixed lineage kinase domain-like protein (p-MLKL) occurred as early as 6 h after reperfusion. RIP3, AIF, and cyclophilin A (CypA) in the neurons after I/R injury were spatially overlapped around and within the nucleus and combined with each other after reperfusion. The survival rate of CA1 neurons in the 3-MA and BAF groups was significantly higher than that in the I/R group. Autophagy was activated significantly after I/R injury, which was partially inhibited by 3-MA and BAF. Pretreatment with both 3-MA and BAF almost completely inhibited nuclear translocation, spatial overlap, and combination of RIP3, AIF, and CypA proteins. These findings suggest that after global cerebral I/R injury, RIP3, AIF, and CypA translocated into the nuclei and formed the DNA degradation complex RIP3/AIF/CypA in hippocampal CA1 neurons. Pretreatment with autophagy inhibitors could reduce neuronal necroptosis by preventing the formation of the RIP3/AIF/CypA complex and its nuclear translocation.
Collapse
Affiliation(s)
- Chen Zhang
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Renhui Liu
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Mengmeng Chen
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yang Xu
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institutes, First Affiliated Hospital of Wannan Medical College, Wuhu, China
- Department of Neurology, First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Xiaoqin Jin
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Bing Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Jingye Wang
- Department of Neurology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
6
|
Hu Y, Liang Y, Tian H, Xu C, Yu D, Zhang P, Ye H, Li M. Microplitis bicoloratus bracovirus regulates cyclophilin A-apoptosis-inducing factor interaction to induce cell apoptosis in the insect immunosuppressive process. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2022; 110:e21877. [PMID: 35218062 PMCID: PMC9285338 DOI: 10.1002/arch.21877] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/28/2022] [Accepted: 02/02/2022] [Indexed: 05/27/2023]
Abstract
Microplitis bicoloratus bracovirus (MbBV) induces apoptosis in hemocytes of the host (Spodoptera litura) via the cyclophilin A (CypA)-mediated signaling pathway. However, the mechanisms underlying CypA-mediated signaling during apoptosis remain largely unknown. Therefore, in this study, we investigated how CypA and apoptosis-inducing factor (AIF) interact during MbBV-mediated apoptosis. Our findings showed that MbBV induces apoptosis through the CypA-AIF axis of insect immune suppression. In MbBV-infected Spli221 cells, both the expression of the cypa gene and the release of AIF from the mitochondria increased the number of apoptotic cells. CypA and AIF underwent concurrent cytoplasm-nuclear translocation. Conversely, blocking of AIF release from mitochondria not only inhibited the CypA-AIF interaction but also inhibited the cytoplasmic-nuclear translocation of AIF and CypA. Importantly, the survival of the apoptotic phenotype was significantly rescued in MbBV-infected Spli221 cells. In addition, we found that the cyclosporine A-mediated inhibition of CypA did not prevent the formation of the CypA and AIF complex; rather, this only suppressed genomic DNA fragmentation. In vitro experiments revealed direct molecular interactions between recombinant CypA and AIF. Taken together, our results demonstrate that the CypA-AIF interaction plays an important role in MbBV-induced innate immune suppression. This study will help to clarify aspects of insect immunological mechanisms and will be relevant to biological pest control.
Collapse
Affiliation(s)
- Yan Hu
- School of Life SciencesYunnan UniversityKunmingChina
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and RegulationsYunnan UniversityKunmingChina
| | - Ya‐Ping Liang
- School of Life SciencesYunnan UniversityKunmingChina
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and RegulationsYunnan UniversityKunmingChina
| | - Hang‐Yu Tian
- School of Life SciencesYunnan UniversityKunmingChina
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and RegulationsYunnan UniversityKunmingChina
| | - Cui‐Xian Xu
- School of Life SciencesYunnan UniversityKunmingChina
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and RegulationsYunnan UniversityKunmingChina
| | - Dan Yu
- School of Life SciencesYunnan UniversityKunmingChina
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and RegulationsYunnan UniversityKunmingChina
| | - Pan Zhang
- School of Life SciencesYunnan UniversityKunmingChina
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and RegulationsYunnan UniversityKunmingChina
| | - Hui Ye
- School of Life SciencesYunnan UniversityKunmingChina
- School of AgricultureYunnan UniversityKunmingChina
| | - Ming Li
- School of Life SciencesYunnan UniversityKunmingChina
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and RegulationsYunnan UniversityKunmingChina
| |
Collapse
|
7
|
Macks C, Jeong D, Lee JS. Therapeutic efficacy of rolipram delivered by PgP nanocarrier on secondary injury and motor function in a rat TBI model. Nanomedicine (Lond) 2022; 17:431-445. [PMID: 35184609 PMCID: PMC8905552 DOI: 10.2217/nnm-2021-0271] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 01/26/2022] [Indexed: 11/21/2022] Open
Abstract
Aim: To develop poly(lactide-co-glycolide)-graft-polyethylenimine (PgP) as a nanocarrier for the delivery of rolipram (Rm) and evaluate the therapeutic efficacy of Rm-loaded PgP (Rm-PgP) on secondary injury and motor function in a rat traumatic brain injury (TBI) model. Materials & methods: Rm-PgP was injected in the injured brain lesion immediately after TBI using a microinjection pump. Secondary injury pathologies such as inflammatory response, apoptosis and astrogliosis were assessed by histological analysis and functional recovery was assessed by assorted motor function tests. Results: Rm-PgP restored cyclic adenosine monophosphate level in the injured brain close to the sham level and Rm-PgP treatment reduced lesion volume, neuroinflammation and apoptosis and improved motor function at 7 days post-TBI. Conclusion: One single injection of Rm-PgP can be effective for acute mild TBI treatment.
Collapse
Affiliation(s)
- Christian Macks
- Department of Bioengineering, Drug Design, Development, & Delivery (4D) Laboratory, Clemson University, Clemson, SC 29634, USA
| | - Daun Jeong
- Department of Bioengineering, Drug Design, Development, & Delivery (4D) Laboratory, Clemson University, Clemson, SC 29634, USA
| | - Jeoung Soo Lee
- Department of Bioengineering, Drug Design, Development, & Delivery (4D) Laboratory, Clemson University, Clemson, SC 29634, USA
| |
Collapse
|
8
|
Relevance of AIF/CypA Lethal Pathway in SH-SY5Y Cells Treated with Staurosporine. Int J Mol Sci 2021; 23:ijms23010265. [PMID: 35008690 PMCID: PMC8745523 DOI: 10.3390/ijms23010265] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/22/2021] [Accepted: 12/26/2021] [Indexed: 11/18/2022] Open
Abstract
The AIF/CypA complex exerts a lethal activity in several rodent models of acute brain injury. Upon formation, it translocates into the nucleus of cells receiving apoptotic stimuli, inducing chromatin condensation, DNA fragmentation, and cell death by a caspase-independent mechanism. Inhibition of this complex in a model of glutamate-induced cell death in HT-22 neuronal cells by an AIF peptide (AIF(370-394)) mimicking the binding site on CypA, restores cell survival and prevents brain injury in neonatal mice undergoing hypoxia-ischemia without apparent toxicity. Here, we explore the effects of the peptide on SH-SY5Y neuroblastoma cells stimulated with staurosporine (STS), a cellular model widely used to study Parkinson’s disease (PD). This will pave the way to understanding the role of the complex and the potential therapeutic efficacy of inhibitors in PD. We find that AIF(370-394) confers resistance to STS-induced apoptosis in SH-SY5Y cells similar to that observed with CypA silencing and that the peptide works on the AIF/CypA translocation pathway and not on caspases activation. These findings suggest that the AIF/CypA complex is a promising target for developing novel therapeutic strategies against PD.
Collapse
|
9
|
Wu L, Chung JY, Cao T, Jin G, Edmiston WJ, Hickman S, Levy ES, Whalen JA, Abrams ESL, Degterev A, Lo EH, Tozzi L, Kaplan DL, El Khoury J, Whalen MJ. Genetic inhibition of RIPK3 ameliorates functional outcome in controlled cortical impact independent of necroptosis. Cell Death Dis 2021; 12:1064. [PMID: 34753914 PMCID: PMC8578385 DOI: 10.1038/s41419-021-04333-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 09/20/2021] [Accepted: 10/04/2021] [Indexed: 02/05/2023]
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability with no specific effective therapy, in part because disease driving mechanisms remain to be elucidated. Receptor interacting protein kinases (RIPKs) are serine/threonine kinases that assemble multi-molecular complexes that induce apoptosis, necroptosis, inflammasome and nuclear factor kappa B activation. Prior studies using pharmacological inhibitors implicated necroptosis in the pathogenesis of TBI and stroke, but these studies cannot be used to conclusively demonstrate a role for necroptosis because of the possibility of off target effects. Using a model of cerebral contusion and RIPK3 and mixed lineage kinase like knockout (MLKL-/-) mice, we found evidence for activation of RIPK3 and MLKL and assembly of a RIPK1-RIPK3-MLKL necrosome complex in pericontusional brain tissue. Phosphorylated forms of RIPK3 and MLKL were detected in endothelium, CD11b + immune cells, and neurons, and RIPK3 was upregulated and activated in three-dimensional human endothelial cell cultures subjected to CCI. RIPK3-/- and MLKL-/- mice had reduced blood-brain barrier damage at 24 h (p < 0.05), but no differences in neuronal death (6 h, p = ns in CA1, CA3 and DG), brain edema (24 h, p = ns), or lesion size (4 weeks, p = ns) after CCI. RIPK3-/-, but not MLKL-/- mice, were protected against postinjury motor and cognitive deficits at 1-4 weeks (RIPK3-/- vs WT: p < 0.05 for group in wire grip, Morris water maze hidden platform trials, p < 0.05 for novel object recognition test, p < 0.01 for rotarod test). RIPK3-/- mice had reduced infiltrating leukocytes (p < 0.05 vs WT in CD11b + cells, microglia and macrophages), HMGB1 release and interleukin-1 beta activation at 24-48 h (p < 0.01) after CCI. Our data indicate that RIPK3 contributes to functional outcome after cerebral contusion by mechanisms involving inflammation but independent of necroptosis.
Collapse
Affiliation(s)
- Limin Wu
- grid.38142.3c000000041936754XDepartment of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
| | - Joon Yong Chung
- grid.38142.3c000000041936754XDepartment of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
| | - Tian Cao
- grid.38142.3c000000041936754XDepartment of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA ,grid.13291.380000 0001 0807 1581Department of Neurology, West China Hospital, Sichuan University, 610041 Chengdu, Sichuan China
| | - Gina Jin
- grid.38142.3c000000041936754XDepartment of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
| | - William J. Edmiston
- grid.38142.3c000000041936754XDepartment of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
| | - Suzanne Hickman
- grid.32224.350000 0004 0386 9924Department of Medicine, Center for Immunology and Inflammatory Disease, Massachusetts General Hospital, Boston, USA
| | - Emily S. Levy
- grid.38142.3c000000041936754XDepartment of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
| | - Jordyn A. Whalen
- grid.38142.3c000000041936754XDepartment of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
| | - Eliza Sophie LaRovere Abrams
- grid.38142.3c000000041936754XDepartment of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
| | - Alexei Degterev
- grid.67033.310000 0000 8934 4045Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA USA
| | - Eng H. Lo
- grid.32224.350000 0004 0386 9924Department of Radiology, Massachusetts General Hospital, Boston, MA 02115 USA ,grid.32224.350000 0004 0386 9924Department of Neurology, Massachusetts General Hospital, Boston, MA 02115 USA
| | - Lorenzo Tozzi
- grid.429997.80000 0004 1936 7531Department of Biomedical Engineering, Tufts University, Medford, MA 02155 USA
| | - David L. Kaplan
- grid.429997.80000 0004 1936 7531Department of Biomedical Engineering, Tufts University, Medford, MA 02155 USA
| | - Joseph El Khoury
- grid.32224.350000 0004 0386 9924Department of Medicine, Center for Immunology and Inflammatory Disease, Massachusetts General Hospital, Boston, USA
| | - Michael J. Whalen
- grid.38142.3c000000041936754XDepartment of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
| |
Collapse
|
10
|
Dhani S, Zhao Y, Zhivotovsky B. A long way to go: caspase inhibitors in clinical use. Cell Death Dis 2021; 12:949. [PMID: 34654807 PMCID: PMC8519909 DOI: 10.1038/s41419-021-04240-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 09/15/2021] [Accepted: 09/28/2021] [Indexed: 12/19/2022]
Abstract
Caspases are an evolutionary conserved family of cysteine-dependent proteases that are involved in many vital cellular processes including apoptosis, proliferation, differentiation and inflammatory response. Dysregulation of caspase-mediated apoptosis and inflammation has been linked to the pathogenesis of various diseases such as inflammatory diseases, neurological disorders, metabolic diseases, and cancer. Multiple caspase inhibitors have been designed and synthesized as a potential therapeutic tool for the treatment of cell death-related pathologies. However, only a few have progressed to clinical trials because of the consistent challenges faced amongst the different types of caspase inhibitors used for the treatment of the various pathologies, namely an inadequate efficacy, poor target specificity, or adverse side effects. Importantly, a large proportion of this failure lies in the lack of understanding various caspase functions. To overcome the current challenges, further studies on understanding caspase function in a disease model is a fundamental requirement to effectively develop their inhibitors as a treatment for the different pathologies. Therefore, the present review focuses on the descriptive properties and characteristics of caspase inhibitors known to date, and their therapeutic application in animal and clinical studies. In addition, a brief discussion on the achievements, and current challenges faced, are presented in support to providing more perspectives for further development of successful therapeutic caspase inhibitors for various diseases.
