1
|
Intracellular DAMPs in Neurodegeneration and Their Role in Clinical Therapeutics. Mol Neurobiol 2023; 60:3600-3616. [PMID: 36859688 DOI: 10.1007/s12035-023-03289-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 02/21/2023] [Indexed: 03/03/2023]
Abstract
Neuroinflammation is the major implication of neurodegeneration. This is a complex process which initiates from the cellular injury triggering the innate immune system which gives rise to damage-associated molecular patterns (DAMPs) which are also recognized as endogenous danger indicators. These originate from various compartments of the cell under pathological stimulus. These are very popular candidates having their origin in the intracellular compartments and organelles of the cell and may have their site of action itself in the intracellular or at the extracellular spaces. Under the influence of the pathological stimuli, they interact with the pattern-recognition receptor to initiate their pro-inflammatory cascade followed by the cytokine release. This provides a good opportunity for diagnostic and therapeutic interventions creating better conditions for repair and reversal. Since the major contributors arise from the intracellular compartment, in this review, we have attempted to focus on the DAMP molecules arising from the intracellular compartments and their specific roles in the neurodegenerative events explaining their downstream mediators and signaling. Moreover, we have tried to cover the latest interventions in terms of DAMPs as clinical biomarkers which can assist in detecting the disease and also target it to reduce the innate-immune activation response which can help in reducing the sterile neuroinflammation having an integral role in the neurodegenerative processes.
Collapse
|
2
|
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: 0.3] [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
|
3
|
Wang X, Ge P. Parthanatos in the pathogenesis of nervous system diseases. Neuroscience 2020; 449:241-250. [DOI: 10.1016/j.neuroscience.2020.09.049] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 09/16/2020] [Accepted: 09/22/2020] [Indexed: 12/12/2022]
|
4
|
Liu J, Guo M, Lv Z, Wang Z, Shao Y, Li C. A cyclophilin A (CypA) from Apostichopus japonicus modulates NF-κB translocation as a cofactor. FISH & SHELLFISH IMMUNOLOGY 2020; 98:728-737. [PMID: 31740398 DOI: 10.1016/j.fsi.2019.11.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 11/05/2019] [Accepted: 11/09/2019] [Indexed: 06/10/2023]
Abstract
As a ubiquitously expressed protein, cyclophilin A (CypA) is involved in a variety of pathological process, including immune suppression, inflammation, cell apoptosis, viral infection and stress response. However, the functional roles of CypA were largely unknown in economic marine animals. In this report, a novel CypA gene from sea cucumber Apostichopus japonicus (designated as AjCypA) was cloned and its function roles in immune responses were explored. The full-length cDNA of AjCypA was 1297 bp containing an open reading frame of 489 bp encoding a putative protein of 162 amino acids (aa). A conserved cyclophilin-like domain (CLD) with PPIase signature was located from 5 to 155 aa sequences in AjCypA, in which five necessary aa residues was totally conserved. In healthy sea cucumbers, AjCypA was expressed in all detected tissues, with highly expressed in muscles and weakly expressed in coelomocytes. AjCypA transcripts was significantly induced 8.08-fold and 5.65-fold in coelomocytes when sea cucumbers challenged with Vibrio splendidus in vivo and LPS in vitro, respectively. The expression pattern is similar with the expression of AjRel in the same condition. Moreover, GST pull-down and immunofluorescence analysis both revealed that AjCypA might be interacted with AjRel. Furthermore, AjCypA knockdown not only inhibited the expression of inflammation cytokines, but also suppressed the translocation of AjRel in nucleus induced by LPS. Taken together, our results suggested that AjCypA play key roles in V. splendidus mediated immune responses via suppressing the nuclear translocation of AjRel activity in sea cucumber.
