51
|
|
52
|
Chung CL, Lawrence I, Hoffman M, Elgindi D, Nadhan K, Potnis M, Jin A, Sershon C, Binnebose R, Lorenzini A, Sell C. Topical rapamycin reduces markers of senescence and aging in human skin: an exploratory, prospective, randomized trial. GeroScience 2019; 41:861-869. [PMID: 31761958 PMCID: PMC6925069 DOI: 10.1007/s11357-019-00113-y] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 10/09/2019] [Indexed: 01/07/2023] Open
Abstract
Aging is a major risk factor for the majority of human diseases, and the development of interventions to reduce the intrinsic rate of aging is expected to reduce the risk for age-related diseases including cardiovascular disease, cancer, and dementia. In the skin, aging manifests itself in photodamage and dermal atrophy, with underlying tissue reduction and impaired barrier function. To determine whether rapamycin, an FDA-approved drug targeting the mechanistic target of rapamycin (mTOR) complex, can reduce senescence and markers of aging in human skin, an exploratory, placebo-controlled, interventional trial was conducted in a clinical dermatology setting. Participants were greater than 40 years of age with evidence of age-related photoaging and dermal volume loss and no major morbidities. Thirty-six participants were enrolled in the study, and nineteen discontinued or were lost to follow-up. A significant (P = 0.008) reduction in p16INK4A protein levels and an increase in collagen VII protein levels (P = 0.0077) were observed among participants at the end of the study. Clinical improvement in skin appearance was noted in multiple participants, and immunohistochemical analysis revealed improvement in histological appearance of skin tissue. Topical rapamycin reduced the expression of the p16INK4A protein consistent with a reduction in cellular senescence. This change was accompanied by relative improvement in clinical appearance of the skin and histological markers of aging and by an increase in collagen VII, which is critical to the integrity of the basement membrane. These results indicate that rapamycin treatment is a potential anti-aging therapy with efficacy in humans.Trial registration ClinicalTrials.gov Identifier: NCT03103893.
Collapse
Affiliation(s)
- Christina Lee Chung
- Department of Dermatology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Ibiyonu Lawrence
- Department of Medicine, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Melissa Hoffman
- Department of Dermatology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Dareen Elgindi
- Department of Dermatology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Kumar Nadhan
- Department of Dermatology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Manali Potnis
- Department of Pathology and Laboratory Medicine, Drexel University College of Medicine, 245 N 15th Street, Philadelphia, PA, 19102, USA
| | - Annie Jin
- Department of Dermatology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Catlin Sershon
- Department of Dermatology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Rhonda Binnebose
- Department of Pathology and Laboratory Medicine, Drexel University College of Medicine, 245 N 15th Street, Philadelphia, PA, 19102, USA
| | - Antonello Lorenzini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Christian Sell
- Department of Pathology and Laboratory Medicine, Drexel University College of Medicine, 245 N 15th Street, Philadelphia, PA, 19102, USA. .,Department of Biochemistry, Drexel University College of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
53
|
Hyttinen JMT, Kannan R, Felszeghy S, Niittykoski M, Salminen A, Kaarniranta K. The Regulation of NFE2L2 (NRF2) Signalling and Epithelial-to-Mesenchymal Transition in Age-Related Macular Degeneration Pathology. Int J Mol Sci 2019; 20:ijms20225800. [PMID: 31752195 PMCID: PMC6888570 DOI: 10.3390/ijms20225800] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/14/2019] [Accepted: 11/14/2019] [Indexed: 12/19/2022] Open
Abstract
Age-related macular degeneration (AMD) is a mounting cause of loss of sight in the elderly in the developed countries, a trend enhanced by the continual ageing of the population. AMD is a multifactorial and only partly understood, malady. Unfortunately, there is no effective treatment for most AMD patients. It is known that oxidative stress (OS) damages the retinal pigment epithelium (RPE) and contributes to the progression of AMD. We review here the potential importance of two OS-related cellular systems in relation to AMD. First, the nuclear factor erythroid 2-related factor 2 (NFE2L2; NRF2)-mediated OS response signalling pathway is important in the prevention of oxidative damage and a failure of this system could be critical in the development of AMD. Second, epithelial-to-mesenchymal transition (EMT) represents a change in the cellular phenotype, which ultimately leads to the fibrosis encountered in RPE, a characteristic of AMD. Many of the pathways triggering EMT are promoted by OS. The possible interconnections between these two signalling routes are discussed here. From a broader perspective, the control of NFE2L2 and EMT as ways of preventing OS-derived cellular damage could be potentially valuable in the therapy of AMD.
Collapse
Affiliation(s)
- Juha M. T. Hyttinen
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland;
- Correspondence:
| | - Ram Kannan
- The Stephen J. Ryan Initiative for Macular Research (RIMR), Doheny Eye Institute, DVRC 203, 1355 San Pablo Street, Los Angeles, CA 90033, USA
| | - Szabolcs Felszeghy
- Institute of Biomedicine, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland;
- Institute of Dentistry, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland
| | - Minna Niittykoski
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland;
| | - Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland;
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland;
- Department of Ophthalmology, Kuopio University Hospital, P.O. Box 100, 70029 KYS Kuopio, Finland
| |
Collapse
|
54
|
Geltinger F, Tevini J, Briza P, Geiser A, Bischof J, Richter K, Felder T, Rinnerthaler M. The transfer of specific mitochondrial lipids and proteins to lipid droplets contributes to proteostasis upon stress and aging in the eukaryotic model system Saccharomyces cerevisiae. GeroScience 2019; 42:19-38. [PMID: 31676965 PMCID: PMC7031196 DOI: 10.1007/s11357-019-00103-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 09/11/2019] [Indexed: 01/12/2023] Open
Abstract
Originally Lipid droplets (LDs) were considered as being droplets for lipid storage only. Increasing evidence, however, demonstrates that LDs fulfill a pleiotropy of additional functions. Among them is the modulation of protein as well as lipid homeostasis. Under unfavorable pro-oxidative conditions, proteins can form aggregates which may exceed the overall proteolytic capacity of the proteasome. After stress termination LDs can adjust and support the removal of these aggregates. Additionally, LDs interact with mitochondria, specifically take over certain proteins and thus prevent apoptosis. LDs, which are loaded with these harmful proteins, are subsequently eliminated via lipophagy. Recently it was demonstrated that this autophagic process is a modulator of longevity. LDs do not only eliminate potentially dangerous proteins, but they are also able to prevent lipotoxicity by storing specific lipids. In the present study we used the model organism Saccharomyces cerevisiae to compare the proteome as well as lipidome of mitochondria and LDs under different conditions: replicative aging, stress and apoptosis. In this context we found an accumulation of proteins at LDs, supporting the role of LDs in proteostasis. Additionally, the composition of main lipid classes such as phosphatidylcholines, phosphatidylethanolamines, phosphatidylinositols, phosphatidylglycerols, triacylglycerols, ceramides, phosphatidic acids and ergosterol of LDs and mitochondria changed during stress conditions and aging.
Collapse
Affiliation(s)
- Florian Geltinger
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Julia Tevini
- Department of Laboratory Medicine, Paracelsus Medical University, Salzburg, Austria
| | - Peter Briza
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Amrito Geiser
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Johannes Bischof
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Klaus Richter
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Thomas Felder
- Department of Laboratory Medicine, Paracelsus Medical University, Salzburg, Austria.
- Obesity Research Unit, Paracelsus Medical University, Salzburg, Austria.
| | - Mark Rinnerthaler
- Department of Biosciences, University of Salzburg, Salzburg, Austria.
| |
Collapse
|
55
|
Lim D, Lee HS, Ku B, Shin HC, Kim SJ. Oligomer Model of PB1 Domain of p62/SQSTM1 Based on Crystal Structure of Homo-Dimer and Calculation of Helical Characteristics. Mol Cells 2019; 42:729-738. [PMID: 31600867 PMCID: PMC6821454 DOI: 10.14348/molcells.2019.0096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 08/14/2019] [Accepted: 09/04/2019] [Indexed: 11/27/2022] Open
Abstract
Autophagy is an important process for protein recycling. Oligomerization of p62/SQSTM1 is an essential step in this process and is achieved in two steps. Phox and Bem1p (PB1) domains can oligomerize through both basic and acidic surfaces in each molecule. The ZZ-type zinc finger (ZZ) domain binds to target proteins and promotes higheroligomerization of p62. This mechanism is an important step in routing target proteins to the autophagosome. Here, we determined the crystal structure of the PB1 homo-dimer and modeled the p62 PB1 oligomers. These oligomer models were represented by a cylindrical helix and were compared with the previously determined electron microscopic map of a PB1 oligomer. To accurately compare, we mathematically calculated the lead length and radius of the helical oligomers. Our PB1 oligomer model fits the electron microscopy map and is both bendable and stretchable as a flexible helical filament.