Collapse
Affiliation(s)
- Shanel Dhani
- Institute of Environmental Medicine, Karolinska Institutet, Box 210, 17177, Stockholm, Sweden
| | - Yun Zhao
- Institute of Environmental Medicine, Karolinska Institutet, Box 210, 17177, Stockholm, Sweden
| | - Boris Zhivotovsky
- Institute of Environmental Medicine, Karolinska Institutet, Box 210, 17177, Stockholm, Sweden.
- Faculty of Medicine, MV Lomonosov Moscow State University, 119991, Moscow, Russia.
| |
Collapse
|
11
|
Inampudi C, Ciccotosto GD, Cappai R, Crack PJ. Genetic Modulators of Traumatic Brain Injury in Animal Models and the Impact of Sex-Dependent Effects. J Neurotrauma 2021; 37:706-723. [PMID: 32027210 DOI: 10.1089/neu.2019.6955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Traumatic brain injury (TBI) is a major health problem causing disability and death worldwide. There is no effective treatment, due in part to the complexity of the injury pathology and factors affecting its outcome. The extent of brain injury depends on the type of insult, age, sex, lifestyle, genetic risk factors, socioeconomic status, other co-injuries, and underlying health problems. This review discusses the genes that have been directly tested in TBI models, and whether their effects are known to be sex-dependent. Sex differences can affect the incidence, symptom onset, pathology, and clinical outcomes following injury. Adult males are more susceptible at the acute phase and females show greater injury in the chronic phase. TBI is not restricted to a single sex; despite variations in the degree of symptom onset and severity, it is important to consider both female and male animals in TBI pre-clinical research studies.
Collapse
Affiliation(s)
- Chaitanya Inampudi
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia
| | - Giuseppe D Ciccotosto
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia
| | - Roberto Cappai
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia
| | - Peter J Crack
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
12
|
Zhang B, Yang M, Yan Q, Xu X, Niu F, Dong J, Zhuang Y, Lu S, Ge Q, Liu B. The Dual Dose-Dependent Effects of Corticosterone on Hippocampal Cell Apoptosis After Traumatic Brain Injury Depend on the Activation Ratio of Mineralocorticoid Receptors to Glucocorticoid Receptors. Front Pharmacol 2021; 12:713715. [PMID: 34381366 PMCID: PMC8350576 DOI: 10.3389/fphar.2021.713715] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/13/2021] [Indexed: 01/11/2023] Open
Abstract
In our recent studies, we reported that mineralocorticoid receptor (MR) had the opposite effects of glucocorticoid receptor (GR) on neural cell survival after traumatic brain injury (TBI). However, whether short-term use of high-dose natural glucocorticoids, which are mixed agonists of both MR and GR, leads to neurotoxic effects by inducing excessive GR activation is unclear, as is the threshold GR activation level and the possible signaling pathways remain unclear. In this study, we examined the dual dose-dependent effects of corticosterone (CORT) on spatial memory, hippocampal cell survival and receptor-mediated downstream signaling pathways after TBI. We found that different doses of CORT exhibited dual effects on hippocampal cell survival and rat spatial memory. Low doses of CORT (0.3 and 3 mg/kg) significantly increased MR activation, upregulated Akt/CREB/Bad phosphorylation and Bcl-2 concentration, reduced the number of apoptotic neural cells, and subsequently improved rat spatial memory. In contrast, a high dose of CORT (30 mg/kg) exerted the opposite effects by overactivating GR, upregulating P53/Bax levels, and inhibiting Erk/CREB activity. The results suggest that the neuroprotective and neurotoxic effects of endogenous GC depend on a threshold level and that a higher dose of GC, even for short-term use, should be avoided after TBI.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Mengshi Yang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Qiongyu Yan
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiaojian Xu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Fei Niu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Jinqian Dong
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yuan Zhuang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shenghua Lu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Qianqian Ge
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Baiyun Liu
- Department of Neurosurgery and Beijing Key Laboratory of Central Nervous System Injury, Beijing Tiantan Hospital and Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Nerve Injury and Repair Center of Beijing Institute for Brain Disorders, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| |
Collapse
|
13
|
Zhang Y, Gliyazova NS, Li PA, Ibeanu G. Phenoxythiophene sulfonamide compound B355252 protects neuronal cells against glutamate-induced excitotoxicity by attenuating mitochondrial fission and the nuclear translocation of AIF. Exp Ther Med 2021; 21:221. [PMID: 33603830 PMCID: PMC7851598 DOI: 10.3892/etm.2021.9652] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 12/03/2020] [Indexed: 01/03/2023] Open
Abstract
Glutamate neurotoxicity has been implicated in the initiation and progression of various neurological and neurodegenerative disorders. Therefore, it is necessary to develop therapeutics for the treatment of patients with these devastating diseases. Mitochondrial fission plays an import role in the mediation of cell death and survival. The objective of the present study was to determine whether B355252, a phenoxythiophene sulfonamide derivative, reduces glutamate-induced cell death by inhibiting mitochondrial fission and the nuclear translocation of apoptosis-inducing factor (AIF) in glutamate-challenged HT22 neuronal cells. The results revealed that glutamate treatment led to large increases in the mitochondrial levels of the major fission proteins dynamin-related protein 1 (Drp1) and mitochondrial fission 1 protein (Fis1), but only small elevations in the fusion proteins mitofusin 1 and 2 (Mfn1/2) and optic atrophy 1 (Opa1). In addition, glutamate toxicity disrupted mitochondrial reticular networks and increased the translocation of AIF to the nucleus. Pretreatment with B35525 reduced glutamate-induced cell death and prevented the increases in the protein levels of Drp1, Fis1, Mfn1/2 and Opa1 in the mitochondrial fraction. More importantly, the architecture of the mitochondria was protected and nuclear translocation of AIF was completely inhibited by B35525. These findings suggest that the regulation of mitochondrial dynamics is central to the neuroprotective properties of B355252, and presents an attractive opportunity for potential development as a therapy for neurodegenerative disorders associated with mitochondria dysfunction.
Collapse
Affiliation(s)
- Yuxin Zhang
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University, Durham, NC 27707, USA.,Institute of Clinical Pharmacology, Department of Pharmacy, General Hospital of Ningxia Medical University, Ningxia 750004, P.R. China.,School of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Nailya S Gliyazova
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University, Durham, NC 27707, USA
| | - P Andy Li
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University, Durham, NC 27707, USA
| | - Gordon Ibeanu
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University, Durham, NC 27707, USA
| |
Collapse
|
14
|
Hummel R, Ulbrich S, Appel D, Li S, Hirnet T, Zander S, Bobkiewicz W, Gölz C, Schäfer MK. Administration of all-trans retinoic acid after experimental traumatic brain injury is brain protective. Br J Pharmacol 2020; 177:5208-5223. [PMID: 32964418 PMCID: PMC7588818 DOI: 10.1111/bph.15259] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 09/02/2020] [Accepted: 09/04/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE All-trans retinoic acid (ATRA) is a vitamin A metabolite, important in the developing and mature brain. Pre-injury ATRA administration ameliorates ischaemic brain insults in rodents. This study examined the effects of post-traumatic ATRA treatment in experimental traumatic brain injury (TBI). EXPERIMENTAL APPROACH Male adult mice were subjected to the controlled cortical impact model of TBI or sham procedure and killed at 7 or 30 days post-injury (dpi). ATRA (10 mg kg-1, i.p.) was given immediately after the injury and 1, 2 and 3 dpi. Neurological function and sensorimotor coordination were evaluated. Brains were processed for (immuno-) histological, mRNA and protein analyses (qPCR and western blot). KEY RESULTS ATRA treatment reduced brain lesion size, reactive astrogliosis and axonal injury at 7 dpi, and hippocampal granule cell layer (GCL) integrity was protected at 7 and 30 dpi, independent of cell proliferation in neurogenic niches and blood-brain barrier damage. Neurological and motor deficits over time and the brain tissue loss at 30 dpi were not affected by ATRA treatment. ATRA decreased gene expression of markers for damage-associated molecular pattern (HMGB1), apoptosis (caspase-3 and Bax), activated microglia (TSPO), and reactive astrogliosis (GFAP, SerpinA3N) at 7 dpi and a subset of markers at 30 dpi (TSPO, GFAP). CONCLUSION AND IMPLICATIONS In experimental TBI, post-traumatic ATRA administration exerted brain protective effects, including long-term protection of GCL integrity, but did not affect neurological and motor deficits. Further investigations are required to optimize treatment regimens to enhance ATRA's brain protective effects and improve outcomes.