Collapse
Affiliation(s)
- Jiqing Liu
- 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
| | - Zhimeng Lv
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, PR China
| | - Zhenhui Wang
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, PR China
| | - Yina Shao
- 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
|
5
|
Chaudhuri P, Prajapati KP, Anand BG, Dubey K, Kar K. Amyloid cross-seeding raises new dimensions to understanding of amyloidogenesis mechanism. Ageing Res Rev 2019; 56:100937. [PMID: 31430565 DOI: 10.1016/j.arr.2019.100937] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 06/21/2019] [Accepted: 07/23/2019] [Indexed: 12/12/2022]
Abstract
Hallmarks of most of the amyloid pathologies are surprisingly found to be heterocomponent entities such as inclusions and plaques which contain diverse essential proteins and metabolites. Experimental studies have already revealed the occurrence of coaggregation and cross-seeding during amyloid formation of several proteins and peptides, yielding multicomponent assemblies of amyloid nature. Further, research reports on the co-occurrence of more than one type of amyloid-linked pathologies in the same individual suggest the possible cross-talk among the disease related amyloidogenic protein species during their amyloid growth. In this review paper, we have tried to gain more insight into the process of coaggregation and cross-seeding during amyloid aggregation of proteins, particularly focusing on their relevance to the pathogenesis of the protein misfolding diseases. Revelation of amyloid cross-seeding and coaggregation seems to open new dimensions in our mechanistic understanding of amyloidogenesis and such knowledge may possibly inspire better designing of anti-amyloid therapeutics.
Collapse
|
6
|
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: 0.3] [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
|
7
|
Saw PE, Zhang A, Nie Y, Zhang L, Xu Y, Xu X. Tumor-Associated Fibronectin Targeted Liposomal Nanoplatform for Cyclophilin A siRNA Delivery and Targeted Malignant Glioblastoma Therapy. Front Pharmacol 2018; 9:1194. [PMID: 30386245 PMCID: PMC6199375 DOI: 10.3389/fphar.2018.01194] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 09/28/2018] [Indexed: 12/11/2022] Open
Abstract
Malignant glioblastoma (GBM) is the most aggressive brain cancer that has a very low survival rate. With the rapid development of nanotechnology in the past few decades, the use of nanoparticles (NPs) for nucleic acid delivery is expected to have a revolutionary impact on GBM therapy. However, clinical success in GBM therapy remains a formidable challenge, mainly due to suboptimal in vivo delivery of therapeutics to glioma cells. Herein, we developed an aptamer-like peptide (aptide)-decorated liposomal nanoplatform for systemic small interfering RNA (siRNA) delivery and targeted GBM therapy. This nanoplatform is mainly composed of the following key components: (i) classic liposome structure with an aqueous core that can encapsulate therapeutic siRNA; (ii) hydrophilic polyethylene glycol (PEG) chains on the outer shell to prolong blood circulation; and (iii) surface-encoded aptide to specifically target the extra-domain B (EDB) of fibronectin that over-expressed on glioma cells. After systemic administration of these new siRNA delivery NPs, they can target the glioma cells and efficiently inhibit the GBM tumor growth by silencing the expression of cyclophilin A (CypA), which is up-regulated in brain cancer and plays an important role in malignant transformation of brain cancer and maintaining glioma cell stemness. These results suggest that the reported RNA interference (RNAi) NP platform herein could become an effective tool for targeted GBM therapy.
Collapse
Affiliation(s)
- Phei Er Saw
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ao Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yan Nie
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lei Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yingjie Xu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoding Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
8
|
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: 1.6] [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
|
9
|
Dongsheng Z, Zhiguang F, Junfeng J, Zifan L, Li W. Cyclophilin A Aggravates Collagen-Induced Arthritis via Promoting Classically Activated Macrophages. Inflammation 2018; 40:1761-1772. [PMID: 28756520 DOI: 10.1007/s10753-017-0619-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Activated macrophages exhibiting diverse phenotypes and various functions contribute to the pathogenesis or amelioration of different diseases like cancer, inflammation, and infectious and autoimmune diseases. However, the mechanisms of macrophage polarization in inflamed joint and its effects on rheumatoid arthritis (RA) are still not clarified. This study is designed to explore the effects of cyclophilin A (CypA) on macrophage polarization and describe the underlying mechanisms. Collagen-induced arthritis (CIA) was employed to address the pro-arthritic effects of CypA. Flow cytometry was performed to investigate the populations of M1 and M2 macrophages in synovial tissues of the mice. Knockdown or overexpression of CypA macrophage cells was used to study the functions of CypA on macrophage polarization. Western blot was carried out to examine the potential signaling pathways. We found that CypA aggravated the severity of CIA in mice, as assessed by the increase of clinical score of inflammation, cartilage damage, and bone erosion. Moreover, the level of cytokines, such as IL-6, IL-1β, and IL-17, and the number of pro-inflammatory macrophages in synovial fluid were significantly elevated. In accordance with our observation, CypA dysregulation could actually influence the M1 macrophages polarization and pro-inflammatory cytokines production. Further mechanism study disclosed that CypA could regulate the transcriptional activity of NF-κB, the pivotal transcriptional factor regulating M1 polarization, dependent of its PPIase activity. Our findings provide evidence that PPIase CypA promoted macrophages polarization toward pro-inflammatory M1 phenotype via transcriptional activating NF-κB, thus leading to aggravated arthritis.