Collapse
Affiliation(s)
- Dahwan Lim
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141,
Korea
- Department of Biochemistry, Chungnam National University, Daejeon 34134,
Korea
| | - Hye Seon Lee
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141,
Korea
- Department of Biology, Chungnam National University, Daejeon 34134,
Korea
| | - Bonsu Ku
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141,
Korea
| | - Ho-Chul Shin
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141,
Korea
| | - Seung Jun Kim
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141,
Korea
| |
Collapse
|
56
|
Battaglia C, Venturin M, Sojic A, Jesuthasan N, Orro A, Spinelli R, Musicco M, De Bellis G, Adorni F. Candidate Genes and MiRNAs Linked to the Inverse Relationship Between Cancer and Alzheimer's Disease: Insights From Data Mining and Enrichment Analysis. Front Genet 2019; 10:846. [PMID: 31608105 PMCID: PMC6771301 DOI: 10.3389/fgene.2019.00846] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 08/14/2019] [Indexed: 12/22/2022] Open
Abstract
The incidence of cancer and Alzheimer’s disease (AD) increases exponentially with age. A growing body of epidemiological evidence and molecular investigations inspired the hypothesis of an inverse relationship between these two pathologies. It has been proposed that the two diseases might utilize the same proteins and pathways that are, however, modulated differently and sometimes in opposite directions. Investigation of the common processes underlying these diseases may enhance the understanding of their pathogenesis and may also guide novel therapeutic strategies. Starting from a text-mining approach, our in silico study integrated the dispersed biological evidence by combining data mining, gene set enrichment, and protein-protein interaction (PPI) analyses while searching for common biological hallmarks linked to AD and cancer. We retrieved 138 genes (ALZCAN gene set), computed a significant number of enriched gene ontology clusters, and identified four PPI modules. The investigation confirmed the relevance of autophagy, ubiquitin proteasome system, and cell death as common biological hallmarks shared by cancer and AD. Then, from a closer investigation of the PPI modules and of the miRNAs enrichment data, several genes (SQSTM1, UCHL1, STUB1, BECN1, CDKN2A, TP53, EGFR, GSK3B, and HSPA9) and miRNAs (miR-146a-5p, MiR-34a-5p, miR-21-5p, miR-9-5p, and miR-16-5p) emerged as promising candidates. The integrative approach uncovered novel miRNA-gene networks (e.g., miR-146 and miR-34 regulating p62 and Beclin1 in autophagy) that might give new insights into the complex regulatory mechanisms of gene expression in AD and cancer.
Collapse
Affiliation(s)
- Cristina Battaglia
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), University of Milan, Segrate, Italy.,Department of Biomedical Sciences, Institute of Biomedical Technologies-National Research Council (ITB-CNR), Segrate, Italy
| | - Marco Venturin
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), University of Milan, Segrate, Italy
| | - Aleksandra Sojic
- Department of Biomedical Sciences, Institute of Biomedical Technologies-National Research Council (ITB-CNR), Segrate, Italy
| | - Nithiya Jesuthasan
- Department of Biomedical Sciences, Institute of Biomedical Technologies-National Research Council (ITB-CNR), Segrate, Italy
| | - Alessandro Orro
- Department of Biomedical Sciences, Institute of Biomedical Technologies-National Research Council (ITB-CNR), Segrate, Italy
| | - Roberta Spinelli
- Istituto Istruzione Superiore Statale IRIS Versari, Cesano Maderno, Italy
| | - Massimo Musicco
- Department of Biomedical Sciences, Institute of Biomedical Technologies-National Research Council (ITB-CNR), Segrate, Italy
| | - Gianluca De Bellis
- Department of Biomedical Sciences, Institute of Biomedical Technologies-National Research Council (ITB-CNR), Segrate, Italy
| | - Fulvio Adorni
- Department of Biomedical Sciences, Institute of Biomedical Technologies-National Research Council (ITB-CNR), Segrate, Italy
| |
Collapse
|
57
|
Lacava G, Laus F, Amaroli A, Marchegiani A, Censi R, Di Martino P, Yanagawa T, Sabbieti MG, Agas D. P62 deficiency shifts mesenchymal/stromal stem cell commitment toward adipogenesis and disrupts bone marrow homeostasis in aged mice. J Cell Physiol 2019; 234:16338-16347. [PMID: 30740681 DOI: 10.1002/jcp.28299] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 12/04/2018] [Accepted: 12/07/2018] [Indexed: 01/24/2023]
Abstract
With advancing age have been observed bone and bone marrow phenotypic alterations due to the impaired bone tissue homeostatic features, involving bone remodeling, and bone marrow niche ontogeny. The complex "inflamm-aging" pathological scenario that culminates with osteopenia and mesenchymal/stromal and hematopoietic stem cell commitment breakdown, is controlled by cellular and molecular intramural components comprising adapter proteins such as the sequestosome 1 (p62/SQSTM1). p62, a "multiway function" protein, has been reported as an effective anti-inflammatory, bone-building factor. In this view, we considered for the first time the involvement of p62 in aging bone and bone marrow of 1 year and 2 years p62-/- mice. Interestingly, p62 deficiency provoked accelerated osteopenia and impaired niche operational activities within the bone marrow. The above findings unearthed the importance of p62 in mesenchymal stem cell maintenance/differentiation schedule in old animals and provide, at least in part, a mechanistic scenario of p62 action.
Collapse
Affiliation(s)
- Giovanna Lacava
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Fulvio Laus
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Andrea Amaroli
- Department of Surgical and Diagnostic Sciences, University of Genova, Genova, Italy
| | - Andrea Marchegiani
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Roberta Censi
- School of Pharmacy, University of Camerino, Camerino, Italy
| | | | - Toru Yanagawa
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | | | - Dimitrios Agas
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| |
Collapse
|
58
|
Yoshida T, Terabe T, Nagai H, Uchida F, Hasegawa S, Nagao T, Miyabe S, Ishibashi‐Kanno N, Yamagata K, Warabi E, Gosho M, Yanagawa T, Bukawa H. Association between p62 expression and clinicopathological characteristics in oral leukoplakia. Clin Exp Dent Res 2019; 5:389-397. [PMID: 31452949 PMCID: PMC6704027 DOI: 10.1002/cre2.193] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 04/30/2019] [Accepted: 04/30/2019] [Indexed: 12/31/2022] Open
Abstract
Objective Oral leukoplakia is keratinized lesions in the buccal mucosa, tongue, and gingiva. It is the most common oral precancerous lesion; oxidative stresses and irrelevant autophagy have been reported to be the cause of oncogenesis. p62, a cytoplasmic protein induced by oxidative stress, is an adaptor protein involved in the formation of protein aggregates and induction and inhibition of autophagy. The inhibition of autophagy induces p62 overexpression and promotes oncogenesis via the oncogenic signaling pathway. The aim of the present study was to elucidate the involvement of intracellular expression of p62 in oral leukoplakia and to address its potential clinical implementation as a biomarker to predict malignant transformation. Material and Methods Fifty samples from subjects with confirmed oral leukoplakia were evaluated by immunohistochemical staining for the expression of p62, 8-hydroxy-2'-deoxyguanosine (8-OHdG), Ki67, and p53. Univariate and multivariate logistic regression analyses were performed to evaluate the association between p62, 8-OHdG, Ki67, and p53 and clinical characteristics, including epithelial dysplasia. Results Significant associations were observed between p62 expression in the nucleus, p62 aggregation, and epithelial dysplasia (adjusted odds ratio [OR] = 5.75; 95% confidence interval [CI]: [1.28, 26.2]; .024 and OR = 6.16; 95% CI: [1.01, 37.4]; .048, respectively). The expression of p62 in the cytoplasm and the levels of 8-OHdG, Ki67, and p53 were not significantly associated with epithelial dysplasia. A significant relationship was found between p62 expression in the nucleus and p53 expression (OR = 3.94; 95% CI: [1.14, 13.6]; .031). Conclusions The results suggested that p62 expression in the nucleus and p62 aggregation can be potential markers to predict the malignant transformation of oral leukoplakia.
Collapse
Affiliation(s)
- Toshio Yoshida
- Oral and Maxillofacial Surgery, Clinical Sciences, Graduate School of Comprehensive Human ScienceUniversity of TsukubaTsukubaJapan
- Yoshida Dental OfficeMedical Cooperation TokuekaiIshiokaJapan
| | - Takehito Terabe
- Department of Oral and Maxillofacial Surgery, Association for Development of Community MedicineIshioka Daiichi HospitalIshiokaJapan
| | - Hiroki Nagai
- Department of Oral and Maxillofacial Surgery, Faculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Fumihiko Uchida
- Department of Oral and Maxillofacial Surgery, Faculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Shogo Hasegawa
- Department of Maxillofacial Surgery, School of DentistryAichi‐Gakuen UniversityNagoyaJapan
- Department of Oral and Maxillofacial SurgeryToyota Wakatake HospitalToyotaJapan
| | - Toru Nagao
- Department of Maxillofacial Surgery, School of DentistryAichi‐Gakuen UniversityNagoyaJapan
| | - Satoru Miyabe
- Department of Maxillofacial Surgery, School of DentistryAichi‐Gakuen UniversityNagoyaJapan
| | - Naomi Ishibashi‐Kanno
- Department of Oral and Maxillofacial Surgery, Faculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Kenji Yamagata
- Department of Oral and Maxillofacial Surgery, Faculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Eiji Warabi
- Department of Anatomy and Embryology, Faculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Masahiko Gosho
- Department of Biostatistics, Faculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Toru Yanagawa
- Department of Oral and Maxillofacial Surgery, Faculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Hiroki Bukawa
- Department of Oral and Maxillofacial Surgery, Faculty of MedicineUniversity of TsukubaTsukubaJapan
| |
Collapse
|
59
|
Hou B, Wang G, Gao Q, Wei Y, Zhang C, Wang Y, Huo Y, Yang H, Jiang X, Xi Z. SQSTM1/p62 loss reverses the inhibitory effect of sunitinib on autophagy independent of AMPK signaling. Sci Rep 2019; 9:11087. [PMID: 31366950 PMCID: PMC6668422 DOI: 10.1038/s41598-019-47597-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 07/19/2019] [Indexed: 02/03/2023] Open
Abstract
Sunitinib (ST), a multitargeted receptor tyrosine kinase inhibitor, has been demonstrated to be effective for the treatment of renal carcinoma. It has been reported that ST is involved in the mediation of autophagy; however, its regulatory role in the autophagic process remains controversial. Furthermore, the mechanism by which activated AMP-activated protein kinase (AMPK) negatively regulates autophagy remains nearly unexplored. In the present study, we revealed that ST inhibited AMPK activity and regulated autophagy in a cell type- and dose-dependent manner. In a number of cell lines, ST was demonstrated to inhibit H2O2-induced autophagy and the phosphorylation of acetyl-CoA carboxylase (ACC), whereas alone it could block the autophagic flux concurrent with increased expression of p62. An immunoprecipitation assay revealed that LC3 directly interacted with p62, whereas ST increased punctate LC3 staining, which was well colocalized with p62. Taken together, we reveal a previously unnoticed pathway for ST to regulate the autophagic process, and p62, although often utilized as a substrate in autophagy, plays a critical role in regulating the inhibition of ST in both basal and induced autophagy.