Collapse
Affiliation(s)
- Regina Hummel
- Department of AnesthesiologyUniversity Medical Center, Johannes Gutenberg‐University MainzMainzGermany
| | - Sebastian Ulbrich
- Department of AnesthesiologyUniversity Medical Center, Johannes Gutenberg‐University MainzMainzGermany
| | - Dominik Appel
- Department of AnesthesiologyUniversity Medical Center, Johannes Gutenberg‐University MainzMainzGermany
| | - Shuailong Li
- Department of AnesthesiologyUniversity Medical Center, Johannes Gutenberg‐University MainzMainzGermany
| | - Tobias Hirnet
- Department of AnesthesiologyUniversity Medical Center, Johannes Gutenberg‐University MainzMainzGermany
| | - Sonja Zander
- Department of AnesthesiologyUniversity Medical Center, Johannes Gutenberg‐University MainzMainzGermany
| | - Wieslawa Bobkiewicz
- Department of AnesthesiologyUniversity Medical Center, Johannes Gutenberg‐University MainzMainzGermany
| | - Christina Gölz
- Department of AnesthesiologyUniversity Medical Center, Johannes Gutenberg‐University MainzMainzGermany
| | - Michael K.E. Schäfer
- Department of AnesthesiologyUniversity Medical Center, Johannes Gutenberg‐University MainzMainzGermany
- Focus Program Translational Neurosciences (FTN)Johannes Gutenberg‐University MainzMainzGermany
- Research Center for ImmunotherapyUniversity Medical Center, Johannes Gutenberg‐University MainzMainzGermany
| |
Collapse
|
15
|
Zhang B, Bai M, Xu X, Yang M, Niu F, Gao F, Liu B. Corticosteroid receptor rebalancing alleviates critical illness-related corticosteroid insufficiency after traumatic brain injury by promoting paraventricular nuclear cell survival via Akt/CREB/BDNF signaling. J Neuroinflammation 2020; 17:318. [PMID: 33100225 PMCID: PMC7586672 DOI: 10.1186/s12974-020-02000-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/16/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND We previously found that high-dose methylprednisolone increased the incidence of critical illness-related corticosteroid insufficiency (CIRCI) and mortality in rats with traumatic brain injury (TBI), whereas low-dose hydrocortisone but not methylprednisolone exerted protective effects. However, the receptor-mediated mechanism remains unclear. This study investigated the receptor-mediated mechanism of the opposite effects of different glucocorticoids on the survival of paraventricular nucleus (PVN) cells and the incidence of CIRCI after TBI. METHODS Based on controlled cortical impact (CCI) and treatments, male SD rats (n = 300) were randomly divided into the sham, CCI, CCI + GCs (methylprednisolone 1 or 30 mg/kg/day; corticosterone 1 mg/kg/day), CCI + methylprednisolone+RU486 (RU486 50 mg/kg/day), and CCI + corticosterone+spironolactone (spironolactone 50 mg/kg/day) groups. Blood samples were collected 7 days before and after CCI. Brain tissues were collected on postinjury day 7 and processed for histology and western blot analysis. RESULTS We examined the incidence of CIRCI, mortality, apoptosis in the PVN, the receptor-mediated mechanism, and downstream signaling pathways on postinjury day 7. We found that methylprednisolone and corticosterone exerted opposite effects on the survival of PVN cells and the incidence of CIRCI by activating different receptors. High-dose methylprednisolone increased the nuclear glucocorticoid receptor (GR) level and subsequently increased cell loss in the PVN and the incidence of CIRCI. In contrast, low-dose corticosterone but not methylprednisolone played a protective role by upregulating mineralocorticoid receptor (MR) activation. The possible downstream receptor signaling mechanism involved the differential effects of GR and MR on the activity of the Akt/CREB/BDNF pathway. CONCLUSION The excessive activation of GR by high-dose methylprednisolone exacerbated apoptosis in the PVN and increased CIRCI. In contrast, refilling of MR by corticosterone protects PVN neurons and reduces the incidence of CIRCI by promoting GR/MR rebalancing after TBI.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Miao Bai
- Department of Neurology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiaojian Xu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Mengshi Yang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Fei Niu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Fei Gao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Baiyun Liu
- Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, No.119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, People's Republic of China.
- Nerve Injury and Repair Center of Beijing Institute for Brain Disorders, Beijing, China.
- China National Clinical Research Center for Neurological Diseases, Beijing, China.
| |
Collapse
|
16
|
Monti A, Sturlese M, Caporale A, Roger JDA, Mascanzoni F, Ruvo M, Doti N. Design, synthesis, structural analysis and biochemical studies of stapled AIF(370-394) analogues as ligand of CypA. Biochim Biophys Acta Gen Subj 2020; 1864:129717. [PMID: 32861757 DOI: 10.1016/j.bbagen.2020.129717] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/28/2020] [Accepted: 08/19/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND The neuronal apoptotic process requires the nuclear translocation of Apoptosis Inducing Factor (AIF) in complex with Cyclophilin A (CypA) with consequent chromatin condensation and DNA degradation events. Targeting CypA by delivering an AIF-blocking peptide (AIF(370-394)) provides a significant neuroprotection, demonstrating the biological relevance of the AIF/CypA complex. To date pharmaceutical compounds targeting this complex are missing. METHODS We designed and synthesized a set of mono and bicyclic AIF(370-394) analogs containing both disulfide and 1,2,3-triazole bridges, in the attempt to both stabilize the peptide conformation and improve its binding affinity to CypA. Peptide structures in solution and in complex with CypA have been studied by circular dichroism (CD), Nuclear Magnetic Resonance (NMR) and molecular modeling. The ability of stapled peptides to interact with CypA was evaluated by using Epic Corning label free technique and Isothermal Titration Calorimetry experiments. RESULTS We identified a stapled peptide analogue of AIF(370-394) with a ten-fold improved affinity for CypA. Molecular modeling studies reveal that the new peptide acquires β-turn/β-fold structures and shares with the parent molecule the same binding region on CypA. CONCLUSIONS Data obtained provide invaluable assistance in designing new ligand of CypA for therapeutic approaches in neurodegenerative diseases. GENERAL SIGNIFICANCE Due to the crucial role of AIF/CypA complex formation in neurodegeneration, identification of selective inhibitors is of high importance for targeted therapies. We describe new bicyclic peptide inhibitors with improved affinity for CypA, investigating the kinetic, thermodynamic and structural effects of conformational constraints on the protein-ligand interaction, and their utility for drug design.
Collapse
Affiliation(s)
- Alessandra Monti
- Istituto di Biostrutture e Bioimmagini-CNR; Via Mezzocannone, 16, 80134 Napoli, Italy; DISTABIF, Università degli Studi della Campania Luigi Vanvitelli, Via Vivaldi 43, 81100 Caserta, CE, Italy
| | - Mattia Sturlese
- Molecular Modeling Section, Dipartimento di Scienze del Farmaco, Università di Padova, via F. Marzolo 5, 35131 Padova, Italy
| | - Andrea Caporale
- Istituto di Biostrutture e Bioimmagini-CNR; Via Mezzocannone, 16, 80134 Napoli, Italy
| | - Jessica De Almeida Roger
- Molecular Modeling Section, Dipartimento di Scienze del Farmaco, Università di Padova, via F. Marzolo 5, 35131 Padova, Italy
| | - Fabiola Mascanzoni
- Istituto di Biostrutture e Bioimmagini-CNR; Via Mezzocannone, 16, 80134 Napoli, Italy
| | - Menotti Ruvo
- Istituto di Biostrutture e Bioimmagini-CNR; Via Mezzocannone, 16, 80134 Napoli, Italy
| | - Nunzianna Doti
- Istituto di Biostrutture e Bioimmagini-CNR; Via Mezzocannone, 16, 80134 Napoli, Italy.
| |
Collapse
|
17
|
Liu J, Zhao X, Lv Z, Guo M, Li C. Cyclophilin A mediates coelomocyte apoptosis via the NF-κB/Bcl-2 signaling pathway in Apostichopus japonicus. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 107:103657. [PMID: 32089518 DOI: 10.1016/j.dci.2020.103657] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 02/13/2020] [Accepted: 02/14/2020] [Indexed: 06/10/2023]
Abstract
As a multifunctional protein, cyclophilin A (CypA) plays an important role in cell apoptosis. In our previous work, we found that CypA from Apostichopus japonicus (AjCypA), as a cofactor, could modulate nuclear translocation of NF-κB. However, the immune function of AjCypA is largely unknown. In the present study, we found that siRNA-mediated AjCypA knockdown in vivo significantly increased the coelomocyte apoptosis rate. In addition, the expression of B-cell lymphoma-2 (AjBcl-2, an anti-apoptosis gene) was synchronously downregulated. To better understand the connection between AjCypA and AjBcl-2 expression, we cloned the promoter of AjBcl-2 via genomic walking, which spanned 1870 bp and contained four potential binding sites of NF-κB. Dual-luciferase reporter assay revealed that the full-length sequence and all truncated fragments exhibited high transcriptional activity. Moreover, 1 μg/mL LPS exposure significantly increased the luciferase activity of P1 (-1870/+57) by 2.31-fold and 3.15-fold at 12 and 24 h, respectively. Furthermore, the four potential NF-κB binding sites and pCMV-Flag2C-AjNF-κB co-transfection assay demonstrated that NF-κB could regulate the expression of AjBcl-2 via the NF-κB binding sites of AjBcl-2 promoter. All results supported that AjCypA mediates coelomocyte apoptosis via NF-κB/AjBcl-2 signaling pathway in A. japonicus.
Collapse
Affiliation(s)
- Jiqing Liu
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, PR China
| | - Xuelin Zhao
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, PR China
| | - Zhimeng Lv
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, PR China
| | - Ming Guo
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, PR China
| | - Chenghua Li
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, PR China.
| |
Collapse
|
18
|
The Attenuation of Traumatic Brain Injury via Inhibition of Oxidative Stress and Apoptosis by Tanshinone IIA. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4170156. [PMID: 32454938 PMCID: PMC7218958 DOI: 10.1155/2020/4170156] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 10/31/2019] [Accepted: 11/28/2019] [Indexed: 12/17/2022]
Abstract
Traumatic brain injury (TBI) is a major source of mortality and long-term disability worldwide. The mechanisms associated with TBI development are poorly understood, and little progress has been made in the treatment of TBI. Tanshinone IIA is an effective agent to treat a variety of disorders; however, the mechanisms of Tanshinone IIA on TBI remain unclear. The aim of the present study was to investigate the therapeutic potential of Tanshinone IIA on TBI and its underlying molecular mechanisms. Changes in microvascular permeability were examined to determine the extent of TBI with Evans blue dye. Brain edema was assessed by measuring the wet weight to dry weight ratio. The expression levels of CD11, interleukin- (IL-) 1β, and tumor necrosis factor- (TNF-) α mRNA were determined by reverse transcription-quantitative PCR. Aquaporin-4 (AQP4), glial fibrillary acidic protein (GFAP), and p47phox protein expression levels were detected by western blotting. Superoxide dismutase (SOD), catalase and glutathione peroxidase (GSH-PX) activities, and malondialdehyde (MDA) content were determined using commercial kits. Cell apoptosis was detected by western blotting and TUNEL staining. Tanshinone IIA (10 mg/kg/day, intraperitoneal administration) significantly reduced brain water content and vascular permeability at 12, 24, 48, and 72 h after TBI. Tanshinone IIA downregulated the mRNA expression levels of various factors induced by TBI, including CD11, IL-1β, and TNF-α. Notably, CD11 mRNA downregulation suggested that Tanshinone IIA inhibited microglia activation. Further results showed that Tanshinone IIA treatment significantly downregulated AQP4 and GFAP expression. TBI-induced oxidative stress and apoptosis were markedly reversed by Tanshinone IIA, with an increase in SOD and GSH-PX activities and a decrease in the MDA content. Moreover, Tanshinone IIA decreased TBI-induced NADPH oxidase activation via the inhibition of p47phox. Tanshinone IIA attenuated TBI, and its mechanism of action may involve the inhibition of oxidative stress and apoptosis.
Collapse
|
19
|
Xu C, Diao YF, Wang J, Liang J, Xu HH, Zhao ML, Zheng B, Luan Z, Wang JJ, Yang XP, Wei MG, Duan JH, Wang KQ, Chen C, Chen F, Ming D, Zhang S, Sun HT, Li XH. Intravenously Infusing the Secretome of Adipose-Derived Mesenchymal Stem Cells Ameliorates Neuroinflammation and Neurological Functioning After Traumatic Brain Injury. Stem Cells Dev 2020; 29:222-234. [DOI: 10.1089/scd.2019.0173] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- Chao Xu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
| | - Yun-Feng Diao
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
| | - Jing Wang
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
- Department of Neurosurgery, Shanxi Dayi Hospital, Taiyuan, China
| | - Jun Liang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Hai-Huan Xu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
| | - Ming-Liang Zhao
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
| | - Bin Zheng
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Zuo Luan
- Department of Pediatrics, Pediatric Surgery of Navy General Hospital, Beijing, China
| | - Jing-Jing Wang
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
| | - Xi-Ping Yang
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
| | - Meng-Guang Wei
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
| | - Jing-Hao Duan
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Ke-Qiang Wang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Chong Chen
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
| | - Feng Chen
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
| | - Dong Ming
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Sai Zhang
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
| | - Hong-Tao Sun
- Institute of Traumatic Brain Injury and Neurology, Characteristic Medical Center of PAPF, Tianjin, China
| | - Xiao-Hong Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| |
Collapse
|
20
|
Ge Z, Wang C, Zhang J, Li X, Hu J. Tempol Protects Against Acetaminophen Induced Acute Hepatotoxicity by Inhibiting Oxidative Stress and Apoptosis. Front Physiol 2019; 10:660. [PMID: 31214044 PMCID: PMC6554449 DOI: 10.3389/fphys.2019.00660] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 05/09/2019] [Indexed: 12/12/2022] Open
Abstract
Acetaminophen (APAP)-induced acute hepatotoxicity is the leading cause of drug-induced acute liver failure. The aim of this study was to evaluate the effects of 4-hydroxy-2,2,6,6-tetramethylpiperidine-N-oxyl (tempol) on the protection of APAP-induced hepatotoxicity in mice. Mice were pretreated with a single dose of tempol (20 mg/kg per day) orally for 7 days. On the seventh day, mice were injected with a single dose of APAP (300 mg/kg) to induce acute hepatotoxicity. Our results showed that tempol treatment markedly improved liver functions with alleviations of histopathological damage induced by APAP. Tempol treatment upregulated levels of antioxidant proteins, including superoxide dismutase, catalase, and glutathione. Also, phosphorylation of phosphoinositide 3-kinase (PI3K) and protein kinase B (Akt) and protein expression of nuclear factor erythroid 2-related factor (Nrf 2) and heme oxygense-1 (HO-1) were all increased by tempol, which indicated tempol protected against APAP-induced hepatotoxicity via the PI3K/Akt/Nrf2 pathway. Moreover, tempol treatment decreased pro-apoptotic protein expressions (cleaved caspase-3 and Bax) and increased anti-apoptotic Bcl-2 in liver, as well as reducing apoptotic cells of TUNEL staining, which suggested apoptotic effects of tempol treatment. Overall, we found that tempol normalizes liver function in APAP-induced acute hepatotoxicity mice via activating PI3K/Akt/Nrf2 pathway, thus enhancing antioxidant response and inhibiting hepatic apoptosis.