Collapse
Affiliation(s)
- Zhai Dongsheng
- State Key Laboratory of Cancer Biology, Department of Pharmacogenomics, School of Pharmacy, Fourth Military Medical University, No. 169 West Changle Road, Xi'an, 710032, Shaanxi, People's Republic of China
| | - Fu Zhiguang
- Institute of Stomatology, General Hospital of Chinese PLA, Beijing, 10085, People's Republic of China
| | - Jia Junfeng
- Department of Clinical Immunology, State Key Discipline of Cell Biology, First Affiliated Hospital, Fourth Military Medical University, No. 15 West Changle Road, Xi'an, 710032, Shaanxi, People's Republic of China
| | - Lu Zifan
- State Key Laboratory of Cancer Biology, Department of Pharmacogenomics, School of Pharmacy, Fourth Military Medical University, No. 169 West Changle Road, Xi'an, 710032, Shaanxi, People's Republic of China.
| | - Wang Li
- State Key Laboratory of Cancer Biology, Department of Pharmacogenomics, School of Pharmacy, Fourth Military Medical University, No. 169 West Changle Road, Xi'an, 710032, Shaanxi, People's Republic of China.
| |
Collapse
|
10
|
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: 756] [Impact Index Per Article: 108.0] [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
|
11
|
Li T, Yan B, Ma Y, Weng J, Yang S, Zhao N, Wang X, Sun X. Ubiquitin-specific protease 4 promotes hepatocellular carcinoma progression via cyclophilin A stabilization and deubiquitination. Cell Death Dis 2018; 9:148. [PMID: 29396555 PMCID: PMC5833721 DOI: 10.1038/s41419-017-0182-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 11/21/2017] [Accepted: 11/22/2017] [Indexed: 12/19/2022]
Abstract
Ubiquitin-specific protease 4 (USP4) is a member of the deubiquitinating enzyme family, which plays an important role in human tumor diseases. However, the mechanisms by which USP4 facilitates tumor development, especially in hepatocellular carcinoma (HCC), remain unclear. Clinically, we found that USP4 is overexpressed in human HCC tissues compared with adjacent non-tumoral tissues and is significantly correlated with malignant phenotype characteristics, including tumor size, tumor number, differentiation, serum alpha-fetoprotein level, and vascular invasion. Moreover, Kaplan-Meier survival analysis showed a poor overall survival rate in patients with USP4-overexpressing tumors. Analyses of univariate and multivariate Cox proportional hazard models indicated that USP4 is a prognostic biomarker for poor outcome. Using in vitro and in vivo assays, we demonstrated that USP4 overexpression enhanced HCC cell growth, migration, and invasion. Mechanistically, cyclophilin A (CypA) was identified as an important molecule for USP4-mediated oncogenic activity in HCC. We observed that USP4 interacted with CypA and inhibited CypA degradation via deubiquitination in HCC cells. Subsequently, the USP4/CypA complex activated the MAPK signaling pathway and prevented CrkII phosphorylation. These data suggest that USP4 acts as a novel prognostic marker, offering potential therapeutic opportunities for HCC.
Collapse
Affiliation(s)
- Tianyi Li
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Bin Yan
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Yang Ma
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Junyong Weng
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Shouwen Yang
- Department of Gynaecology, International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Nan Zhao
- Department of General Surgery, Shanghai General Hospital, Nanjing Medical University, Shanghai, China
| | - Xiaoliang Wang
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China.
| | - Xing Sun
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China.