Collapse
Affiliation(s)
- Bolin Hou
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Gang Wang
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
| | - Quan Gao
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Yanjie Wei
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Caining Zhang
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Yange Wang
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Yuqing Huo
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, 30912, Georgia, USA
| | - Huaiyi Yang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xuejun Jiang
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Zhijun Xi
- Department of Urology, Peking University First Hospital, Beijing, 100034, China.
| |
Collapse
|
60
|
Li J, Zhao R, Zhao H, Chen G, Jiang Y, Lyu X, Wu T. Reduction of Aging-Induced Oxidative Stress and Activation of Autophagy by Bilberry Anthocyanin Supplementation via the AMPK-mTOR Signaling Pathway in Aged Female Rats. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:7832-7843. [PMID: 31242723 DOI: 10.1021/acs.jafc.9b02567] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Oxidative-stress-induced senescence constitutes a great risk factor for chronic diseases. Therefore, ameliorating oxidative-stress-induced senescence is expected to prevent chronic diseases. The beneficial effects of bilberry anthocyanin (BA) on healthy aging were evaluated using 12 month old, aging female SD rats in this study. The experimental results suggested that consumption of a middle-dose of BA (MBA) appreciably increased the relative liver mass by 7.34% when compared with that of the AC group. Furthermore, BA significantly increased the total antioxidant capacity, total superoxide dismutase activity, and catalase activities; decreased malondialdehyde, serum low-density lipoprotein cholesterol (LDL-C), serum total cholesterol (TC), serum triglyceride (TG), and glycated serum protein (GSP) levels; and reduced TC/high-density lipoprotein cholesterol (HDL-C) and LDL-C/HDL-C ratios. In addition, MBA decreased the activity of fecal bacterial enzymes and increased the content of fecal short-chain fatty acids. The Western blot results showed that MBA significantly upregulated the expression of OCLN, ZO-1, and autophagy-related proteins (ATP6 V0C, ATG4D, and CTSB) in aging rats. Moreover, it also showed that MBA induced the phosphorylation of AMPK and FOXO3a and inhibited the phosphorylation of mTOR, which indicated that bilberry anthocyanin induced autophagy via the AMPK-mTOR signaling pathways. This induction of autophagy further promoted oxidative stress resistance effects and intestinal epithelial barrier function of bilberry anthocyanin in aging female rats.
Collapse
Affiliation(s)
- Jing Li
- State Key Laboratory of Food Nutrition and Safety , Tianjin University of Science and Technology , Tianjin 300457 , China
| | - Runtian Zhao
- State Key Laboratory of Food Nutrition and Safety , Tianjin University of Science and Technology , Tianjin 300457 , China
| | - Huan Zhao
- State Key Laboratory of Food Nutrition and Safety , Tianjin University of Science and Technology , Tianjin 300457 , China
| | - Guiyun Chen
- State Key Laboratory of Food Nutrition and Safety , Tianjin University of Science and Technology , Tianjin 300457 , China
| | - Yuhan Jiang
- State Key Laboratory of Food Nutrition and Safety , Tianjin University of Science and Technology , Tianjin 300457 , China
| | - Xiaoling Lyu
- State Key Laboratory of Food Nutrition and Safety , Tianjin University of Science and Technology , Tianjin 300457 , China
| | - Tao Wu
- State Key Laboratory of Food Nutrition and Safety , Tianjin University of Science and Technology , Tianjin 300457 , China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health , Beijing Technology & Business University , Beijing 100193 , China
| |
Collapse
|
61
|
Liao W, Wang Z, Fu Z, Ma H, Jiang M, Xu A, Zhang W. p62/SQSTM1 protects against cisplatin-induced oxidative stress in kidneys by mediating the cross talk between autophagy and the Keap1-Nrf2 signalling pathway. Free Radic Res 2019; 53:800-814. [PMID: 31223046 DOI: 10.1080/10715762.2019.1635251] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Weitang Liao
- Department of Nephrology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
- Department of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, PR China
| | - Zhiyu Wang
- Department of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, PR China
| | - Zongjie Fu
- Department of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, PR China
| | - Hongkun Ma
- Department of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, PR China
| | - Mengdi Jiang
- Department of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, PR China
| | - Anping Xu
- Department of Nephrology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Wen Zhang
- Department of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, PR China
| |
Collapse
|
62
|
Chen X, Miller NM, Afghah Z, Geiger JD. Development of AD-Like Pathology in Skeletal Muscle. JOURNAL OF PARKINSON'S DISEASE AND ALZHEIMER'S DISEASE 2019; 6:10.13188/2376-922x.1000028. [PMID: 32190732 PMCID: PMC7079679 DOI: 10.13188/2376-922x.1000028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Effective therapeutic strategy against Alzheimer's disease (AD) requires early detection of AD; however, clinical diagnosis of Alzheimer's disease (AD) is not precise and a definitive diagnosis of AD is only possible via postmortem examination for AD pathological hallmarks including senile plaques composed of Aβ and neuro fibrillary tangles composed of phosphorylated tau. Although a variety of biomarker has been developed and used in clinical setting, none of them robustly predicts subsequent clinical course of AD. Thus, it is essential to identify new biomarkers that may facilitate the diagnosis of early stages of AD, prediction of subsequent clinical course, and development of new therapeutic strategies. Given that pathological hallmarks of AD including Aβaccumulation and the presence of phosphorylated tau are also detected in peripheral tissues, AD is considered a systemic disease. Without the protection of blood-brain barrier, systemic factors can affect peripheral tissues much earlier than neurons in brain. Here, we will discuss the development of AD-like pathology in skeletal muscle and the potential use of skeletal muscle biopsy (examination for Aβaccumulation and phosphorylated tau) as a biomarker for AD.
Collapse
Affiliation(s)
- X Chen
- Department of Biomedical Sciences, University of North Dakota, USA
| | - NM Miller
- Department of Biomedical Sciences, University of North Dakota, USA
| | - Z Afghah
- Department of Biomedical Sciences, University of North Dakota, USA
| | - JD Geiger
- Department of Biomedical Sciences, University of North Dakota, USA
| |
Collapse
|
63
|
Wang L, Howell MEA, Sparks-Wallace A, Hawkins C, Nicksic CA, Kohne C, Hall KH, Moorman JP, Yao ZQ, Ning S. p62-mediated Selective autophagy endows virus-transformed cells with insusceptibility to DNA damage under oxidative stress. PLoS Pathog 2019; 15:e1007541. [PMID: 31017975 PMCID: PMC6502431 DOI: 10.1371/journal.ppat.1007541] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 05/06/2019] [Accepted: 03/26/2019] [Indexed: 12/26/2022] Open
Abstract
DNA damage response (DDR) and selective autophagy both can be activated by reactive oxygen/nitrogen species (ROS/RNS), and both are of paramount importance in cancer development. The selective autophagy receptor and ubiquitin (Ub) sensor p62 plays a key role in their crosstalk. ROS production has been well documented in latent infection of oncogenic viruses including Epstein-Barr Virus (EBV). However, p62-mediated selective autophagy and its interplay with DDR have not been investigated in these settings. In this study, we provide evidence that considerable levels of p62-mediated selective autophagy are spontaneously induced, and correlate with ROS-Keap1-NRF2 pathway activity, in virus-transformed cells. Inhibition of autophagy results in p62 accumulation in the nucleus, and promotes ROS-induced DNA damage and cell death, as well as downregulates the DNA repair proteins CHK1 and RAD51. In contrast, MG132-mediated proteasome inhibition, which induces rigorous autophagy, promotes p62 degradation but accumulation of the DNA repair proteins CHK1 and RAD51. However, pretreatment with an autophagy inhibitor offsets the effects of MG132 on CHK1 and RAD51 levels. These findings imply that p62 accumulation in the nucleus in response to autophagy inhibition promotes proteasome-mediated CHK1 and RAD51 protein instability. This claim is further supported by the findings that transient expression of a p62 mutant, which is constitutively localized in the nucleus, in B cell lines with low endogenous p62 levels recaptures the effects of autophagy inhibition on CHK1 and RAD51 protein stability. These results indicate that proteasomal degradation of RAD51 and CHK1 is dependent on p62 accumulation in the nucleus. However, small hairpin RNA (shRNA)-mediated p62 depletion in EBV-transformed lymphoblastic cell lines (LCLs) had no apparent effects on the protein levels of CHK1 and RAD51, likely due to the constitutive localization of p62 in the cytoplasm and incomplete knockdown is insufficient to manifest its nuclear effects on these proteins. Rather, shRNA-mediated p62 depletion in EBV-transformed LCLs results in significant increases of endogenous RNF168-γH2AX damage foci and chromatin ubiquitination, indicative of activation of RNF168-mediated DNA repair mechanisms. Our results have unveiled a pivotal role for p62-mediated selective autophagy that governs DDR in the setting of oncogenic virus latent infection, and provide a novel insight into virus-mediated oncogenesis.