Collapse
Affiliation(s)
- Zheng Ge
- Department of General Surgery, Huaihe Hospital of Henan University, Kaifeng, China
| | - Chenyu Wang
- Department of General Surgery, Huaihe Hospital of Henan University, Kaifeng, China
| | - Junjie Zhang
- Department of General Surgery, Huaihe Hospital of Henan University, Kaifeng, China
| | - Xiwang Li
- Department of General Surgery, Huaihe Hospital of Henan University, Kaifeng, China
| | - Junhong Hu
- Department of General Surgery, Huaihe Hospital of Henan University, Kaifeng, China
| |
Collapse
|
21
|
Glotfelty EJ, Delgado TE, Tovar-y-Romo LB, Luo Y, Hoffer BJ, Olson L, Karlsson TE, Mattson MP, Harvey BK, Tweedie D, Li Y, Greig NH. Incretin Mimetics as Rational Candidates for the Treatment of Traumatic Brain Injury. ACS Pharmacol Transl Sci 2019; 2:66-91. [PMID: 31396586 PMCID: PMC6687335 DOI: 10.1021/acsptsci.9b00003] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Indexed: 12/17/2022]
Abstract
Traumatic brain injury (TBI) is becoming an increasing public health issue. With an annually estimated 1.7 million TBIs in the United States (U.S) and nearly 70 million worldwide, the injury, isolated or compounded with others, is a major cause of short- and long-term disability and mortality. This, along with no specific treatment, has made exploration of TBI therapies a priority of the health system. Age and sex differences create a spectrum of vulnerability to TBI, with highest prevalence among younger and older populations. Increased public interest in the long-term effects and prevention of TBI have recently reached peaks, with media attention bringing heightened awareness to sport and war related head injuries. Along with short-term issues, TBI can increase the likelihood for development of long-term neurodegenerative disorders. A growing body of literature supports the use of glucagon-like peptide-1 (GLP-1), glucose-dependent insulinotropic peptide (GIP), and glucagon (Gcg) receptor (R) agonists, along with unimolecular combinations of these therapies, for their potent neurotrophic/neuroprotective activities across a variety of cellular and animal models of chronic neurodegenerative diseases (Alzheimer's and Parkinson's diseases) and acute cerebrovascular disorders (stroke). Mild or moderate TBI shares many of the hallmarks of these conditions; recent work provides evidence that use of these compounds is an effective strategy for its treatment. Safety and efficacy of many incretin-based therapies (GLP-1 and GIP) have been demonstrated in humans for the treatment of type 2 diabetes mellitus (T2DM), making these compounds ideal for rapid evaluation in clinical trials of mild and moderate TBI.
Collapse
Affiliation(s)
- Elliot J. Glotfelty
- Translational
Gerontology Branch, and Laboratory of Neurosciences, Intramural
Research Program, National Institute on
Aging, National Institutes of Health, Baltimore, Maryland 21224, United States
- Department
of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Thomas E. Delgado
- Translational
Gerontology Branch, and Laboratory of Neurosciences, Intramural
Research Program, National Institute on
Aging, National Institutes of Health, Baltimore, Maryland 21224, United States
| | - Luis B. Tovar-y-Romo
- Division
of Neuroscience, Institute of Cellular Physiology, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Yu Luo
- Department
of Molecular Genetics, University of Cincinnati, Cincinnati, Ohio 45221, United States
| | - Barry J. Hoffer
- Department
of Neurosurgery, Case Western Reserve University
School of Medicine, Cleveland, Ohio 44106, United States
| | - Lars Olson
- Department
of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Mark P. Mattson
- Translational
Gerontology Branch, and Laboratory of Neurosciences, Intramural
Research Program, National Institute on
Aging, National Institutes of Health, Baltimore, Maryland 21224, United States
| | - Brandon K. Harvey
- Molecular
Mechanisms of Cellular Stress and Inflammation Unit, Integrative Neuroscience
Department, National Institute on Drug Abuse,
National Institutes of Health, Baltimore, Maryland 21224, United States
| | - David Tweedie
- Translational
Gerontology Branch, and Laboratory of Neurosciences, Intramural
Research Program, National Institute on
Aging, National Institutes of Health, Baltimore, Maryland 21224, United States
| | - Yazhou Li
- Translational
Gerontology Branch, and Laboratory of Neurosciences, Intramural
Research Program, National Institute on
Aging, National Institutes of Health, Baltimore, Maryland 21224, United States
| | - Nigel H. Greig
- Translational
Gerontology Branch, and Laboratory of Neurosciences, Intramural
Research Program, National Institute on
Aging, National Institutes of Health, Baltimore, Maryland 21224, United States
| |
Collapse
|
22
|
Tian HY, Hu Y, Zhang P, Xing WX, Xu C, Yu D, Yang Y, Luo K, Li M. Spodoptera litura cyclophilin A is required for Microplitis bicoloratus bracovirus-induced apoptosis during insect cellular immune response. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2019; 100:e21534. [PMID: 30623473 DOI: 10.1002/arch.21534] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 12/17/2018] [Accepted: 12/19/2018] [Indexed: 05/27/2023]
Abstract
Microplitis bicoloratus bracovirus (MbBV) is a polydnavirus found in the parasitic wasp M. bicoloratus. Although MbBV is a known inducer of apoptosis in host hemocytes, the mechanism by which this occurs remains elusive. In this study, we found that expression of cyclophilin A (CypA) was significantly upregulated in Spodoptera litura hemocytes at 6-day post-parasitization. Similar results were reported in High Five cells (Hi5 cells) infected by MbBV, suggesting that the upregulation of CypA is linked to MbBV infection in insect cells. cDNA encoding CypA was cloned from parasitized hemocytes of S. litura, and bioinformatic analyses showed that S. litura CypA belongs to the cyclophilin family of proteins. Overexpression of S. litura CypA in Hi5 cells revealed that the protein promotes MbBV-induced apoptosis in vitro. Conversely, suppression of the expression and activity of CypA protein significantly rescued the apoptotic phenotype observed in MbBV-infected Hi5 cells, suggesting that it plays a key role in this process. MbBV infection also promoted the cytoplasmic-nuclear translocation of CypA in Hi5 cells. Taken together, these results suggest that MbBV infection upregulates the expression of CypA, which is required for MbBV-mediated apoptosis. Our findings provide insight into the role that CypA plays in insect cellular immune response.
Collapse
Affiliation(s)
- Hang-Yu Tian
- Center for Life Science, School of Life Sciences, Yunnan University, Kunming, People's Republic of China
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and Regulations, Yunnan University, Kunming, People's Republic of China
| | - Yan Hu
- Center for Life Science, School of Life Sciences, Yunnan University, Kunming, People's Republic of China
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and Regulations, Yunnan University, Kunming, People's Republic of China
| | - Pan Zhang
- Center for Life Science, School of Life Sciences, Yunnan University, Kunming, People's Republic of China
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and Regulations, Yunnan University, Kunming, People's Republic of China
| | - Wen-Xi Xing
- Center for Life Science, School of Life Sciences, Yunnan University, Kunming, People's Republic of China
| | - Cuixian Xu
- Center for Life Science, School of Life Sciences, Yunnan University, Kunming, People's Republic of China
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and Regulations, Yunnan University, Kunming, People's Republic of China
| | - Dan Yu
- Center for Life Science, School of Life Sciences, Yunnan University, Kunming, People's Republic of China
| | - Yang Yang
- Center for Life Science, School of Life Sciences, Yunnan University, Kunming, People's Republic of China
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and Regulations, Yunnan University, Kunming, People's Republic of China
| | - Kaijun Luo
- Center for Life Science, School of Life Sciences, Yunnan University, Kunming, People's Republic of China
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and Regulations, Yunnan University, Kunming, People's Republic of China
| | - Ming Li
- Center for Life Science, School of Life Sciences, Yunnan University, Kunming, People's Republic of China
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and Regulations, Yunnan University, Kunming, People's Republic of China
| |
Collapse
|
23
|
Piao CS, Holloway AL, Hong-Routson S, Wainwright MS. Depression following traumatic brain injury in mice is associated with down-regulation of hippocampal astrocyte glutamate transporters by thrombin. J Cereb Blood Flow Metab 2019; 39:58-73. [PMID: 29135354 PMCID: PMC6311670 DOI: 10.1177/0271678x17742792] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Depression after traumatic brain injury (TBI) is common but the mechanisms by which TBI causes depression are unknown. TBI decreases glutamate transporters GLT-1 and GLAST and allows extravasation of thrombin. We examined the effects of thrombin on transporter expression in primary hippocampal astrocytes. Application of a PAR-1 agonist caused down-regulation of GLT-1, which was prevented by inhibition of Rho kinase (ROCK). To confirm these mechanisms in vivo, we subjected mice to closed-skull TBI. Thrombin activity in the hippocampus increased one day following TBI. Seven days following TBI, expression of GLT-1 and GLAST was reduced in the hippocampus, and this was prevented by administration of the PAR-1 antagonist SCH79797. Inhibition of ROCK attenuated the decrease in GLT-1, but not GLAST, after TBI. We measured changes in glutamate levels in the hippocampus seven days after TBI using an implanted biosensor. Stress-induced glutamate levels were significantly increased following TBI and this was attenuated by treatment with the ROCK inhibitor fasudil. We quantified depressive behavior following TBI and found that inhibition of PAR-1 or ROCK decreased these behaviors. These results identify a novel mechanism by which TBI results in down-regulation of astrocyte glutamate transporters and implicate astrocyte and glutamate transporter dysfunction in depression following TBI.
Collapse
Affiliation(s)
- Chun-Shu Piao
- 1 Ruth D. & Ken M. Davee Pediatric Neurocritical Care Program, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,2 Division of Neurology, Ann & Robert H Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ashley L Holloway
- 1 Ruth D. & Ken M. Davee Pediatric Neurocritical Care Program, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,2 Division of Neurology, Ann & Robert H Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Sue Hong-Routson
- 1 Ruth D. & Ken M. Davee Pediatric Neurocritical Care Program, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,2 Division of Neurology, Ann & Robert H Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,3 Division of Critical Care, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Mark S Wainwright
- 1 Ruth D. & Ken M. Davee Pediatric Neurocritical Care Program, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,2 Division of Neurology, Ann & Robert H Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,3 Division of Critical Care, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
24
|
Tian H, Yu D, Hu Y, Zhang P, Yang Y, Hu Q, Li M. Angiotensin II upregulates cyclophilin A by enhancing ROS production in rat cardiomyocytes. Mol Med Rep 2018; 18:4349-4355. [PMID: 30221707 PMCID: PMC6172398 DOI: 10.3892/mmr.2018.9448] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 06/15/2018] [Indexed: 12/20/2022] Open
Abstract
Angiotensin II (Ang II) is a principal molecule of the renin-angiotensin system, which promotes hypertrophy and fibrosis. It has been demonstrated that Ang II upregulates the expression of cyclophilin A (CypA), which is a potential myocardial hypertrophy factor. However, the mechanisms by which Ang II induces the expression of CypA in cardiomyocytes remain unclear. In the present study, reactive oxygen species (ROS) were detected by fluorescence microscopy, and western blot analysis and ELISA were used to measure CypA expression. It was identified that Ang II enhanced the production of ROS in rat cardiomyocytes. ROS, in turn, promoted CypA expression and secretion. Notably, the action of Ang II was primarily dependent on the angiotensin type 2 receptor (AT2R), not the type 1 receptor. These results provided an insight into the role of the AT2R signaling pathway in Ang II-induced myocardial hypertrophy.