| |
Collapse
|
12
|
Zhou T, Ahmad TK, Gozda K, Truong J, Kong J, Namaka M. Implications of white matter damage in amyotrophic lateral sclerosis (Review). Mol Med Rep 2017; 16:4379-4392. [PMID: 28791401 PMCID: PMC5646997 DOI: 10.3892/mmr.2017.7186] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Accepted: 06/09/2017] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease, which involves the progressive degeneration of motor neurons. ALS has long been considered a disease of the grey matter; however, pathological alterations of the white matter (WM), including axonal loss, axonal demyelination and oligodendrocyte death, have been reported in patients with ALS. The present review examined motor neuron death as the primary cause of ALS and evaluated the associated WM damage that is guided by neuronal‑glial interactions. Previous studies have suggested that WM damage may occur prior to the death of motor neurons, and thus may be considered an early indicator for the diagnosis and prognosis of ALS. However, the exact molecular mechanisms underlying early‑onset WM damage in ALS have yet to be elucidated. The present review explored the detailed anatomy of WM and identified several pathological mechanisms that may be implicated in WM damage in ALS. In addition, it associated the pathophysiological alterations of WM, which may contribute to motor neuron death in ALS, with similar mechanisms of WM damage that are involved in multiple sclerosis (MS). Furthermore, the early detection of WM damage in ALS, using neuroimaging techniques, may lead to earlier therapeutic intervention, using immunomodulatory treatment strategies similar to those used in relapsing‑remitting MS, aimed at delaying WM damage in ALS. Early therapeutic approaches may have the potential to delay motor neuron damage and thus prolong the survival of patients with ALS. The therapeutic interventions that are currently available for ALS are only marginally effective. However, early intervention with immunomodulatory drugs may slow the progression of WM damage in the early stages of ALS, thus delaying motor neuron death and increasing the life expectancy of patients with ALS.
Collapse
Affiliation(s)
- Ting Zhou
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
- Department of Human Anatomy and Cell Science, College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Tina Khorshid Ahmad
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Kiana Gozda
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Jessica Truong
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Jiming Kong
- Department of Human Anatomy and Cell Science, College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Michael Namaka
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
- Department of Human Anatomy and Cell Science, College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- College of Pharmacy, Third Military Medical University, Chongqing 400038, P.R. China
- Department of Medical Rehabilitation, College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T6, Canada
- Department of Internal Medicine, College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 1R9, Canada
| |
Collapse
|
13
|
Targeting Extracellular Cyclophilin A Reduces Neuroinflammation and Extends Survival in a Mouse Model of Amyotrophic Lateral Sclerosis. J Neurosci 2016; 37:1413-1427. [PMID: 28011744 DOI: 10.1523/jneurosci.2462-16.2016] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 10/24/2016] [Accepted: 11/15/2016] [Indexed: 12/13/2022] Open
Abstract
Neuroinflammation is a major hallmark of amyotrophic lateral sclerosis (ALS), which is currently untreatable. Several anti-inflammatory compounds have been evaluated in patients and in animal models of ALS, but have been proven disappointing in part because effective targets have not yet been identified. Cyclophilin A, also known as peptidylprolyl cis-/trans-isomerase A (PPIA), as a foldase is beneficial intracellularly, but extracellularly has detrimental functions. We found that extracellular PPIA is a mediator of neuroinflammation in ALS. It is a major inducer of matrix metalloproteinase 9 and is selectively toxic for motor neurons. High levels of PPIA were found in the CSF of SOD1G93A mice and rats and sporadic ALS patients, suggesting that our findings may be relevant for familial and sporadic cases. A specific inhibitor of extracellular PPIA, MM218, given at symptom onset, rescued motor neurons and extended survival in the SOD1G93A mouse model of familial ALS by 11 d. The treatment resulted in the polarization of glia toward a prohealing phenotype associated with reduced NF-κB activation, proinflammatory markers, endoplasmic reticulum stress, and insoluble phosphorylated TDP-43. Our results indicates that extracellular PPIA is a promising druggable target for ALS and support further studies to develop a therapy to arrest or slow the progression of the disease in patients.SIGNIFICANCE STATEMENT We provide evidence that extracellular cyclophilin A, also known as peptidylprolyl cis-/trans-isomerase A (PPIA), is a mediator of the neuroinflammatory reaction in amyotrophic lateral sclerosis (ALS) and is toxic for motor neurons. Supporting this, a specific extracellular PPIA inhibitor reduced neuroinflammation, rescued motor neurons, and extended survival in the SOD1G93A mouse model of familial ALS. Our findings suggest selective pharmacological inhibition of extracellular PPIA as a novel therapeutic strategy, not only for SOD1-linked ALS, but possibly also for sporadic ALS. This approach aims to address the neuroinflammatory reaction that is a major hallmark of ALS. However, given the complexity of the disease, a combination of therapeutic approaches may be necessary.