Collapse
Affiliation(s)
- Ling Wang
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States of America
- Center of Excellence for Inflammation, Infectious Diseases and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States of America
| | - Mary E. A. Howell
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States of America
| | - Ayrianna Sparks-Wallace
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States of America
| | - Caroline Hawkins
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States of America
| | - Camri A. Nicksic
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States of America
| | - Carissa Kohne
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States of America
| | - Kenton H. Hall
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States of America
| | - Jonathan P. Moorman
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States of America
- Center of Excellence for Inflammation, Infectious Diseases and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States of America
- The HCV/HIV Program, James H Quillen VA Medical Center, Johnson City, TN, United States of America
| | - Zhi Q. Yao
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States of America
- Center of Excellence for Inflammation, Infectious Diseases and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States of America
- The HCV/HIV Program, James H Quillen VA Medical Center, Johnson City, TN, United States of America
| | - Shunbin Ning
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States of America
- Center of Excellence for Inflammation, Infectious Diseases and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States of America
| |
Collapse
|
64
|
Germic N, Frangez Z, Yousefi S, Simon HU. Regulation of the innate immune system by autophagy: neutrophils, eosinophils, mast cells, NK cells. Cell Death Differ 2019; 26:703-714. [PMID: 30737478 PMCID: PMC6460399 DOI: 10.1038/s41418-019-0295-8] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 01/16/2019] [Accepted: 01/17/2019] [Indexed: 12/15/2022] Open
Abstract
Autophagy is an evolutionally conserved, highly regulated catabolic process that combines cellular functions required for the regulation of metabolic balance under conditions of stress with those needed for the degradation of damaged cell organelles via the lysosomal machinery. The importance of autophagy for cell homeostasis and survival has long been appreciated. Recent data suggest that autophagy is also involved in non-metabolic functions that impact the immune system. Here, we reflect in two review articles the recent literature pointing to an important role for autophagy in innate immune cells. In this article, we focus on neutrophils, eosinophils, mast cells, and natural killer cells. We mainly discuss the influence of autophagy on functional cellular responses and its importance for overall host defense. In the companion review, we present the role of autophagy in the functions performed by monocytes/macrophages and dendritic cells.
Collapse
Affiliation(s)
- Nina Germic
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Ziva Frangez
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Shida Yousefi
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland.
- Department of Clinical Immunology and Allergology, Sechenov University, Moscow, Russia.
| |
Collapse
|
65
|
Salvestrini V, Sell C, Lorenzini A. Obesity May Accelerate the Aging Process. Front Endocrinol (Lausanne) 2019; 10:266. [PMID: 31130916 PMCID: PMC6509231 DOI: 10.3389/fendo.2019.00266] [Citation(s) in RCA: 146] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 04/10/2019] [Indexed: 12/27/2022] Open
Abstract
Lines of evidence from several studies have shown that increases in life expectancy are now accompanied by increased disability rate. The expanded lifespan of the aging population imposes a challenge on the continuous increase of chronic disease. The prevalence of overweight and obesity is increasing at an alarming rate in many parts of the world. Further to increasing the onset of metabolic imbalances, obesity leads to reduced life span and affects cellular and molecular processes in a fashion resembling aging. Nine key hallmarks of the aging process have been proposed. In this review, we will review these hallmarks and discuss pathophysiological changes that occur with obesity, that are similar to or contribute to those that occur during aging. We present and discuss the idea that obesity, in addition to having disease-specific effects, may accelerate the rate of aging affecting all aspects of physiology and thus shortening life span and health span.
Collapse
Affiliation(s)
- Valentina Salvestrini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Christian Sell
- Department of Pathology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Antonello Lorenzini
- Department of Biomedical and Neuromotor Sciences, Biochemistry Unit, University of Bologna, Bologna, Italy
- *Correspondence: Antonello Lorenzini
| |
Collapse
|
66
|
Nrf2: Molecular and epigenetic regulation during aging. Ageing Res Rev 2018; 47:31-40. [PMID: 29913211 DOI: 10.1016/j.arr.2018.06.003] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 06/11/2018] [Accepted: 06/12/2018] [Indexed: 12/23/2022]
Abstract
Increase in life-span is commonly related with age-related diseases and with gradual loss of genomic, proteomic and metabolic integrity. Nrf2 (Nuclear factor-erythroid 2-p45 derived factor 2) controls the expression of genes whose products include antioxidant proteins, detoxifying enzymes, drug transporters and numerous cytoprotective proteins. Several experimental approaches have evaluated the potential regulation of the transcription factor Nrf2 to enhance the expression of genes that contend against accumulative oxidative stress and promote healthy aging. Negative regulators of Nrf2 that act preventing it´s binding to DNA-responsive elements, have been identified in young and adult animal models. However, it is not clearly established if Nrf2 decreased activity in several models of aging results from disruption of that regulation. In this review, we present a compilation of evidences showing that changes in the levels or activity of Keap1 (Kelch-like ECH associated protein 1), GSK-3β (glycogen synthase kinase-3), Bach1, p53, Hrd1 (E3 ubiquitin ligase) and miRNAs might impact on Nrf2 activity during elderly. We conclude that understanding Nrf2 regulatory mechanisms is essential to develop a rational strategy to prevent the loss of cellular protection response during aging.
Collapse
|
67
|
Yamada T, Murata D, Adachi Y, Itoh K, Kameoka S, Igarashi A, Kato T, Araki Y, Huganir RL, Dawson TM, Yanagawa T, Okamoto K, Iijima M, Sesaki H. Mitochondrial Stasis Reveals p62-Mediated Ubiquitination in Parkin-Independent Mitophagy and Mitigates Nonalcoholic Fatty Liver Disease. Cell Metab 2018; 28:588-604.e5. [PMID: 30017357 PMCID: PMC6170673 DOI: 10.1016/j.cmet.2018.06.014] [Citation(s) in RCA: 199] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 03/07/2018] [Accepted: 06/15/2018] [Indexed: 12/15/2022]
Abstract
It is unknown what occurs if both mitochondrial division and fusion are completely blocked. Here, we introduced mitochondrial stasis by deleting two dynamin-related GTPases for division (Drp1) and fusion (Opa1) in livers. Mitochondrial stasis rescues liver damage and hypotrophy caused by the single knockout (KO). At the cellular level, mitochondrial stasis re-establishes mitochondrial size and rescues mitophagy defects caused by division deficiency. Using Drp1KO livers, we found that the autophagy adaptor protein p62/sequestosome-1-which is thought to function downstream of ubiquitination-promotes mitochondrial ubiquitination. p62 recruits two subunits of a cullin-RING ubiquitin E3 ligase complex, Keap1 and Rbx1, to mitochondria. Resembling Drp1KO, diet-induced nonalcoholic fatty livers enlarge mitochondria and accumulate mitophagy intermediates. Resembling Drp1Opa1KO, Opa1KO rescues liver damage in this disease model. Our data provide a new concept that mitochondrial stasis leads the spatial dimension of mitochondria to a stationary equilibrium and a new mechanism for mitochondrial ubiquitination in mitophagy.