Collapse
Affiliation(s)
- Hangyu Tian
- Key Laboratory for Animal Genetic Diversity and Evolution of High Education in Yunnan Province, School of Life Sciences, Yunnan University, Kunming, Yunnan 650091, P.R. China
| | - Dan Yu
- Key Laboratory for Animal Genetic Diversity and Evolution of High Education in Yunnan Province, School of Life Sciences, Yunnan University, Kunming, Yunnan 650091, P.R. China
| | - Yan Hu
- Key Laboratory for Animal Genetic Diversity and Evolution of High Education in Yunnan Province, School of Life Sciences, Yunnan University, Kunming, Yunnan 650091, P.R. China
| | - Pan Zhang
- Key Laboratory for Animal Genetic Diversity and Evolution of High Education in Yunnan Province, School of Life Sciences, Yunnan University, Kunming, Yunnan 650091, P.R. China
| | - Yang Yang
- Key Laboratory for Animal Genetic Diversity and Evolution of High Education in Yunnan Province, School of Life Sciences, Yunnan University, Kunming, Yunnan 650091, P.R. China
| | - Qiang Hu
- Department of Emergency Medicine, The Second Affiliated Hospital, Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Ming Li
- Key Laboratory for Animal Genetic Diversity and Evolution of High Education in Yunnan Province, School of Life Sciences, Yunnan University, Kunming, Yunnan 650091, P.R. China
| |
Collapse
|
25
|
Binding mode of AIF(370-394) peptide to CypA: insights from NMR, label-free and molecular docking studies. Biochem J 2018; 475:2377-2393. [PMID: 29891613 DOI: 10.1042/bcj20180177] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 05/28/2018] [Accepted: 06/11/2018] [Indexed: 01/16/2023]
Abstract
The complex formation between the proteins apoptosis-inducing factor (AIF) and cyclophilin A (CypA) following oxidative stress in neuronal cells has been suggested as a main target for reverting ischemia-stroke damage. Recently, a peptide encompassing AIF residues 370-394 has been developed to target the AIF-binding site on CypA, to prevent the association between the two proteins and suppress glutamate-induced cell death in neuronal cells. Using a combined approach based on NMR spectroscopy, synthesis and in vitro testing of all Ala-scan mutants of the peptide and molecular docking/molecular dynamics, we have generated a detailed model of the AIF (370-394)/CypA complex. The model suggests us that the central region of the peptide spanning residues V374-K384 mostly interacts with the protein and that for efficient complex inhibition and preservation of CypA activity, it is bent around amino acids F46-G75 of the protein. The model is consistent with experimental data also from previous works and supports the concept that the peptide does not interfere with other CypA activities unrelated to AIF activation; therefore, it may serve as an ideal template for generating future non-peptidic antagonists.
Collapse
|
26
|
Fricker M, Tolkovsky AM, Borutaite V, Coleman M, Brown GC. Neuronal Cell Death. Physiol Rev 2018; 98:813-880. [PMID: 29488822 PMCID: PMC5966715 DOI: 10.1152/physrev.00011.2017] [Citation(s) in RCA: 692] [Impact Index Per Article: 115.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 05/23/2017] [Accepted: 07/10/2017] [Indexed: 02/07/2023] Open
Abstract
Neuronal cell death occurs extensively during development and pathology, where it is especially important because of the limited capacity of adult neurons to proliferate or be replaced. The concept of cell death used to be simple as there were just two or three types, so we just had to work out which type was involved in our particular pathology and then block it. However, we now know that there are at least a dozen ways for neurons to die, that blocking a particular mechanism of cell death may not prevent the cell from dying, and that non-neuronal cells also contribute to neuronal death. We review here the mechanisms of neuronal death by intrinsic and extrinsic apoptosis, oncosis, necroptosis, parthanatos, ferroptosis, sarmoptosis, autophagic cell death, autosis, autolysis, paraptosis, pyroptosis, phagoptosis, and mitochondrial permeability transition. We next explore the mechanisms of neuronal death during development, and those induced by axotomy, aberrant cell-cycle reentry, glutamate (excitoxicity and oxytosis), loss of connected neurons, aggregated proteins and the unfolded protein response, oxidants, inflammation, and microglia. We then reassess which forms of cell death occur in stroke and Alzheimer's disease, two of the most important pathologies involving neuronal cell death. We also discuss why it has been so difficult to pinpoint the type of neuronal death involved, if and why the mechanism of neuronal death matters, the molecular overlap and interplay between death subroutines, and the therapeutic implications of these multiple overlapping forms of neuronal death.
Collapse
Affiliation(s)
- Michael Fricker
- Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales , Australia ; Department of Clinical Neurosciences, University of Cambridge , Cambridge , United Kingdom ; Neuroscience Institute, Lithuanian University of Health Sciences , Kaunas , Lithuania ; and Department of Biochemistry, University of Cambridge , Cambridge , United Kingdom
| | - Aviva M Tolkovsky
- Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales , Australia ; Department of Clinical Neurosciences, University of Cambridge , Cambridge , United Kingdom ; Neuroscience Institute, Lithuanian University of Health Sciences , Kaunas , Lithuania ; and Department of Biochemistry, University of Cambridge , Cambridge , United Kingdom
| | - Vilmante Borutaite
- Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales , Australia ; Department of Clinical Neurosciences, University of Cambridge , Cambridge , United Kingdom ; Neuroscience Institute, Lithuanian University of Health Sciences , Kaunas , Lithuania ; and Department of Biochemistry, University of Cambridge , Cambridge , United Kingdom
| | - Michael Coleman
- Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales , Australia ; Department of Clinical Neurosciences, University of Cambridge , Cambridge , United Kingdom ; Neuroscience Institute, Lithuanian University of Health Sciences , Kaunas , Lithuania ; and Department of Biochemistry, University of Cambridge , Cambridge , United Kingdom
| | - Guy C Brown
- Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales , Australia ; Department of Clinical Neurosciences, University of Cambridge , Cambridge , United Kingdom ; Neuroscience Institute, Lithuanian University of Health Sciences , Kaunas , Lithuania ; and Department of Biochemistry, University of Cambridge , Cambridge , United Kingdom
| |
Collapse
|
27
|
6-Hydroxydopamine induces nuclear translocation of apoptosis inducing factor in nigral dopaminergic neurons in rat. Mol Cell Toxicol 2017. [DOI: 10.1007/s13273-017-0034-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
28
|
Farina B, Di Sorbo G, Chambery A, Caporale A, Leoni G, Russo R, Mascanzoni F, Raimondo D, Fattorusso R, Ruvo M, Doti N. Structural and biochemical insights of CypA and AIF interaction. Sci Rep 2017; 7:1138. [PMID: 28442737 PMCID: PMC5430804 DOI: 10.1038/s41598-017-01337-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 03/28/2017] [Indexed: 01/08/2023] Open
Abstract
The Cyclophilin A (CypA)/Apoptosis Inducing Factor (AIF) complex is implicated in the DNA degradation in response to various cellular stress conditions, such as oxidative stress, cerebral hypoxia-ischemia and traumatic brain injury. The pro-apoptotic form of AIF (AIF(Δ1-121)) mainly interacts with CypA through the amino acid region 370–394. The AIF(370-394) synthetic peptide inhibits complex formation in vitro by binding to CypA and exerts neuroprotection in a model of glutamate-mediated oxidative stress. Here, the binding site of AIF(Δ1-121) and AIF(370-394) on CypA has been mapped by NMR spectroscopy and biochemical studies, and a molecular model of the complex has been proposed. We show that AIF(370-394) interacts with CypA on the same surface recognized by AIF(Δ1-121) protein and that the region is very close to the CypA catalytic pocket. Such region partially overlaps with the binding site of cyclosporin A (CsA), the strongest catalytic inhibitor of CypA. Our data point toward distinct CypA structural determinants governing the inhibitor selectivity and the differential biological effects of AIF and CsA, and provide new structural insights for designing CypA/AIF selective inhibitors with therapeutic relevance in neurodegenerative diseases.
Collapse
Affiliation(s)
- Biancamaria Farina
- Istituto di Biostrutture e Bioimmagini, C.N.R. and CIRPEB, Via Mezzocannone 16, 80134, Napoli, Italy
| | - Gianluigi Di Sorbo
- Istituto di Biostrutture e Bioimmagini, C.N.R. and CIRPEB, Via Mezzocannone 16, 80134, Napoli, Italy.,Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università degli Studi della Campania Luigi Vanvitelli, via Vivaldi 46, 81100, Caserta, Italy
| | - Angela Chambery
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università degli Studi della Campania Luigi Vanvitelli, via Vivaldi 46, 81100, Caserta, Italy
| | - Andrea Caporale
- Istituto di Biostrutture e Bioimmagini, C.N.R. and CIRPEB, Via Mezzocannone 16, 80134, Napoli, Italy
| | - Guido Leoni
- Nouscom s.r.l. via di Castel Romano 100, 00128, Roma, Italy
| | - Rosita Russo
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università degli Studi della Campania Luigi Vanvitelli, via Vivaldi 46, 81100, Caserta, Italy
| | - Fabiola Mascanzoni
- Istituto di Biostrutture e Bioimmagini, C.N.R. and CIRPEB, Via Mezzocannone 16, 80134, Napoli, Italy
| | - Domenico Raimondo
- Sapienza, Università di Roma- Viale Regina Elena 324, 00161, Roma, Italy
| | - Roberto Fattorusso
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università degli Studi della Campania Luigi Vanvitelli, via Vivaldi 46, 81100, Caserta, Italy
| | - Menotti Ruvo
- Istituto di Biostrutture e Bioimmagini, C.N.R. and CIRPEB, Via Mezzocannone 16, 80134, Napoli, Italy
| | - Nunzianna Doti
- Istituto di Biostrutture e Bioimmagini, C.N.R. and CIRPEB, Via Mezzocannone 16, 80134, Napoli, Italy.
| |
Collapse
|
29
|
Xue Z, Song Z, Wan Y, Wang K, Mo L, Wang Y. Calcium-sensing receptor antagonist NPS2390 attenuates neuronal apoptosis though intrinsic pathway following traumatic brain injury in rats. Biochem Biophys Res Commun 2017; 486:589-594. [PMID: 28336431 DOI: 10.1016/j.bbrc.2017.03.097] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 03/19/2017] [Indexed: 10/19/2022]
Abstract
Traumatic brain injury (TBI) initiates a complex cascade of neurochemical and signaling changes that leads to neuronal apoptosis, which contributes to poor outcomes for patients with TBI. Previous study indicates that calcium-sensing receptor (CaSR) activation contributes to neuron death in focal cerebral ischemia-reperfusion mice, however, its role in neuronal apoptosis after TBI is not well-established. Using a controlled cortical impact model in rats, the present study was designed to determine the effect of CaSR inhibitor NPS2390 upon neuronal apoptosis after TBI. Rats were randomly distributed into three groups undergoing the sham surgery or TBI procedure, and NPS2390 (1.5 mg/kg) was infused subcutaneously at 30 min and 120 min after TBI. All rats were sacrificed at 24 h after TBI. Our data indicated that NPS2390 significantly reduced the brain edema and improved the neurological function after TBI. In addition, NPS2390 decreased caspase-3 levels and the number of apoptotic neurons. Furthermore, NPS2390 up-regulated anti-apoptotic protein Bcl-2 expression and down-regulated pro-apoptotic protein Bax, and reduced subsequent release of cytochrome c into the cytosol. In summary, this study indicated that inhibition of CaSR by NPS2390 attenuates neuronal apoptosis after TBI, in part, through modulating intrinsic apoptotic pathway.