Collapse
|
14
|
Caballero-Hernandez D, Toscano MG, Cejudo-Guillen M, Garcia-Martin ML, Lopez S, Franco JM, Quintana FJ, Roodveldt C, Pozo D. The ‘Omics’ of Amyotrophic Lateral Sclerosis. Trends Mol Med 2016; 22:53-67. [DOI: 10.1016/j.molmed.2015.11.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 10/29/2015] [Accepted: 11/08/2015] [Indexed: 12/11/2022]
|
15
|
Zhang L, Li Z, Zhang B, He H, Bai Y. PPIA is a novel adipogenic factor implicated in obesity. Obesity (Silver Spring) 2015; 23:2093-100. [PMID: 26347493 DOI: 10.1002/oby.21208] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 05/26/2015] [Accepted: 06/08/2015] [Indexed: 12/22/2022]
Abstract
OBJECTIVE To assess the role of peptidyl-prolyl cis/trans isomerase a (PPIA) in adipogenesis and obesity. METHODS Fat mass and adipocyte sizes of PPIA-/- and wild-type mice were compared. The role of PPIA in adipocyte differentiation of 3T3L1 and MEFs cells was analyzed by gene silencing and overexpression. The roles of PPIA in obesity were observed on a high-fat diet obesity model and a gestational diabetes obesity model. RESULTS PPIA-/- mice had significantly less fat than PPIA+/+ mice. The adipocyte size of PPIA-/- mice was significantly smaller than wild type. Silencing PPIA in 3T3L1 cells significantly impaired its adipocyte differentiation ability. Similarly, MEFs from PPIA-/- mice differentiated less than wild type, while their differentiation ability was restored by PPIA overexpression. PPIA-silenced 3T3L1 cells had significantly lower expression of PPARG, C/EBPA, and C/EBPB at late stage of adipocyte differentiation, which was the same in PPIA-/- MEFs. When fed a high-fat diet, PPIA-/- mice gained significantly less weight than wild type, accompanied by reduced PPARG, C/EBPA, and C/EBPB expression. PPIA expression was significantly higher in adipose tissue of gestational diabetes rat offspring, which had higher inguinal fat/body weight ratios than normal rat offspring. CONCLUSIONS PPIA was a novel adipogenic factor important in obesity.
Collapse
Affiliation(s)
- Lihong Zhang
- Department of Medical Genetics, Third Military Medical University, Chongqing, People's Republic of China
- Department of Gynecology and Obstetrics, Xinqiao Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Zhen Li
- Department of Gynecology and Obstetrics, Xinqiao Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Bo Zhang
- Department of Medical Genetics, Third Military Medical University, Chongqing, People's Republic of China
| | - Haiyang He
- Institute of Immunology, Third Military Medical University, Chongqing, People's Republic of China
| | - Yun Bai
- Department of Medical Genetics, Third Military Medical University, Chongqing, People's Republic of China
| |
Collapse
|
16
|
Li W, Fotinos A, Wu Q, Chen Y, Zhu Y, Baranov S, Tu Y, Zhou EW, Sinha B, Kristal BS, Wang X. N-acetyl-l-tryptophan delays disease onset and extends survival in an amyotrophic lateral sclerosis transgenic mouse model. Neurobiol Dis 2015; 80:93-103. [DOI: 10.1016/j.nbd.2015.05.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 04/25/2015] [Accepted: 05/08/2015] [Indexed: 12/14/2022] Open
|
17
|
Blair LJ, Baker JD, Sabbagh JJ, Dickey CA. The emerging role of peptidyl-prolyl isomerase chaperones in tau oligomerization, amyloid processing, and Alzheimer's disease. J Neurochem 2015; 133:1-13. [PMID: 25628064 DOI: 10.1111/jnc.13033] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 12/12/2014] [Accepted: 01/05/2015] [Indexed: 12/20/2022]
Abstract
Peptidyl-prolyl cis/trans isomerases (PPIases), a unique family of molecular chaperones, regulate protein folding at proline residues. These residues are abundant within intrinsically disordered proteins, like the microtubule-associated protein tau. Tau has been shown to become hyperphosphorylated and accumulate as one of the two main pathological hallmarks in Alzheimer's disease, the other being amyloid beta (Ab). PPIases, including Pin1, FK506-binding protein (FKBP) 52, FKBP51, and FKBP12, have been shown to interact with and regulate tau biology. This interaction is particularly important given the numerous proline-directed phosphorylation sites found on tau and the role phosphorylation has been found to play in pathogenesis. This regulation then affects downstream aggregation and oligomerization of tau. However, many PPIases have yet to be explored for their effects on tau biology, despite the high likelihood of interaction based on proline content. Moreover, Pin1, FKBP12, FKBP52, cyclophilin (Cyp) A, CypB, and CypD have been shown to also regulate Ab production or the toxicity associated with Ab pathology. Therefore, PPIases directly and indirectly regulate pathogenic protein multimerization in Alzheimer's disease and represent a family rich in targets for modulating the accumulation and toxicity.