Collapse
Affiliation(s)
- Tatsuya Yamada
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Daisuke Murata
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yoshihiro Adachi
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kie Itoh
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Shoichiro Kameoka
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Graduate School of Frontier Biosciences, Osaka University, Osaka 565-0871, Japan
| | - Atsushi Igarashi
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Takashi Kato
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yoichi Araki
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Richard L Huganir
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ted M Dawson
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130, USA
| | - Toru Yanagawa
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Koji Okamoto
- Graduate School of Frontier Biosciences, Osaka University, Osaka 565-0871, Japan
| | - Miho Iijima
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Hiromi Sesaki
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
68
|
Wu Y, Li Y, Wang B, He X, Li Y, Wu B, Yu G, Wang H, Xu B. Role of p62/SQSTM1 in lipopolysaccharide (LPS)-induced mucus hypersecretion in bronchial epithelial cells. Life Sci 2018; 211:270-278. [DOI: 10.1016/j.lfs.2018.09.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 09/12/2018] [Accepted: 09/18/2018] [Indexed: 12/21/2022]
|
69
|
Lee HS, Park YJ, Cho DW, Han SC, Jun SY, Jung GM, Lee WJ, Choi CM, Park EJ, Pak SI. Repeated injection of KMRC011, a medical countermeasure for radiation, can cause adverse health effects in cynomolgus monkeys. J Appl Toxicol 2018; 39:294-304. [DOI: 10.1002/jat.3719] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 07/27/2018] [Accepted: 07/28/2018] [Indexed: 01/08/2023]
Affiliation(s)
- Hong-Soo Lee
- Jeonbuk Department of Inhalation Research; Korea Institute of Toxicology; Jeongeup-si Jeollabuk-do 56212 Republic of Korea
| | - Yoo-Jin Park
- Graduate School of East-West Medical Science; Kyung Hee University; Yongin-si Gyeonggi-do 17104 Republic of Korea
| | - Doo-Wan Cho
- Jeonbuk Department of Inhalation Research; Korea Institute of Toxicology; Jeongeup-si Jeollabuk-do 56212 Republic of Korea
| | - Su-Cheol Han
- Jeonbuk Department of Inhalation Research; Korea Institute of Toxicology; Jeongeup-si Jeollabuk-do 56212 Republic of Korea
| | - Soo Youn Jun
- iNtRON Biotechnology Inc.; Seongnam-si Gyeonggi-do 13202 Republic of Korea
| | - Gi Mo Jung
- iNtRON Biotechnology Inc.; Seongnam-si Gyeonggi-do 13202 Republic of Korea
| | - Woo-Jong Lee
- Biomedical Manufacturing Technology Center; Korea Institute of Industrial Technology; Yeongcheon-si Gyeongsangbuk-do 38822 Republic of Korea
| | - Chi-Min Choi
- Biomedical Manufacturing Technology Center; Korea Institute of Industrial Technology; Yeongcheon-si Gyeongsangbuk-do 38822 Republic of Korea
| | - Eun-Jung Park
- Graduate School of East-West Medical Science; Kyung Hee University; Yongin-si Gyeonggi-do 17104 Republic of Korea
| | - Son-Il Pak
- College of Veterinary Medicine and Institute of Veterinary Science; Kangwon National University; Chuncheon-si Gangwon-do 24341 Republic of Korea
| |
Collapse
|
70
|
Huang X, Wu H, Jiang R, Sun G, Shen J, Ma M, Ma C, Zhang S, Huang Z, Wu Q, Chen G, Tao W. The antidepressant effects of ɑ-tocopherol are related to activation of autophagy via the AMPK/mTOR pathway. Eur J Pharmacol 2018; 833:1-7. [DOI: 10.1016/j.ejphar.2018.05.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 04/28/2018] [Accepted: 05/17/2018] [Indexed: 12/25/2022]
|
71
|
Bejarano E, Murray JW, Wang X, Pampliega O, Yin D, Patel B, Yuste A, Wolkoff AW, Cuervo AM. Defective recruitment of motor proteins to autophagic compartments contributes to autophagic failure in aging. Aging Cell 2018; 17:e12777. [PMID: 29845728 PMCID: PMC6052466 DOI: 10.1111/acel.12777] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2018] [Indexed: 12/11/2022] Open
Abstract
Inability to preserve proteostasis with age contributes to the gradual loss of function that characterizes old organisms. Defective autophagy, a component of the proteostasis network for delivery and degradation of intracellular materials in lysosomes, has been described in multiple old organisms, while a robust autophagy response has been linked to longevity. The molecular mechanisms responsible for defective autophagic function with age remain, for the most part, poorly characterized. In this work, we have identified differences between young and old cells in the intracellular trafficking of the vesicular compartments that participate in autophagy. Failure to reposition autophagosomes and lysosomes toward the perinuclear region with age reduces the efficiency of their fusion and the subsequent degradation of the sequestered cargo. Hepatocytes from old mice display lower association of two microtubule-based minus-end-directed motor proteins, the well-characterized dynein, and the less-studied KIFC3, with autophagosomes and lysosomes, respectively. Using genetic approaches to mimic the lower levels of KIFC3 observed in old cells, we confirmed that reduced content of this motor protein in fibroblasts leads to failed lysosomal repositioning and diminished autophagic flux. Our study connects defects in intracellular trafficking with insufficient autophagy in old organisms and identifies motor proteins as a novel target for future interventions aiming at correcting autophagic activity with anti-aging purposes.
Collapse
Affiliation(s)
- Eloy Bejarano
- Department of Developmental and Molecular Biology; Albert Einstein College of Medicine; Bronx New York
- Institute for Aging Studies; Albert Einstein College of Medicine; Bronx New York
| | - John W. Murray
- Marion Bessin Liver Research Center; Albert Einstein College of Medicine; Bronx New York
- Department of Anatomy and Structural Biology; Albert Einstein College of Medicine; Bronx New York
| | - Xintao Wang
- Marion Bessin Liver Research Center; Albert Einstein College of Medicine; Bronx New York
- Department of Anatomy and Structural Biology; Albert Einstein College of Medicine; Bronx New York
| | - Olatz Pampliega
- Institut des Maladies Neurodégénératives UMR5293; Universite de Bordeaux; Bordeaux France
- CNRS; Institut des Maladies Neurodégénératives; UMR 5293 C Bordeaux Cedex France
| | - David Yin
- Marion Bessin Liver Research Center; Albert Einstein College of Medicine; Bronx New York
| | - Bindi Patel
- Department of Developmental and Molecular Biology; Albert Einstein College of Medicine; Bronx New York
- Institute for Aging Studies; Albert Einstein College of Medicine; Bronx New York
| | - Andrea Yuste
- Department of Developmental and Molecular Biology; Albert Einstein College of Medicine; Bronx New York
- Institute for Aging Studies; Albert Einstein College of Medicine; Bronx New York
| | - Allan W. Wolkoff
- Marion Bessin Liver Research Center; Albert Einstein College of Medicine; Bronx New York
- Department of Anatomy and Structural Biology; Albert Einstein College of Medicine; Bronx New York
- Division of Hepatology, Albert Einstein College of Medicine and; Montefiore Medical Center; Bronx New York
| | - Ana Maria Cuervo
- Department of Developmental and Molecular Biology; Albert Einstein College of Medicine; Bronx New York
- Institute for Aging Studies; Albert Einstein College of Medicine; Bronx New York
- Marion Bessin Liver Research Center; Albert Einstein College of Medicine; Bronx New York
- Department of Anatomy and Structural Biology; Albert Einstein College of Medicine; Bronx New York
| |
Collapse
|
72
|
Cabe M, Rademacher DJ, Karlsson AB, Cherukuri S, Bakowska JC. PB1 and UBA domains of p62 are essential for aggresome-like induced structure formation. Biochem Biophys Res Commun 2018; 503:2306-2311. [PMID: 29966650 DOI: 10.1016/j.bbrc.2018.06.153] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 06/27/2018] [Indexed: 11/26/2022]
Abstract
ALIS are large, transient, cytosolic aggregates that serve as storage compartments for ubiquitin-tagged defective ribosomal products. We determined the importance of the protein p62 in the formation of ALIS and demonstrated that two domains of p62-PB1 and UBA-are essential for ALIS assembly. Those two major binding domains of p62, also known as sequestosome 1, were shown to play a critical role in the formation of autophagosomes or cytoplasmic aggregates. Specifically, the PB1 domain is essential for self-oligomerization, and the UBA domain allows p62 to bind to polyubiquitin chains or ubiquitinated proteins. After stimulation of RAW 264.7 macrophages with lipopolysaccharide, we observed a significant decrease in the number of cells with ALIS. Importantly, cells overexpressing either a PB1 mutant or UBA-deleted p62 construct also exhibited a substantially diminished number of cells containing ALIS. Since both p62 and ubiquitin are found in ALIS, we evaluated the dynamics of YFP-tagged p62 in ALIS. In contrast to the findings of a previous study that evaluated GFP-tagged ubiquitin motility in ALIS, we determined that YFP-tagged p62 has very limited mobility. Lastly, we determined that GST-tagged full-length p62 binds to Lys-63-linked polyubiquitin chains but not to Lys-48-linked chains. Overall, our findings provide insight on the essential role that p62, particularly its PB1 and UBA domains, has in the formation of ALIS.
Collapse
Affiliation(s)
- Maleen Cabe
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Maywood, IL 60153, USA
| | - David J Rademacher
- Core Imaging Facility and Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL 60153, USA
| | - Amelia B Karlsson
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Maywood, IL 60153, USA
| | - Srinivas Cherukuri
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Maywood, IL 60153, USA
| | - Joanna C Bakowska
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Maywood, IL 60153, USA.
| |
Collapse
|
73
|
Xu W, Wei Q, Han M, Zhou B, Wang H, Zhang J, Wang Q, Sun J, Feng L, Wang S, Ye Y, Wang X, Zhou J, Jin H. CCL2-SQSTM1 positive feedback loop suppresses autophagy to promote chemoresistance in gastric cancer. Int J Biol Sci 2018; 14:1054-1066. [PMID: 29989092 PMCID: PMC6036739 DOI: 10.7150/ijbs.25349] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Accepted: 05/10/2018] [Indexed: 12/11/2022] Open
Abstract
Chemotherapy is one of the most important approaches for the treatment of various cancers. However, tumor cells often develop resistance to chemotherapeutic drugs. The tumor microenvironment reconstituted by various cytokines secreted from immune cells was recently found to play important roles in affecting therapeutic response of tumor cells. Herein, we reported that tumor cells can secrete autocrine cytokines to confer chemoresistance by inactivating proapoptotic autophagy. Through cytokine screening, we found that drug resistant cancer cells secreted more CCL2 than drug sensitive cells. Such secreted CCL2 could not only maintain chemoresistance in drug-resistant cancer cells but also confer drug resistance to drug-sensitive cancer cells. CCL2 attenuated drug-induced cytotoxicity by activating PI3K-Akt-mTOR signaling to inhibit proapoptotic autophagy and increase SQSTM1 expression. CCL2 expression in primary carcinoma tissues also correlated well with SQSTM1 expression. Either CCL2 knock-down or autophagy induction successfully reversed drug resistance of tumor cells. Moreover, increased expression of SQSTM1 in turn activated CCL2 transcription via NF-κB signal pathway, representing a positive feedback loop to maintain drug resistance. Therefore, our results provided a new insight to understand drug resistance, and indicated the potential value of CCL2 as a biomarker and intervention target for chemotherapy resistance.