Collapse
Affiliation(s)
- Zhaoliang Xue
- Department of Neurosurgery, Sir Run Run Shaw Hospital, Affiliated to Zhejiang University School of Medicine, China
| | - Zhengfei Song
- Department of Neurosurgery, Sir Run Run Shaw Hospital, Affiliated to Zhejiang University School of Medicine, China
| | - Yingfeng Wan
- Department of Neurosurgery, Sir Run Run Shaw Hospital, Affiliated to Zhejiang University School of Medicine, China
| | - Kun Wang
- Department of Neurosurgery, Sir Run Run Shaw Hospital, Affiliated to Zhejiang University School of Medicine, China
| | - Lianjie Mo
- Department of Neurosurgery, Sir Run Run Shaw Hospital, Affiliated to Zhejiang University School of Medicine, China
| | - Yirong Wang
- Department of Neurosurgery, Sir Run Run Shaw Hospital, Affiliated to Zhejiang University School of Medicine, China.
| |
Collapse
|
30
|
Doti N, Ruvo M. Relevance and therapeutic potential of CypA targeting to block apoptosis inducing factor-mediated neuronal cell death. Neural Regen Res 2017; 12:1428-1429. [PMID: 29089982 PMCID: PMC5649457 DOI: 10.4103/1673-5374.215248] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Affiliation(s)
- Nunzianna Doti
- Institute of Biostructures and Bioimaging (IBB)-CNR, Via Mezzocannone, 16, 80134 Napoli, Italy
| | - Menotti Ruvo
- Institute of Biostructures and Bioimaging (IBB)-CNR, Via Mezzocannone, 16, 80134 Napoli, Italy
| |
Collapse
|
31
|
Kumar A, Barrett JP, Alvarez-Croda DM, Stoica BA, Faden AI, Loane DJ. NOX2 drives M1-like microglial/macrophage activation and neurodegeneration following experimental traumatic brain injury. Brain Behav Immun 2016; 58:291-309. [PMID: 27477920 PMCID: PMC5067217 DOI: 10.1016/j.bbi.2016.07.158] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 07/20/2016] [Accepted: 07/27/2016] [Indexed: 12/16/2022] Open
Abstract
Following traumatic brain injury (TBI), activation of microglia and peripherally derived inflammatory macrophages occurs in association with tissue damage. This neuroinflammatory response may have beneficial or detrimental effects on neuronal survival, depending on the functional polarization of these cells along a continuum from M1-like to M2-like activation states. The mechanisms that regulate M1-like and M2-like activation after TBI are not well understood, but appear in part to reflect the redox state of the lesion microenvironment. NADPH oxidase (NOX2) is a critical enzyme system that generates reactive oxygen species in microglia/macrophages. After TBI, NOX2 is strongly up-regulated in M1-like, but not in M2-like polarized cells. Therefore, we hypothesized that NOX2 drives M1-like neuroinflammation and contributes to neurodegeneration and loss of neurological function after TBI. In the present studies we inhibited NOX2 activity using NOX2-knockout mice or the selective peptide inhibitor gp91ds-tat. We show that NOX2 is highly up-regulated in infiltrating macrophages after injury, and that NOX2 deficiency reduces markers of M1-like activation, limits tissue loss and neurodegeneration, and improves motor recovery after moderate-level control cortical injury (CCI). NOX2 deficiency also promotes M2-like activation after CCI, through increased IL-4Rα signaling in infiltrating macrophages, suggesting that NOX2 acts as a critical switch between M1- and M2-like activation states after TBI. Administration of gp91ds-tat to wild-type CCI mice starting at 24h post-injury reduces deficits in cognitive function and increased M2-like activation in the hippocampus. Collectively, our data indicate that increased NOX2 activity after TBI drives M1-like activation that contributes to inflammatory-mediated neurodegeneration, and that inhibiting this pathway provides neuroprotection, in part by altering M1-/M2-like balance towards the M2-like neuroinflammatory response.
Collapse
Affiliation(s)
- Alok Kumar
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - James P. Barrett
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dulce-Mariely Alvarez-Croda
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA,Posgrado en Neuroetologia, Universidad Veracruzana, Xalapa, Mexico,Centro de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa, Mexico
| | - Bogdan A. Stoica
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alan I. Faden
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - David J. Loane
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA,Correspondence: David J. Loane PhD, Department of Anesthesiology, University of Maryland School of Medicine, 655 West Baltimore Street, #6-011, Baltimore, MD 21201. Tel: 410-706-5188 Fax: 410-706-1639,
| |
Collapse
|
32
|
CHU HUI, YU HANG, REN DING, ZHU KEJUN, HUANG HONG. Plumbagin exerts protective effects in nucleus pulposus cells by attenuating hydrogen peroxide-induced oxidative stress, inflammation and apoptosis through NF-κB and Nrf-2. Int J Mol Med 2016; 37:1669-76. [DOI: 10.3892/ijmm.2016.2564] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 04/01/2016] [Indexed: 11/06/2022] Open
|
33
|
Thrombin decreases expression of the glutamate transporter GLAST and inhibits glutamate uptake in primary cortical astrocytes via the Rho kinase pathway. Exp Neurol 2015; 273:288-300. [PMID: 26391563 DOI: 10.1016/j.expneurol.2015.09.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 09/17/2015] [Indexed: 01/08/2023]
Abstract
Astrocyte glutamate transporters GLAST and GLT1 play a key role in regulating neuronal excitation and their levels are altered in patients with epilepsy, and after traumatic brain injury. The mechanisms which regulate their expression are not well understood. We tested the hypothesis that exposure of astrocytes to high levels of thrombin, as may occur after a compromise of the blood-brain barrier, would reduce astrocyte glutamate transporter levels. In isolated rat cortical astrocytes we examined the effects of thrombin on the expression and function of glutamate transporters, and the signaling pathways involved in these responses by using Western blotting and selective inhibitors. Thrombin induced a selective decrease in the expression of GLAST but not GLT1, with a corresponding decrease in the capacity of astrocytes to take up glutamate. Activation of the thrombin receptor PAR-1 with an activating peptide induced a similar decrease in the expression of GLAST and compromise of glutamate uptake. The downregulation of GLAST induced by thrombin was mediated by the mitogen activated protein kinases p38 MAPK, ERK and JNK, but inhibition of these kinases did not prevent the decrease in glutamate uptake induced by thrombin. In contrast, inhibition of the Rho kinase pathway using the specific inhibitor, Y27632, suppressed both the decrease in the expression of GLAST and the decrease in glutamate uptake induced by thrombin. In hippocampal astrocyte cultures, thrombin caused a decrease in both GLAST and GLT1. In tissue resected from brains of children with intractable epilepsy, we found a decrease in the integrity of the blood-brain barrier along with a reduction in immunoreactivity for both transporters which was associated with an increase in cleaved thrombin and reactive astrogliosis. The in vitro results suggest a specific mechanism by which thrombin may lead to a compromise of astrocyte function and enhanced synaptic excitability after the blood-brain barrier is compromised. The human in vivo results provide indirect support evidence linking the compromise of the blood-brain barrier to thrombin-induced reduction in glutamate transporter expression and an increase in neuronal excitation.
Collapse
|
34
|
Temporal pattern of neurodegeneration, programmed cell death, and neuroplastic responses in the thalamus after lateral fluid percussion brain injury in the rat. J Neuropathol Exp Neurol 2015; 74:512-26. [PMID: 25933386 DOI: 10.1097/nen.0000000000000194] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The effects of traumatic brain injury (TBI) on the thalamus are not well characterized. We analyzed neuronal degeneration and loss, apoptosis, programmed cell death-executing pathways, and neuroplastic responses in the rat thalamus during the first week after lateral fluid percussion injury (LFPI). The most prominent neurodegenerative and neuroplastic changes were observed in the region containing the posterior thalamic nuclear group and ventral posteromedial and posterolateral thalamic nuclei ipsilateral to the LFPI. There was progressive neurodegeneration in these regions, with maximal neuronal loss on Day 7. Increases in numbers of apoptotic cells were detected on Day 1 and were enhanced on Days 3 and 7 after TBI. There was unchanged expression of active caspase-3 at all postinjury time points, but there was increased expression of apoptosis-inducing factor (AIF) on Day 7. The AIF nuclear translocation was detected on Day 1 and was maximal on Day 7. Total thalamic synaptophysin expression was unchanged, but immunostaining intensities were increased at all time points after TBI. Decreased growth-associated protein-43 expression and signal intensity were observed on Day 1. Our results suggest that progressive neuronal damage and loss, AIF signaling pathway-dependent programmed cell death, and limited neuroplastic changes occur in the rat thalamus during the first week after LFPI induction.
Collapse
|
35
|
Skendelas JP, Muccigrosso M, Eiferman DS, Godbout JP. Chronic Inflammation After TBI and Associated Behavioral Sequelae. CURRENT PHYSICAL MEDICINE AND REHABILITATION REPORTS 2015. [DOI: 10.1007/s40141-015-0091-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
36
|
Abstract
Traumatic brain injury (TBI) is a major cause of mortality and morbidity worldwide. Despite extensive preclinical research supporting the effectiveness of neuroprotective therapies for brain trauma, there have been no successful randomized controlled clinical trials to date. TBI results in delayed secondary tissue injury due to neurochemical, metabolic and cellular changes; modulating such effects has provided the basis for neuroprotective interventions. To establish more effective neuroprotective treatments for TBI it is essential to better understand the complex cellular and molecular events that contribute to secondary injury. Here we critically review relevant research related to causes and modulation of delayed tissue damage, with particular emphasis on cell death mechanisms and post-traumatic neuroinflammation. We discuss the concept of utilizing multipotential drugs that target multiple secondary injury pathways, rather than more specific "laser"-targeted strategies that have uniformly failed in clinical trials. Moreover, we assess data supporting use of neuroprotective drugs that are currently being evaluated in human clinical trials for TBI, as well as promising emerging experimental multipotential drug treatment strategies. Finally, we describe key challenges and provide suggestions to improve the likelihood of successful clinical translation.
Collapse
Affiliation(s)
- David J Loane
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), National Study Center for Trauma and EMS, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bogdan A Stoica
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), National Study Center for Trauma and EMS, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alan I Faden
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), National Study Center for Trauma and EMS, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
37
|
Lazarus RC, Buonora JE, Jacobowitz DM, Mueller GP. Protein carbonylation after traumatic brain injury: cell specificity, regional susceptibility, and gender differences. Free Radic Biol Med 2015; 78:89-100. [PMID: 25462645 DOI: 10.1016/j.freeradbiomed.2014.10.507] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 10/07/2014] [Accepted: 10/10/2014] [Indexed: 12/15/2022]
Abstract
Protein carbonylation is a well-documented and quantifiable consequence of oxidative stress in several neuropathologies, including multiple sclerosis, Alzheimer׳s disease, and Parkinson׳s disease. Although oxidative stress is a hallmark of traumatic brain injury (TBI), little work has explored the specific neural regions and cell types in which protein carbonylation occurs. Furthermore, the effect of gender on protein carbonylation after TBI has not been studied. The present investigation was designed to determine the regional and cell specificity of TBI-induced protein carbonylation and how this response to injury is affected by gender. Immunohistochemistry was used to visualize protein carbonylation in the brains of adult male and female Sprague-Dawley rats subjected to controlled cortical impact (CCI) as an injury model of TBI. Cell-specific markers were used to colocalize the presence of carbonylated proteins in specific cell types, including astrocytes, neurons, microglia, and oligodendrocytes. Results also indicated that the injury lesion site, ventral portion of the dorsal third ventricle, and ventricular lining above the median eminence showed dramatic increases in protein carbonylation after injury. Specifically, astrocytes and limited regions of ependymal cells adjacent to the dorsal third ventricle and the median eminence were most susceptible to postinjury protein carbonylation. However, these patterns of differential susceptibility to protein carbonylation were gender dependent, with males showing significantly greater protein carbonylation at sites distant from the lesion. Proteomic analyses were also conducted and determined that the proteins most affected by carbonylation in response to TBI include glial fibrillary acidic protein, dihydropyrimidase-related protein 2, fructose-bisphosphate aldolase C, and fructose-bisphosphate aldolase A. Many other proteins, however, were not carbonylated by CCI. These findings indicate that there is both regional and protein specificity in protein carbonylation after TBI. The marked increase in carbonylation seen in ependymal layers distant from the lesion suggests a mechanism involving the transmission of a cerebral spinal fluid-borne factor to these sites. Furthermore, this process is affected by gender, suggesting that hormonal mechanisms may serve a protective role against oxidative stress.