Collapse
Affiliation(s)
- Laura J Blair
- Department of Molecular Medicine, Byrd Alzheimer's Institute, University of South Florida, Tampa, Florida, USA
| | | | | | | |
Collapse
|
18
|
Takata M, Tanaka H, Kimura M, Nagahara Y, Tanaka K, Kawasaki K, Seto M, Tsuruma K, Shimazawa M, Hara H. Fasudil, a rho kinase inhibitor, limits motor neuron loss in experimental models of amyotrophic lateral sclerosis. Br J Pharmacol 2014; 170:341-51. [PMID: 23763343 DOI: 10.1111/bph.12277] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 05/28/2013] [Accepted: 06/01/2013] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder with no effective treatment. Fasudil hydrochloride (fasudil), a potent rho kinase (ROCK) inhibitor, is useful for the treatment of ischaemic diseases. In previous reports, fasudil improved pathology in mouse models of Alzheimer's disease and spinal muscular atrophy, but there is no evidence in that it can affect ALS. We therefore investigated its effects on experimental models of ALS. EXPERIMENTAL APPROACH In mice motor neuron (NSC34) cells, the neuroprotective effect of hydroxyfasudil (M3), an active metabolite of fasudil, and its mechanism were evaluated. Moreover, the effects of fasudil, 30 and 100 mg·kg(-1), administered via drinking water to mutant superoxide dismutase 1 (SOD1(G93A)) mice were tested by measuring motor performance, survival time and histological changes, and its mechanism investigated. KEY RESULTS M3 prevented motor neuron cell death induced by SOD1(G93A). Furthermore, M3 suppressed both the increase in ROCK activity and phosphorylated phosphatase and tensin homologue deleted on chromosome 10 (PTEN), and the reduction in phosphorylated Akt induced by SOD1(G93A). These effects of M3 were attenuated by treatment with a PI3K inhibitor (LY294002). Moreover, fasudil slowed disease progression, increased survival time and reduced motor neuron loss, in SOD1(G93A) mice. Fasudil also attenuated the increase in ROCK activity and PTEN, and the reduction in Akt in SOD1(G93A) mice. CONCLUSIONS AND IMPLICATIONS These findings indicate that fasudil may be effective at suppressing motor neuron degeneration and symptom progression in ALS. Hence, fasudil may have potential as a therapeutic agent for ALS treatment.
Collapse
Affiliation(s)
- M Takata
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Ghavami S, Shojaei S, Yeganeh B, Ande SR, Jangamreddy JR, Mehrpour M, Christoffersson J, Chaabane W, Moghadam AR, Kashani HH, Hashemi M, Owji AA, Łos MJ. Autophagy and apoptosis dysfunction in neurodegenerative disorders. Prog Neurobiol 2013; 112:24-49. [PMID: 24211851 DOI: 10.1016/j.pneurobio.2013.10.004] [Citation(s) in RCA: 773] [Impact Index Per Article: 64.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2012] [Revised: 10/08/2013] [Accepted: 10/15/2013] [Indexed: 12/12/2022]
Abstract
Autophagy and apoptosis are basic physiologic processes contributing to the maintenance of cellular homeostasis. Autophagy encompasses pathways that target long-lived cytosolic proteins and damaged organelles. It involves a sequential set of events including double membrane formation, elongation, vesicle maturation and finally delivery of the targeted materials to the lysosome. Apoptotic cell death is best described through its morphology. It is characterized by cell rounding, membrane blebbing, cytoskeletal collapse, cytoplasmic condensation, and fragmentation, nuclear pyknosis, chromatin condensation/fragmentation, and formation of membrane-enveloped apoptotic bodies, that are rapidly phagocytosed by macrophages or neighboring cells. Neurodegenerative disorders are becoming increasingly prevalent, especially in the Western societies, with larger percentage of members living to an older age. They have to be seen not only as a health problem, but since they are care-intensive, they also carry a significant economic burden. Deregulation of autophagy plays a pivotal role in the etiology and/or progress of many of these diseases. Herein, we briefly review the latest findings that indicate the involvement of autophagy in neurodegenerative diseases. We provide a brief introduction to autophagy and apoptosis pathways focusing on the role of mitochondria and lysosomes. We then briefly highlight pathophysiology of common neurodegenerative disorders like Alzheimer's diseases, Parkinson's disease, Huntington's disease and Amyotrophic lateral sclerosis. Then, we describe functions of autophagy and apoptosis in brain homeostasis, especially in the context of the aforementioned disorders. Finally, we discuss different ways that autophagy and apoptosis modulation may be employed for therapeutic intervention during the maintenance of neurodegenerative disorders.