Collapse
Affiliation(s)
- Wenxia Xu
- Labortaory of Cancer Biology, Key Laboratory of Biotherapy in Zhejiang, Sir Runrun Shaw hospital, Medical School of Zhejiang University, China
| | - Qi Wei
- Labortaory of Cancer Biology, Key Laboratory of Biotherapy in Zhejiang, Sir Runrun Shaw hospital, Medical School of Zhejiang University, China
| | - Mengjiao Han
- Department of Medical Oncology, Sir Runrun Shaw hospital, Medical School of Zhejiang University, China
| | - Bingluo Zhou
- Department of Medical Oncology, Sir Runrun Shaw hospital, Medical School of Zhejiang University, China
| | - Hanying Wang
- Department of Medical Oncology, Sir Runrun Shaw hospital, Medical School of Zhejiang University, China
| | - Jianbing Zhang
- Pathology Center, Shanghai General Hospital, Medical School of Shanghai Jiaotong University
| | - Qiang Wang
- Department of Molecular Cell Biology and Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jie Sun
- Labortaory of Cancer Biology, Key Laboratory of Biotherapy in Zhejiang, Sir Runrun Shaw hospital, Medical School of Zhejiang University, China
| | - Lifeng Feng
- Labortaory of Cancer Biology, Key Laboratory of Biotherapy in Zhejiang, Sir Runrun Shaw hospital, Medical School of Zhejiang University, China
| | - Shouyu Wang
- Department of Molecular Cell Biology and Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yang Ye
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xian Wang
- Department of Medical Oncology, Sir Runrun Shaw hospital, Medical School of Zhejiang University, China
| | - Jianwei Zhou
- Department of Molecular Cell Biology and Toxicology, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Hongchuan Jin
- Labortaory of Cancer Biology, Key Laboratory of Biotherapy in Zhejiang, Sir Runrun Shaw hospital, Medical School of Zhejiang University, China
| |
Collapse
|
74
|
Thomas-Jardin SE, Kanchwala MS, Jacob J, Merchant S, Meade RK, Gahnim NM, Nawas AF, Xing C, Delk NA. Identification of an IL-1-induced gene expression pattern in AR + PCa cells that mimics the molecular phenotype of AR - PCa cells. Prostate 2018; 78. [PMID: 29527701 PMCID: PMC5893432 DOI: 10.1002/pros.23504] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND In immunosurveillance, bone-derived immune cells infiltrate the tumor and secrete inflammatory cytokines to destroy cancer cells. However, cancer cells have evolved mechanisms to usurp inflammatory cytokines to promote tumor progression. In particular, the inflammatory cytokine, interleukin-1 (IL-1), is elevated in prostate cancer (PCa) patient tissue and serum, and promotes PCa bone metastasis. IL-1 also represses androgen receptor (AR) accumulation and activity in PCa cells, yet the cells remain viable and tumorigenic; suggesting that IL-1 may also contribute to AR-targeted therapy resistance. Furthermore, IL-1 and AR protein levels negatively correlate in PCa tumor cells. Taken together, we hypothesize that IL-1 reprograms AR positive (AR+ ) PCa cells into AR negative (AR- ) PCa cells that co-opt IL-1 signaling to ensure AR-independent survival and tumor progression in the inflammatory tumor microenvironment. METHODS LNCaP and PC3 PCa cells were treated with IL-1β or HS-5 bone marrow stromal cell (BMSC) conditioned medium and analyzed by RNA sequencing and RT-QPCR. To verify genes identified by RNA sequencing, LNCaP, MDA-PCa-2b, PC3, and DU145 PCa cell lines were treated with the IL-1 family members, IL-1α or IL-1β, or exposed to HS-5 BMSC in the presence or absence of Interleukin-1 Receptor Antagonist (IL-1RA). Treated cells were analyzed by western blot and/or RT-QPCR. RESULTS Comparative analysis of sequencing data from the AR+ LNCaP PCa cell line versus the AR- PC3 PCa cell line reveals an IL-1-conferred gene suite in LNCaP cells that is constitutive in PC3 cells. Bioinformatics analysis of the IL-1 regulated gene suite revealed that inflammatory and immune response pathways are primarily elicited; likely facilitating PCa cell survival and tumorigenicity in an inflammatory tumor microenvironment. CONCLUSIONS Our data supports that IL-1 reprograms AR+ PCa cells to mimic AR- PCa gene expression patterns that favor AR-targeted treatment resistance and cell survival.
Collapse
Affiliation(s)
| | - Mohammed S. Kanchwala
- McDermott Center of Human Growth and Development, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Joan Jacob
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080
| | - Sana Merchant
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080
| | - Rachel K. Meade
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080
| | - Nagham M. Gahnim
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080
| | - Afshan F. Nawas
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080
| | - Chao Xing
- McDermott Center of Human Growth and Development, The University of Texas Southwestern Medical Center, Dallas, TX 75390
- Department of Bioinformatics, The University of Texas Southwestern Medical Center, Dallas, TX 75390
- Department of Clinical Sciences, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Nikki A. Delk
- Biological Sciences Department, The University of Texas at Dallas, Richardson, TX 75080
| |
Collapse
|
75
|
Candeias E, Sebastião I, Cardoso S, Carvalho C, Santos MS, Oliveira CR, Moreira PI, Duarte AI. Brain GLP-1/IGF-1 Signaling and Autophagy Mediate Exendin-4 Protection Against Apoptosis in Type 2 Diabetic Rats. Mol Neurobiol 2018; 55:4030-4050. [PMID: 28573460 DOI: 10.1007/s12035-017-0622-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 05/16/2017] [Indexed: 02/08/2023]
Abstract
Type 2 diabetes (T2D) is a modern socioeconomic burden, mostly due to its long-term complications affecting nearly all tissues. One of them is the brain, whose dysfunctional intracellular quality control mechanisms (namely autophagy) may upregulate apoptosis, leading to cognitive dysfunction and Alzheimer disease (AD). Since impaired brain insulin signaling may constitute the crosslink between T2D and AD, its restoration may be potentially therapeutic herein. Accordingly, the insulinotropic anti-T2D drugs from glucagon-like peptide-1 (GLP-1) mimetics, namely, exendin-4 (Ex-4), could be a promising therapy. In line with this, we hypothesized that peripherally administered Ex-4 rescues brain intracellular signaling pathways, promoting autophagy and ultimately protecting against chronic T2D-induced apoptosis. Thus, we aimed to explore the effects of chronic, continuous, subcutaneous (s.c.) exposure to Ex-4 in brain cortical GLP-1/insulin/insulin-like growth factor-1 (IGF-1) signaling, and in autophagic and cell death mechanisms in middle-aged (8 months old), male T2D Goto-Kakizaki (GK) rats. We used brain cortical homogenates obtained from middle-aged (8 months old) male Wistar (control) and T2D GK rats. Ex-4 was continuously administered for 28 days, via s.c. implanted micro-osmotic pumps (5 μg/kg/day; infusion rate 2.5 μL/h). Peripheral characterization of the animal models was given by the standard biochemical analyses of blood or plasma, the intraperitoneal glucose tolerance test, and the heart rate. GLP-1, insulin, and IGF-1, their downstream signaling and autophagic markers were evaluated by specific ELISA kits and Western blotting. Caspase-like activities and other apoptotic markers were given by colorimetric methods and Western blotting. Chronic Ex-4 treatment attenuated peripheral features of T2D in GK rats, including hyperglycemia and insulin resistance. Furthermore, s.c. Ex-4 enhanced their brain cortical GLP-1 and IGF-1 levels, and subsequent signaling pathways. Specifically, Ex-4 stimulated protein kinase A (PKA) and phosphoinositide 3-kinase (PI3K)/Akt signaling, increasing cGMP and AMPK levels, and decreasing GSK3β and JNK activation in T2D rat brains. Moreover, Ex-4 regulated several markers for autophagy in GK rat brains (as mTOR, PI3K class III, LC3 II, Atg7, p62, LAMP-1, and Parkin), ultimately protecting against apoptosis (by decreasing several caspase-like activities and mitochondrial cytochrome c, and increasing Bcl2 levels upon T2D). Altogether, this study demonstrates that peripheral Ex-4 administration may constitute a promising therapy against the chronic complications of T2D affecting the brain.
Collapse
Affiliation(s)
- Emanuel Candeias
- CNC-Center for Neuroscience and Cell Biology, Rua Larga, Faculty of Medicine (Pólo 1, 1st Floor), University of Coimbra, 3004-517, Coimbra, Portugal
- Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Casa Costa Alemão - Pólo II, Rua D. Francisco de Lemos, 3030-789, Coimbra, Portugal
| | - Inês Sebastião
- CNC-Center for Neuroscience and Cell Biology, Rua Larga, Faculty of Medicine (Pólo 1, 1st Floor), University of Coimbra, 3004-517, Coimbra, Portugal
| | - Susana Cardoso
- CNC-Center for Neuroscience and Cell Biology, Rua Larga, Faculty of Medicine (Pólo 1, 1st Floor), University of Coimbra, 3004-517, Coimbra, Portugal
- Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Casa Costa Alemão - Pólo II, Rua D. Francisco de Lemos, 3030-789, Coimbra, Portugal
| | - Cristina Carvalho
- CNC-Center for Neuroscience and Cell Biology, Rua Larga, Faculty of Medicine (Pólo 1, 1st Floor), University of Coimbra, 3004-517, Coimbra, Portugal
- Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Casa Costa Alemão - Pólo II, Rua D. Francisco de Lemos, 3030-789, Coimbra, Portugal
| | - Maria Sancha Santos
- CNC-Center for Neuroscience and Cell Biology, Rua Larga, Faculty of Medicine (Pólo 1, 1st Floor), University of Coimbra, 3004-517, Coimbra, Portugal
- Life Sciences Department, University of Coimbra, Largo Marquês de Pombal, 3004-517, Coimbra, Portugal
| | - Catarina Resende Oliveira
- CNC-Center for Neuroscience and Cell Biology, Rua Larga, Faculty of Medicine (Pólo 1, 1st Floor), University of Coimbra, 3004-517, Coimbra, Portugal
- Institute of Biochemistry, Faculty of Medicine, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Paula I Moreira
- CNC-Center for Neuroscience and Cell Biology, Rua Larga, Faculty of Medicine (Pólo 1, 1st Floor), University of Coimbra, 3004-517, Coimbra, Portugal.