Collapse
Affiliation(s)
- Rachel C Lazarus
- Program in Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - John E Buonora
- Program in Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - David M Jacobowitz
- Program in Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Gregory P Mueller
- Program in Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA.
| |
Collapse
|
38
|
Mo J, Marshall B, Covar J, Zhang NY, Smith SB, Atherton SS, Zhang M. Role of Bax in death of uninfected retinal cells during murine cytomegalovirus retinitis. Invest Ophthalmol Vis Sci 2014; 55:7137-46. [PMID: 25298417 DOI: 10.1167/iovs.14-15404] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Extensive death of uninfected bystander neuronal cells is an important component of the pathogenesis of cytomegalovirus retinitis. Our previous results have shown that caspase 3-dependent and -independent pathways are involved in death of uninfected bystander cells during murine cytomegalovirus (MCMV) retinitis and also that Bcl-2, an important inhibitor of apoptosis via the Bax-mediated mitochondrial pathway, is downregulated during this process. The purpose of this study was to determine whether Bax-mediated mitochondrial damage has a significant role in the death of uninfected retinal cells. METHODS BALB/c mice, Bax(-/-) mice, or Bax(+/+) mice were immunosuppressed with methylprednisolone and infected with 5 × 10(3) plaque-forming units (PFU) of the K181 strain of MCMV via the supraciliary route. Injected eyes were analyzed by plaque assay, electron microscopy, hematoxylin and eosin (H&E) staining, TUNEL assay, Western blot (for caspase 3, caspase 12, Bax, receptor interacting protein-1 [RIP1] and receptor interacting protein-3 [RIP3]), as well as immunohistochemical staining for MCMV early antigen and cleaved caspase 3. RESULTS Significantly more Bax was detected in mitochondrial fractions of MCMV-infected eyes than in mitochondrial fractions of mock-infected control eyes. Furthermore, the level of cleaved caspase 3 was significantly lower in MCMV-infected Bax(-/-) eyes than in MCMV-infected Bax(+/+) eyes. However, more caspase 3-independent cell death of uninfected bystander retinal cells and more cleaved RIP1 were observed in Bax(-/-) than in Bax(+/+) eyes. CONCLUSIONS During MCMV retinitis, Bax is activated and has an important role in death of uninfected bystander retinal cells by caspase 3-dependent apoptosis. Although the exact mechanism remains to be deciphered, active Bax might also prevent death of some types of uninfected retinal cells by a caspase 3-independent pathway.
Collapse
Affiliation(s)
- Juan Mo
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, United States
| | - Brendan Marshall
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, United States
| | - Jason Covar
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, United States
| | - Nancy Y Zhang
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, United States
| | - Sylvia B Smith
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, United States
| | - Sally S Atherton
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, United States The James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, United States
| | - Ming Zhang
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, United States The James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, United States
| |
Collapse
|
39
|
Zhang X, Yu Q, Jiang W, Bi Y, Zhang Y, Gong M, Wei X, Li T, Chen J. All-trans retinoic acid suppresses apoptosis in PC12 cells injured by oxygen and glucose deprivation via the retinoic acid receptor α signaling pathway. Mol Med Rep 2014; 10:2549-55. [PMID: 25231683 DOI: 10.3892/mmr.2014.2568] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Accepted: 05/29/2014] [Indexed: 11/06/2022] Open
Abstract
Vitamin A (VA) has a number of important biological functions in human growth and development. Previous studies by our group demonstrated that the normal VA levels improved recovery of learning and memory function and decreased apoptosis in rats with hypoxic‑ischemic brain damage (HIBD). However, it has not been fully elucidated how VA regulates the apoptosis of neuronal cells. To investigate the anti‑apoptotic effect of VA, an in vitro oxygen glucose deprivation (OGD) model in PC12 cells was treated with four concentrations of all‑trans‑retinoic acid (ATRA), an active in vivo product of VA. Following in vitro OGD injury in PC12 cells, the percentage of apoptosis and the fluorescence intensity of the mitochondrial membrane potential (MMP) were increased in the cells, and the expression levels of B-cell lymphoma-associated X (Bax) were enhanced. ATRA treatment at 2‑4 µmol/l for 24 h decreased the percentage of apoptosis and the MMP of the PC12 cells injured by OGD. ATRA at 4 µmol/l also reduced the expression levels of Bax and enhanced the expression of B-cell lymphoma 2. Furthermore, RNA interference with retinoic acid receptor α (RARα) reversed the observed effect in PC12 cells following ATRA treatment at 4 µmol/l alone. In conclusion, the present study suggested that treatment with ATRA at 4 µmol/l suppressed apoptosis of PC12 cells following OGD injury, potentially through regulation of the RARα signaling pathway.
Collapse
Affiliation(s)
- Xiaojian Zhang
- Children's Nutritional Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
| | - Qin Yu
- Children's Nutritional Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
| | - Wei Jiang
- Children's Nutritional Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
| | - Yang Bi
- Children's Nutritional Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
| | - Yun Zhang
- Children's Nutritional Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
| | - Min Gong
- Children's Nutritional Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
| | - Xiaoping Wei
- Children's Nutritional Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
| | - Tingyu Li
- Children's Nutritional Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
| | - Jie Chen
- Children's Nutritional Research Center, Children's Hospital of Chongqing Medical University, Chongqing 400014, P.R. China
| |
Collapse
|
40
|
Nazıroğlu M, Senol N, Ghazizadeh V, Yürüker V. Neuroprotection induced by N-acetylcysteine and selenium against traumatic brain injury-induced apoptosis and calcium entry in hippocampus of rat. Cell Mol Neurobiol 2014; 34:895-903. [PMID: 24842665 DOI: 10.1007/s10571-014-0069-2] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 04/21/2014] [Indexed: 12/17/2022]
Abstract
Neurodegeneration associated with acute central nervous system injuries and diseases such as spinal cord injury and traumatic brain injury (TBI) are reported to be mediated by the regulation of apoptosis and oxidative stress through Ca(2+) influx. The thiol redox system antioxidants, such as N-acetylcysteine (NAC) and selenium (Se), display neuroprotective activities mediated at least in part by their antioxidant and anti-inflammatory properties. However, there are no reports on hippocampal apoptosis, cytosolic reactive oxygen species (ROS), or Ca(2+) values in rats with an induced TBI. Therefore, we tested the effects of Se and NAC administration on apoptosis, oxidative stress, and Ca(2+) influx through TRPV1 channel activations in the hippocampus of TBI-induced rats. The 32 rats were divided into four groups: control, TBI, TBI + NAC, and TBI + Se groups. Intraperitoneal administrations of NAC and Se were performed at 1, 24, 48, and 72 h after TBI induction. After 3 days, the hippocampal neurons were freshly isolated from the rats. In cytosolic-free Ca(2+) analyses, the neurons were stimulated with the TRPV1 channel agonist capsaicin, a pungent compound found in hot chili peppers. Cytosolic-free Ca(2+), apoptosis, cytosolic ROS levels, and caspase-3 and -9 activities were higher in the TBI group than control. The values in the hippocampus were decreased by Se and NAC administrations. In conclusion, we observed that NAC and Se have protective effects on oxidative stress, apoptosis, and Ca(2+) entry via TRPV1 channel activation in the hippocampus of this TBI model, but the effect of NAC appears to be much greater than that of Se. They are both interesting candidates for studying the amelioration of TBIs.
Collapse
Affiliation(s)
- Mustafa Nazıroğlu
- Neuroscience Research Center, University of SuleymanDemirel, TR-32260, Isparta, Turkey,
| | | | | | | |
Collapse
|
41
|
Galluzzi L, Kepp O, Krautwald S, Kroemer G, Linkermann A. Molecular mechanisms of regulated necrosis. Semin Cell Dev Biol 2014; 35:24-32. [PMID: 24582829 DOI: 10.1016/j.semcdb.2014.02.006] [Citation(s) in RCA: 181] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Accepted: 02/12/2014] [Indexed: 01/01/2023]
Abstract
It is now clear that apoptosis does not constitute the sole genetically encoded form of cell death. Rather, cells can spontaneously undertake or exogenously be driven into a cell death subroutine that manifests with necrotic features, yet can be inhibited by pharmacological and genetic interventions. As regulated necrosis (RN) plays a major role in both physiological scenarios (e.g., embryonic development) and pathological settings (e.g., ischemic disorders), consistent efforts have been made throughout the last decade toward the characterization of the molecular mechanisms that underlie this cell death modality. Contrarily to initial beliefs, RN does not invariably result from the activation of a receptor interacting protein kinase 3 (RIPK3)-dependent signaling pathway, but may be ignited by distinct molecular networks. Nowadays, various types of RN have been characterized, including (but not limited to) necroptosis, mitochondrial permeability transition (MPT)-dependent RN and parthanatos. Of note, the inhibition of only one of these modules generally exerts limited cytoprotective effects in vivo, underscoring the degree of interconnectivity that characterizes RN. Here, we review the signaling pathways, pathophysiological relevance and therapeutic implications of the major molecular cascades that underlie RN.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Gustave Roussy, F-94805 Villejuif, France; Université Paris Descartes/Paris V, Sorbonne Paris Cité, F-75005 Paris, France; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, F-75005 Paris, France
| | - Oliver Kepp
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, F-75005 Paris, France; INSERM, U848, F-94805 Villejuif, France; Metabolomics and Cell Biology Platforms, Gustave Roussy, F-94805 Villejuif, France
| | - Stefan Krautwald
- Division for Nephrology and Hypertension, Christian-Albrechts-University, D-24118 Kiel, Germany
| | - Guido Kroemer
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, F-75005 Paris, France; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, F-75005 Paris, France; INSERM, U848, F-94805 Villejuif, France; Metabolomics and Cell Biology Platforms, Gustave Roussy, F-94805 Villejuif, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, F-75015 Paris, France.
| | - Andreas Linkermann
- Division for Nephrology and Hypertension, Christian-Albrechts-University, D-24118 Kiel, Germany
| |
Collapse
|
42
|
Stoica BA, Loane DJ, Zhao Z, Kabadi SV, Hanscom M, Byrnes KR, Faden AI. PARP-1 inhibition attenuates neuronal loss, microglia activation and neurological deficits after traumatic brain injury. J Neurotrauma 2014; 31:758-72. [PMID: 24476502 DOI: 10.1089/neu.2013.3194] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Traumatic brain injury (TBI) causes neuronal cell death as well as microglial activation and related neurotoxicity that contribute to subsequent neurological dysfunction. Poly (ADP-ribose) polymerase (PARP-1) induces neuronal cell death through activation of caspase-independent mechanisms, including release of apoptosis inducing factor (AIF), and microglial activation. Administration of PJ34, a selective PARP-1 inhibitor, reduced cell death of primary cortical neurons exposed to N-Methyl-N'-Nitro-N-Nitrosoguanidine (MNNG), a potent inducer of AIF-dependent cell death. PJ34 also attenuated lipopolysaccharide and interferon-γ-induced activation of BV2 or primary microglia, limiting NF-κB activity and iNOS expression as well as decreasing generation of reactive oxygen species and TNFα. Systemic administration of PJ34 starting as late as 24 h after controlled cortical impact resulted in improved motor function recovery in mice with TBI. Stereological analysis demonstrated that PJ34 treatment reduced the lesion volume, attenuated neuronal cell loss in the cortex and thalamus, and reduced microglial activation in the TBI cortex. PJ34 treatment did not improve cognitive performance in a Morris water maze test or reduce neuronal cell loss in the hippocampus. Overall, our data indicate that PJ34 has a significant, albeit selective, neuroprotective effect after experimental TBI, and its therapeutic effect may be from multipotential actions on neuronal cell death and neuroinflammatory pathways.