Collapse
Affiliation(s)
- Saeid Ghavami
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Canada; Manitoba Institute of Child Health, Department of Physiology, University of Manitoba, Winnipeg, Canada; St. Boniface Research Centre, University of Manitoba, Winnipeg, Canada
| | - Shahla Shojaei
- Department of Biochemistry, Recombinant Protein Laboratory, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Behzad Yeganeh
- Manitoba Institute of Child Health, Department of Physiology, University of Manitoba, Winnipeg, Canada; Hospital for Sick Children Research Institute, Department of Physiology and Experimental Medicine, University of Toronto, Canada
| | - Sudharsana R Ande
- Department of Internal Medicine, University of Manitoba, Winnipeg, Canada
| | - Jaganmohan R Jangamreddy
- Department of Clinical and Experimental Medicine (IKE), Integrative Regenerative Medicine Center (IGEN), Division of Cell Biology, Linkoping University, Linkoping, Sweden
| | - Maryam Mehrpour
- INSERM U845, Research Center "Growth & Signaling" Paris Descartes University Medical School, France
| | - Jonas Christoffersson
- Department of Clinical and Experimental Medicine (IKE), Integrative Regenerative Medicine Center (IGEN), Division of Cell Biology, Linkoping University, Linkoping, Sweden
| | - Wiem Chaabane
- Department of Clinical and Experimental Medicine (IKE), Integrative Regenerative Medicine Center (IGEN), Division of Cell Biology, Linkoping University, Linkoping, Sweden; Department of Biology, Faculty of Sciences, Tunis University, Tunis, Tunisia
| | | | - Hessam H Kashani
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Canada; Manitoba Institute of Child Health, Department of Physiology, University of Manitoba, Winnipeg, Canada
| | - Mohammad Hashemi
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran; Cellular and Molecular Biology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Ali A Owji
- Department of Biochemistry, Recombinant Protein Laboratory, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Marek J Łos
- Department of Clinical and Experimental Medicine (IKE), Integrative Regenerative Medicine Center (IGEN), Division of Cell Biology, Linkoping University, Linkoping, Sweden.
| |
Collapse
|
20
|
Cyclophilin A: a key player for human disease. Cell Death Dis 2013; 4:e888. [PMID: 24176846 PMCID: PMC3920964 DOI: 10.1038/cddis.2013.410] [Citation(s) in RCA: 333] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 09/10/2013] [Accepted: 09/11/2013] [Indexed: 01/23/2023]
Abstract
Cyclophilin A (CyPA) is a ubiquitously distributed protein belonging to the immunophilin family. CyPA has peptidyl prolyl cis-trans isomerase (PPIase) activity, which regulates protein folding and trafficking. Although CyPA was initially believed to function primarily as an intracellular protein, recent studies have revealed that it can be secreted by cells in response to inflammatory stimuli. Current research in animal models and humans has provided compelling evidences supporting the critical function of CyPA in several human diseases. This review discusses recently available data about CyPA in cardiovascular diseases, viral infections, neurodegeneration, cancer, rheumatoid arthritis, sepsis, asthma, periodontitis and aging. It is believed that further elucidations of the role of CyPA will provide a better understanding of the molecular mechanisms underlying these diseases and will help develop novel pharmacological therapies.