- Institute of Physiology, Faculty of Medicine, University of Coimbra, 3004-504, Coimbra, Portugal.
| | - Ana I Duarte
- CNC-Center for Neuroscience and Cell Biology, Rua Larga, Faculty of Medicine (Pólo 1, 1st Floor), University of Coimbra, 3004-517, Coimbra, Portugal.
- Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Casa Costa Alemão - Pólo II, Rua D. Francisco de Lemos, 3030-789, Coimbra, Portugal.
| |
Collapse
|
76
|
Lu Y, Wang Q, Zhou Y, Sun L, Hu B, Xue H, Li M, Zhang K, Ren C, Duan N, Liu H, Zhang C, Li Z, Ma T. Overexpression of p62 is associated with poor prognosis and aggressive phenotypes in osteosarcoma. Oncol Lett 2018; 15:9889-9895. [PMID: 29928361 PMCID: PMC6004647 DOI: 10.3892/ol.2018.8579] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Accepted: 03/16/2018] [Indexed: 12/31/2022] Open
Abstract
p62 (also known as sequestosome 1) protein, is a small regulatory protein that accumulates in autophagy-defective cells that has been demonstrated to be involved in the prognosis and survival of patients with several types of cancer. However, to the best of our knowledge, there have been no such studies for osteosarcoma (OS). In the present study, the expression of p62 in 70 OS samples was determined using immunohistochemistry and its association with various clinicopathological factors was assessed. The results demonstrated that the overexpression of p62 protein was detected in 77.1% (54/70) samples, and the expression levels were significantly associated with tumor size (P=0.001), metastasis (P=0.036), clinical staging (P=0.003) and poor prognosis (P=0.0058). Furthermore, suppression of the p62 expression by short hairpin RNA interference in F5M2 and F4 cells lines led to decreased cell proliferation, migration and invasion in vitro. These results suggested that increased expression of p62 may be involved in OS progression, and therefore the excess expression of p62 may serve as a novel prognostic biomarker for patients with OS.
Collapse
Affiliation(s)
- Yao Lu
- Department of Orthopaedic Surgery, Hong Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710054, P.R. China
| | - Qian Wang
- Department of Orthopaedic Surgery, Hong Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710054, P.R. China
| | - Yong Zhou
- Department of Orthopaedic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Liang Sun
- Department of Orthopaedic Surgery, Hong Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710054, P.R. China
| | - Bin Hu
- Department of Hematology, Xi'an Gao Xin Hospital, Xi'an, Shaanxi 710075, P.R. China
| | - Hanzhong Xue
- Department of Orthopaedic Surgery, Hong Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710054, P.R. China
| | - Ming Li
- Department of Orthopaedic Surgery, Hong Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710054, P.R. China
| | - Kun Zhang
- Department of Orthopaedic Surgery, Hong Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710054, P.R. China
| | - Cheng Ren
- Department of Orthopaedic Surgery, Hong Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710054, P.R. China
| | - Ning Duan
- Department of Orthopaedic Surgery, Hong Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710054, P.R. China
| | - Hongliang Liu
- Department of Orthopaedic Surgery, Hong Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710054, P.R. China
| | - Congming Zhang
- Department of Orthopaedic Surgery, Hong Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710054, P.R. China
| | - Zhong Li
- Department of Orthopaedic Surgery, Hong Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710054, P.R. China
| | - Teng Ma
- Department of Orthopaedic Surgery, Hong Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710054, P.R. China
| |
Collapse
|
77
|
Wu Y, Xu B, He X, Wu B, Li Y, Yu G, Tan C, Wang H. Correlation between autophagy levels in peripheral blood mononuclear cells and clinical parameters in patients with chronic obstructive pulmonary disease. Mol Med Rep 2018; 17:8003-8009. [PMID: 29620199 DOI: 10.3892/mmr.2018.8831] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 03/02/2018] [Indexed: 11/05/2022] Open
Abstract
Autophagy serves a role in the pathogenesis of chronic inflammatory diseases. The aim of the present study was to compare the autophagy levels in the peripheral blood mononuclear cells (PBMCs) of patients with chronic obstructive pulmonary disease (COPD) and healthy individuals and to assess the association between autophagy and the clinical parameters of COPD. Samples of peripheral blood from 20 patients with stable COPD and 20 healthy controls were collected. PBMCs were harvested using Ficoll density gradient centrifugation. Levels of the autophagy‑associated proteins ubiquitin‑binding protein p62 (p62), microtubule‑associated proteins 1A/1B light chain 3A (LC3I/II) and beclin‑1 in PBMCs were detected by western blotting. Enzyme‑linked immunosorbent assay kits were used to detect the serum concentrations of interleukin (IL)‑6, IL‑8 and tumor necrosis factor (TNF)‑α. Associations between the levels of autophagy and forced expiratory volume in 1 sec % predicted (FEV1%) and pro‑inflammatory factors were assessed. Western blotting demonstrated that the protein expression of p62 was decreased, but LC3II/I and beclin‑1 levels increased in patients with COPD compared with healthy controls. Serum levels of IL‑6, IL‑8 and TNF‑α were increased in patients with COPD. The extent of PBMC autophagy was negatively correlated with FEV1% predicted, but positively correlated with levels of pro‑inflammatory cytokines. The levels of autophagy in PBMCs in patients with COPD were increased and were negatively correlated with FEV1% predicted and positively correlated with circulating levels of pro‑inflammatory cytokines. Autophagy may serve a role as a biomarker of the severity of COPD or as a therapeutic target for treatment of COPD.
Collapse
Affiliation(s)
- Yanjun Wu
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Bo Xu
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Xin He
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Bo Wu
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Yunxiao Li
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Ganggang Yu
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Chunting Tan
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Haoyan Wang
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| |
Collapse
|
78
|
Li X, Li Y, Zhao J, Li L, Wang Y, Zhang Y, Li Y, Chen Y, Liu W, Gao L. Administration of Ketamine Causes Autophagy and Apoptosis in the Rat Fetal Hippocampus and in PC12 Cells. Front Cell Neurosci 2018; 12:21. [PMID: 29456493 PMCID: PMC5801406 DOI: 10.3389/fncel.2018.00021] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 01/15/2018] [Indexed: 11/18/2022] Open
Abstract
Drug abuse during pregnancy is a serious problem. Like alcohol, anticonvulsants, sedatives, and anesthetics, such as ketamine, can pass through the placental barrier and affect the growing fetus. However, the mechanism by which ketamine causes damage to fetal rats is not well understood. Therefore, in this study, we anesthetized pregnant rats with ketamine and evaluated the Total Antioxidant Capacity (T-AOC), Reactive Oxygen Species (ROS), and Malondialdehyde (MDA). Moreover, we determined changes in the levels of Cleaved-Caspase-3 (C-Caspase-3), Beclin-1, B-cell lymphoma-2 (Bcl-2), Bcl-2 Associated X Protein (Bax), Autophagy-related gene 4 (Atg4), Atg5, p62 (SQSTM1), and marker of autophagy Light Chain 3 (LC3). In addition, we cultured PC12 cells in vitro to determine the relationship between ROS, autophagy, and apoptosis following ketamine treatment. The results showed that ketamine induced changes in autophagy- and apoptosis-related proteins, reduced T-AOC, and generated excessive levels of ROS and MDA. In vitro experiments showed similar results, indicating that apoptosis levels can be inhibited by 3-MA. We also found that autophagy and apoptosis can be inhibited by N-acetyl-L-cysteine (Nac). Thus, anesthesia with ketamine in pregnant rats may increase the rate of autophagy and apoptosis in the fetal hippocampus and the mechanism may be through inhibition of antioxidant activity and ROS accumulation.
Collapse
Affiliation(s)
- Xinran Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, China
| | - Yanan Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, China
| | - Jinghua Zhao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, China
| | - Lina Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, China
| | - Yuxin Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, China
| | - Yiming Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, China
| | - Yue Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, China
| | - Yu Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, China
| | - Wenhan Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, China
| | - Li Gao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Key Laboratory of the Provincial Education, Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, China
| |
Collapse
|
79
|
Honda A, Komiya K, Hara A, Fukunaka A, Suzuki L, Miyatsuka T, Ogihara T, Fujitani Y, Watada H. Normal pancreatic β-cell function in mice with RIP-Cre-mediated inactivation of p62/SQSTM1. Endocr J 2018; 65:83-89. [PMID: 28978813 DOI: 10.1507/endocrj.ej17-0333] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/09/2022] Open
Abstract
Recent studies have suggested that decreased pancreatic β-cell function and mass are common features of patients with type 2 diabetes mellitus. Pancreatic β-cell homeostasis is regulated by various types of signaling molecules and stress responses. Sequestosome 1/p62 (SQSTM1, hereafter referred to as p62) is a ubiquitin-binding adaptor protein involved in cell signaling, oxidative stress, and autophagy. Because p62 appears to play an important role in maintaining mitochondrial quality control, it is possible that the loss of p62 in pancreatic β cells contributes to mitochondrial dysfunction, and thus leading to impaired glucose tolerance. In this study we investigated the physiological roles of p62 by inactivating p62 in a β-cell specific manner. We found that firstly, rat insulin-2 promoter-Cre (RIP-Cre)-mediated p62 inactivation did not cause body weight gain, although ubiquitous inactivation of p62 was previously shown to result in severe obesity. Secondly, we found no gross structural disorganization of the islets of p62-deficient mice. Consistent with normal islet morphology, no impairment in glucose tolerance was observed in mice with RIP-Cre-mediated p62 deletion. These results suggest that p62 is dispensable for normal islet organization and β-cell function.