Collapse
Affiliation(s)
- Bogdan A Stoica
- 1 Department of Anesthesiology, Center for Shock, Trauma and Anesthesiology Research (STAR), National Study Center for Trauma and EMS, University of Maryland , School of Medicine, Baltimore, Maryland
| | | | | | | | | | | | | |
Collapse
|
43
|
Kabadi SV, Faden AI. Neuroprotective strategies for traumatic brain injury: improving clinical translation. Int J Mol Sci 2014; 15:1216-36. [PMID: 24445258 PMCID: PMC3907865 DOI: 10.3390/ijms15011216] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 01/07/2014] [Accepted: 01/13/2014] [Indexed: 01/15/2023] Open
Abstract
Traumatic brain injury (TBI) induces secondary biochemical changes that contribute to delayed neuroinflammation, neuronal cell death, and neurological dysfunction. Attenuating such secondary injury has provided the conceptual basis for neuroprotective treatments. Despite strong experimental data, more than 30 clinical trials of neuroprotection in TBI patients have failed. In part, these failures likely reflect methodological differences between the clinical and animal studies, as well as inadequate pre-clinical evaluation and/or trial design problems. However, recent changes in experimental approach and advances in clinical trial methodology have raised the potential for successful clinical translation. Here we critically analyze the current limitations and translational opportunities for developing successful neuroprotective therapies for TBI.
Collapse
Affiliation(s)
- Shruti V Kabadi
- Department of Anesthesiology, Center for Shock, Trauma and Anesthesiology Research (STAR), National Study Center for Trauma and EMS, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Alan I Faden
- Department of Anesthesiology, Center for Shock, Trauma and Anesthesiology Research (STAR), National Study Center for Trauma and EMS, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
44
|
Inhibition of the AIF/CypA complex protects against intrinsic death pathways induced by oxidative stress. Cell Death Dis 2014; 5:e993. [PMID: 24434516 PMCID: PMC4040673 DOI: 10.1038/cddis.2013.518] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 10/28/2013] [Accepted: 11/15/2013] [Indexed: 01/12/2023]
Abstract
Delayed neuronal cell death largely contributes to the progressive infarct development and associated functional impairments after cerebral ischemia or brain trauma. Previous studies exposed a key role for the interaction of the mitochondrial protein apoptosis-inducing factor (AIF) and cytosolic cyclophilin A (CypA) in pathways of programmed cell death in neurons in vitro and in vivo. These studies suggested that pro-apoptotic activities of AIF, such as its translocation to the nucleus and subsequent DNA degradation, depend on the physical interaction of AIF with CypA. Hence, this protein complex may represent a new pharmacological target for inhibiting the lethal action of AIF on the brain tissue. In this study, we show that the AIF amino-acid residues 370–394 mediate the protein complex formation of AIF with CypA. The synthetic AIF(370–394) peptide inhibited AIF/CypA complex formation in vitro by binding CypA with a KD of 12 μM. Further, the peptide exerted pronounced neuroprotective effects in a model of glutamate-induced oxidative stress in cultured HT-22 cells. In this model system of AIF-dependent cell death, the AIF(370–394) peptide preserved mitochondrial integrity, as detected by measurements of the mitochondrial membrane potential and quantification of mitochondrial fragmentation. Further, the AIF(370–394) peptide inhibited perinuclear accumulation of fragmented mitochondria, mitochondrial release of AIF to the nucleus and glutamate-induced cell death to a similar extent as CypA-siRNA. These data indicate that the targeting of the AIF-CypA axis is an effective strategy of neuroprotection.
Collapse
|
45
|
Neuroprotective effects of geranylgeranylacetone in experimental traumatic brain injury. J Cereb Blood Flow Metab 2013; 33:1897-908. [PMID: 23942364 PMCID: PMC3851897 DOI: 10.1038/jcbfm.2013.144] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 07/19/2013] [Accepted: 07/22/2013] [Indexed: 12/11/2022]
Abstract
Geranylgeranylacetone (GGA) is an inducer of heat-shock protein 70 (HSP70) that has been used clinically for many years as an antiulcer treatment. It is centrally active after oral administration and is neuroprotective in experimental brain ischemia/stroke models. We examined the effects of single oral GGA before treatment (800 mg/kg, 48 hours before trauma) or after treatment (800 mg/kg, 3 hours after trauma) on long-term functional recovery and histologic outcomes after moderate-level controlled cortical impact, an experimental traumatic brain injury (TBI) model in mice. The GGA pretreatment increased the number of HSP70(+) cells and attenuated posttraumatic α-fodrin cleavage, a marker of apoptotic cell death. It also improved sensorimotor performance on a beam walk task; enhanced recovery of cognitive/affective function in the Morris water maze, novel object recognition, and tail-suspension tests; and improved outcomes using a composite neuroscore. Furthermore, GGA pretreatment reduced the lesion size and neuronal loss in the hippocampus, cortex, and thalamus, and decreased microglial activation in the cortex when compared with vehicle-treated TBI controls. Notably, GGA was also effective in a posttreatment paradigm, showing significant improvements in sensorimotor function, and reducing cortical neuronal loss. Given these neuroprotective actions and considering its longstanding clinical use, GGA should be considered for the clinical treatment of TBI.
Collapse
|
46
|
Walker KR, Tesco G. Molecular mechanisms of cognitive dysfunction following traumatic brain injury. Front Aging Neurosci 2013; 5:29. [PMID: 23847533 PMCID: PMC3705200 DOI: 10.3389/fnagi.2013.00029] [Citation(s) in RCA: 179] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 06/18/2013] [Indexed: 12/12/2022] Open
Abstract
Traumatic brain injury (TBI) results in significant disability due to cognitive deficits particularly in attention, learning and memory, and higher-order executive functions. The role of TBI in chronic neurodegeneration and the development of neurodegenerative diseases including Alzheimer's disease (AD), Parkinson's disease (PD), Amyotrophic Lateral Sclerosis (ALS) and most recently chronic traumatic encephalopathy (CTE) is of particular importance. However, despite significant effort very few therapeutic options exist to prevent or reverse cognitive impairment following TBI. In this review, we present experimental evidence of the known secondary injury mechanisms which contribute to neuronal cell loss, axonal injury, and synaptic dysfunction and hence cognitive impairment both acutely and chronically following TBI. In particular we focus on the mechanisms linking TBI to the development of two forms of dementia: AD and CTE. We provide evidence of potential molecular mechanisms involved in modulating Aβ and Tau following TBI and provide evidence of the role of these mechanisms in AD pathology. Additionally we propose a mechanism by which Aβ generated as a direct result of TBI is capable of exacerbating secondary injury mechanisms thereby establishing a neurotoxic cascade that leads to chronic neurodegeneration.
Collapse
Affiliation(s)
- Kendall R Walker
- Alzheimer's Disease Research Laboratory, Department of Neuroscience, Tufts University School of Medicine Boston, MA, USA
| | | |
Collapse
|
47
|
Moretti L, Cristofori I, Weaver SM, Chau A, Portelli JN, Grafman J. Cognitive decline in older adults with a history of traumatic brain injury. Lancet Neurol 2013; 11:1103-12. [PMID: 23153408 DOI: 10.1016/s1474-4422(12)70226-0] [Citation(s) in RCA: 154] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Traumatic brain injury (TBI) is an important public health problem with potentially serious long-term neurobehavioural sequelae. There is evidence to suggest that a history of TBI can increase a person's risk of developing Alzheimer's disease. However, individuals with dementia do not usually have a history of TBI, and survivors of TBI do not invariably acquire dementia later in life. Instead, a history of traumatic brain injury, combined with brain changes associated with normal ageing, might lead to exacerbated cognitive decline in older adults. Strategies to increase or maintain cognitive reserve might help to prevent exacerbated decline after TBI. Systematic clinical assessment could help to differentiate between exacerbated cognitive decline and mild cognitive impairment, a precursor of Alzheimer's disease, with important implications for patients and their families.
Collapse
Affiliation(s)
- Laura Moretti
- Traumatic Brain Injury Research Laboratory, Kessler Foundation, West Orange, NJ, USA
| | | | | | | | | | | |
Collapse
|
48
|
Piao CS, Stoica BA, Wu J, Sabirzhanov B, Zhao Z, Cabatbat R, Loane DJ, Faden AI. Late exercise reduces neuroinflammation and cognitive dysfunction after traumatic brain injury. Neurobiol Dis 2013; 54:252-63. [PMID: 23313314 DOI: 10.1016/j.nbd.2012.12.017] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 12/21/2012] [Accepted: 12/28/2012] [Indexed: 11/29/2022] Open
Abstract
Delayed secondary biochemical and cellular changes after traumatic brain injury continue for months to years, and are associated with chronic neuroinflammation and progressive neurodegeneration. Physical activity can reduce inflammation and facilitate recovery after brain injury. Here, we investigated the time-dependent effects, and underlying mechanisms of post-traumatic exercise initiation on outcome after moderate traumatic brain injury using a well-characterized mouse controlled cortical impact model. Late exercise initiation beginning at 5weeks after trauma, but not early initiation of exercise at 1week, significantly reduced working and retention memory impairment at 3months, and decreased lesion volume compared to non-exercise injury controls. Cognitive recovery was associated with attenuation of classical inflammatory pathways, activation of alternative inflammatory responses and enhancement of neurogenesis. In contrast, early initiation of exercise failed to alter behavioral recovery or lesion size, while increasing the neurotoxic pro-inflammatory responses. These data underscore the critical importance of timing of exercise initiation after trauma and its relation to neuroinflammation, and challenge the widely held view that effective neuroprotection requires early intervention.
Collapse
Affiliation(s)
- Chun-Shu Piao
- Center for Shock, Trauma and Anesthesiology Research (STAR) and Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Sun Y, Zhang Y, Wang X, Blomgren K, Zhu C. Apoptosis-inducing factor downregulation increased neuronal progenitor, but not stem cell, survival in the neonatal hippocampus after cerebral hypoxia-ischemia. Mol Neurodegener 2012; 7:17. [PMID: 22534064 PMCID: PMC3464153 DOI: 10.1186/1750-1326-7-17] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 04/25/2012] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND A considerable proportion of all newly generated cells in the hippocampus will die before becoming fully differentiated, both under normal and pathological circumstances. The caspase-independent apoptosis-inducing factor (AIF) has not been investigated previously in this context. RESULTS Postnatal day 8 (P8) harlequin (Hq) mutant mice, expressing lower levels of AIF, and wild type littermates were injected with BrdU once daily for two days to label newborn cells. On P10 mice were subjected to hypoxia-ischemia (HI) and their brains were analyzed 4 h, 24 h or 4 weeks later. Overall tissue loss was 63.5% lower in Hq mice 4 weeks after HI. Short-term survival (4 h and 24 h) of labeled cells in the subgranular zone was neither affected by AIF downregulation, nor by HI. Long-term (4 weeks) survival of undifferentiated, BLBP-positive stem cells was reduced by half after HI, but this was not changed by AIF downregulation. Neurogenesis, however, as judged by BrdU/NeuN double labeling, was reduced by half after HI in wild type mice but preserved in Hq mice, indicating that primarily neural progenitors and neurons were protected. A wave of cell death started early after HI in the innermost layers of the granule cell layer (GCL) and moved outward, such that 24 h after HI dying cells could be detected in the entire GCL. CONCLUSIONS These findings demonstrate that AIF downregulation provides not only long-term overall neuroprotection after HI, but also protects neural progenitor cells, thereby rescuing hippocampal neurogenesis.
Collapse
Affiliation(s)
- Yanyan Sun
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Pediatrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Pediatrics, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Yu Zhang
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Pediatrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoyang Wang
- Department of Pediatrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Perinatal Center, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Klas Blomgren
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Pediatrics, The Queen Silvia Children’s Hospital, University of Gothenburg, Gothenburg, Sweden
- Department of Women’s and Children’s Health, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Changlian Zhu
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Pediatrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|