Collapse
|
21
|
Tanaka H, Shimazawa M, Takata M, Kaneko H, Tsuruma K, Ikeda T, Warita H, Aoki M, Yamada M, Takahashi H, Hozumi I, Minatsu H, Inuzuka T, Hara H. ITIH4 and Gpx3 are potential biomarkers for amyotrophic lateral sclerosis. J Neurol 2013; 260:1782-97. [PMID: 23436019 DOI: 10.1007/s00415-013-6877-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 02/12/2013] [Accepted: 02/14/2013] [Indexed: 11/30/2022]
Abstract
The diagnosis of amyotrophic lateral sclerosis (ALS) is difficult due to lack of definitive biomarkers. Our aim was to identify characteristic serum protein patterns that could provide candidate biomarkers for ALS. We divided mutant superoxide dismutase-1 (SOD1)(H46R) rats into three groups based on disease progression: pre-symptom (90 days), onset, and end-stage. After separation of serum proteins using two-dimensional electrophoresis, we selected clear protein spots and identified two candidate proteins-inter-alpha-trypsin inhibitor heavy chain H4 (ITIH4) and glutathione peroxidase 3 (Gpx3). The 120 kDa ITIH4 increased at the onset of the disease and the 85 kDa ITIH4, a cleaved form, at the end-stage in the sera of the SOD1(H46R) rats. Expression of the 85 kDa ITIH4 was substantial in ALS compared with controls or patients with muscular dystrophy, Alzheimer diseases, or Parkinson diseases. The Gpx3 protein levels in the sera of SOD1(H46R) rats were upregulated pre-symptom and gradually decreased as the disease progressed. The Gpx3 protein levels were lower in the sera of the patients with ALS than in other diseases. These results indicate that ITIH4 and Gpx3 are potential biomarkers for ALS.
Collapse
Affiliation(s)
- Hirotaka Tanaka
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu 501-1196, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
The potential of GPNMB as novel neuroprotective factor in amyotrophic lateral sclerosis. Sci Rep 2012; 2:573. [PMID: 22891158 PMCID: PMC3417778 DOI: 10.1038/srep00573] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 07/27/2012] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an incurable and fatal neurodegenerative disease characterized by the loss of motor neurons. Despite substantial research, the causes of ALS remain unclear. Glycoprotein nonmetastatic melanoma protein B (GPNMB) was identified as an ALS-related factor using DNA microarray analysis with mutant superoxide dismutase (SOD1G93A) mice. GPNMB was greatly induced in the spinal cords of ALS patients and a mouse model as the disease progressed. It was especially expressed in motor neurons and astrocytes. In an NSC34 cell line, glycosylation of GPNMB was inhibited by interaction with SOD1G93A, increasing motor neuron vulnerability, whereas extracellular fragments of GPNMB secreted from activated astrocytes attenuated the neurotoxicity of SOD1G93A in neural cells. Furthermore, GPNMB expression was substantial in the sera of sporadic ALS patients than that of other diseased patients. This study suggests that GPNMB can be a target for therapeutic intervention for suppressing motor neuron degeneration in ALS.
Collapse
|
23
|
Abstract
Apoptosis-inducing factor (AIF) is a flavin adenine dinucleotide-containing, NADH-dependent oxidoreductase residing in the mitochondrial intermembrane space whose specific enzymatic activity remains unknown. Upon an apoptotic insult, AIF undergoes proteolysis and translocates to the nucleus, where it triggers chromatin condensation and large-scale DNA degradation in a caspase-independent manner. Besides playing a key role in execution of caspase-independent cell death, AIF has emerged as a protein critical for cell survival. Analysis of in vivo phenotypes associated with AIF deficiency and defects, and identification of its mitochondrial, cytoplasmic, and nuclear partners revealed the complexity and multilevel regulation of AIF-mediated signal transduction and suggested an important role of AIF in the maintenance of mitochondrial morphology and energy metabolism. The redox activity of AIF is essential for optimal oxidative phosphorylation. Additionally, the protein is proposed to regulate the respiratory chain indirectly, through assembly and/or stabilization of complexes I and III. This review discusses accumulated data with respect to the AIF structure and outlines evidence that supports the prevalent mechanistic view on the apoptogenic actions of the flavoprotein, as well as the emerging concept of AIF as a redox sensor capable of linking NAD(H)-dependent metabolic pathways to apoptosis.
Collapse
Affiliation(s)
- Irina F Sevrioukova
- Department of Molecular Biology and Biochemistry, University of California-Irvine, CA 92697-3900, USA.
| |
Collapse
|