Collapse
Affiliation(s)
- Akira Honda
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Koji Komiya
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Akemi Hara
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
- Center for Identification of Diabetic Therapeutic Targets, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Ayako Fukunaka
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
- Laboratory of Developmental Biology and Metabolism, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi 371-8512, Japan
| | - Luka Suzuki
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Takeshi Miyatsuka
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
- Center for Identification of Diabetic Therapeutic Targets, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Takeshi Ogihara
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Yoshio Fujitani
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
- Laboratory of Developmental Biology and Metabolism, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi 371-8512, Japan
| | - Hirotaka Watada
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
- Center for Identification of Diabetic Therapeutic Targets, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
- Center for Therapeutic Innovations in Diabetes, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
- Sportology Center, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| |
Collapse
|
80
|
Tang M, Ji C, Pallo S, Rahman I, Johnson GVW. Nrf2 mediates the expression of BAG3 and autophagy cargo adaptor proteins and tau clearance in an age-dependent manner. Neurobiol Aging 2017; 63:128-139. [PMID: 29304346 DOI: 10.1016/j.neurobiolaging.2017.12.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 11/07/2017] [Accepted: 12/01/2017] [Indexed: 11/25/2022]
Abstract
During aging, decreased efficiency of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) activation and autophagic processes in the brain may be a contributing factor in the pathogenesis of neurodegenerative diseases such as Alzheimer's disease. Therefore, we analyzed the expression of Bcl-2-associated athanogene 3, a cochaperone that mediates autophagy, and the autophagy adaptors NBR1, NDP52, and sequestosome 1/p62 in the brains of 4-, 8-, and 12-month-old wild-type and Nrf2 knockout (-/-) mice. We also analyzed the levels of total tau and phospho-tau species. There were minimal differences in the expression of autophagy-related genes or tau species in 4-month-old animals; however, by 12 months, all of these autophagy-associated genes were expressed at significantly lower levels in the Nrf2 (-/-) mice. The decreases in the autophagy-associated genes were accompanied by significantly elevated levels of phospho-tau species in the 12-month-old Nrf2 (-/-) brains. These findings indicate that Nrf2 regulation of autophagy-related genes likely plays a greater role in mediating the clearance of tau as an organism ages.
Collapse
Affiliation(s)
- Maoping Tang
- Department of Anesthesiology, University of Rochester Medical Center, University of Rochester, Rochester, NY, USA
| | - Changyi Ji
- Department of Anesthesiology, University of Rochester Medical Center, University of Rochester, Rochester, NY, USA
| | - Susanne Pallo
- Department of Anesthesiology, University of Rochester Medical Center, University of Rochester, Rochester, NY, USA
| | - Irfan Rahman
- Department of Environmental Medicine, Lung Biology and Disease Program, University of Rochester Medical Center, University of Rochester, Rochester, NY, USA
| | - Gail V W Johnson
- Department of Anesthesiology, University of Rochester Medical Center, University of Rochester, Rochester, NY, USA.
| |
Collapse
|
81
|
Ning H, Liu D, Yu X, Guan X. Oxidized low-density lipoprotein-induced p62/SQSTM1 accumulation in THP-1-derived macrophages promotes IL-18 secretion and cell death. Exp Ther Med 2017; 14:5417-5423. [PMID: 29285070 PMCID: PMC5740607 DOI: 10.3892/etm.2017.5221] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 10/05/2016] [Indexed: 11/29/2022] Open
Abstract
Macrophage autophagy has a protective role in the development of atherosclerosis; however, it turns dysfunctional in advanced lesions with an increase in p62/sequestosome-1 protein. Little is known about the role and significance of p62 accumulation in atherosclerosis. The present study investigated the association between p62 expression and the process of foam cell formation. Foam cell models were established through incubation of THP-1-derived macrophages with oxidized low-density lipoprotein, and the process of foam cell formation was detected by Oil red O staining. Furthermore, the dynamic change of p62 expression was detected by western blotting and quantitative polymerase chain reaction. Additionally, using gene silencing techniques, the roles of p62 in foam cells were investigated with ELISA, MTT and flow cytometry. The results indicated that besides serving as a marker of autophagy deficiency, the p62 protein could also mediate inflammation and cytotoxicity in advanced foam cells. Additionally, the implication of p62 in autophagy inhibition and foam cell formation makes it a key atherogenic factor under autophagy-deficient conditions.
Collapse
Affiliation(s)
- Haofeng Ning
- Department of Laboratory Diagnostics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Dan Liu
- Department of Laboratory Diagnostics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Xiaochen Yu
- Department of Laboratory Diagnostics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Xiuru Guan
- Department of Laboratory Diagnostics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
82
|
Fiuza-Luces C, Nogales-Gadea G, García-Consuegra I, Pareja-Galeano H, Rufián-Vázquez L, Pérez LM, Andreu AL, Arenas J, Martín MA, Pinós T, Lucia A, Morán M. Muscle Signaling in Exercise Intolerance: Insights from the McArdle Mouse Model. Med Sci Sports Exerc 2017; 48:1448-58. [PMID: 27031745 DOI: 10.1249/mss.0000000000000931] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION We recently generated a knock-in mouse model (PYGM p.R50X/p.R50X) of the McArdle disease (myophosphorylase deficiency). One mechanistic approach to unveil the molecular alterations caused by myophosphorylase deficiency, which is arguably the paradigm of "exercise intolerance," is to compare the skeletal muscle tissue of McArdle, heterozygous, and healthy (wild-type [wt]) mice. METHODS We analyzed in quadriceps muscle of p.R50X/p.R50X (n = 4), p.R50X/wt (n = 6), and wt/wt mice (n = 5) (all male, 8 wk old) molecular markers of energy-sensing pathways, oxidative phosphorylation and autophagy/proteasome systems, oxidative damage, and sarcoplasmic reticulum Ca handling. RESULTS We found a significant group effect for total adenosine monophosphate-(AMP)-activated protein kinase (tAMPK) and ratio of phosphorylated (pAMPK)/tAMPK (P = 0.012 and 0.033), with higher mean values in p.R50X/p.R50X mice versus the other two groups. The absence of a massive accumulation of ubiquitinated proteins, autophagosomes, or lysosomes in p.R50X/p.R50X mice suggested no major alterations in autophagy/proteasome systems. Citrate synthase activity was lower in p.R50X/p.R50X mice versus the other two groups (P = 0.036), but no statistical effect existed for respiratory chain complexes. We found higher levels of 4-hydroxy-2-nonenal-modified proteins in p.R50X/p.R50X and p.R50X/wt mice compared with the wt/wt group (P = 0.011). Sarco(endo)plasmic reticulum ATPase 1 levels detected at 110 kDa tended to be higher in p.R50X/p.R50X and p.R50X/wt mice compared with wt/wt animals (P = 0.076), but their enzyme activity was normal. We also found an accumulation of phosphorylated sarco(endo)plasmic reticulum ATPase 1 in p.R50X/p.R50X animals. CONCLUSION Myophosphorylase deficiency causes alterations in sensory energetic pathways together with some evidence of oxidative damage and alterations in Ca handling but with no major alterations in oxidative phosphorylation capacity or autophagy/ubiquitination pathways, which suggests that the muscle tissue of patients is likely to adapt overall favorably to exercise training interventions.
Collapse
Affiliation(s)
- Carmen Fiuza-Luces
- 1Mitochondrial and Neuromuscular Diseases Laboratory and "MITOLAB-CM," Research Institute of Hospital "12 de Octubre" ("i + 12"), Madrid, SPAIN; 2Neuromuscular and Neuropediatric Research Group, Neurosciences Department, Germans Trias i Pujol Research Institute and Campus Can Ruti, Autonomous University of Barcelona, Badalona, SPAIN; 3Department of Research and Doctorate Studies, European University, Madrid, SPAIN; 4Neuromuscular and Mitochondrial Pathology Department, Vall d'Hebron University Hospital, Research Institute (VHIR), Autonomous University of Barcelona, Barcelona, SPAIN; and 5Spanish Network for Biomedical Research in Rare Diseases (CIBERER), U723, Madrid, SPAIN
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Wang Y, Wang Q, Song J. Inhibition of autophagy potentiates the proliferation inhibition activity of microRNA-7 in human hepatocellular carcinoma cells. Oncol Lett 2017; 14:3566-3572. [PMID: 28927113 PMCID: PMC5588049 DOI: 10.3892/ol.2017.6573] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 04/21/2017] [Indexed: 12/21/2022] Open
Abstract
MicroRNAs (miRNAs/miRs) are important molecules that are able to regulate multiple cellular processes in cancer cells. miR-7 has been previously identified as a tumor suppressive miRNA in several types of cancer. The aim of the present study was to investigate whether miR-7 is able to regulate autophagy in hepatocellular carcinoma (HCC) cells. It was identified that miR-7 was significantly downregulated in tumor tissues compared with adjacent normal tissues. Overexpression of miR-7 inhibited cell proliferative activity, which was partially reversed by miR-7 inhibitor. In addition, overexpression of miR-7 significantly induced an increasen in autophagic activity, and luciferase activity assay and western blot analysis identified that mammalian target of rapamycin (mTOR) was a direct target of miR-7. In addition, inhibition of autophagy by 3-methyladenine resulted in a marked enhancement of the proliferation inhibition effect of miR-7. In conclusion, miR-7 was identified to induce proliferation inhibition and autophagy in HCC cells by targeting mTOR, and inhibition of autophagy may be utilized to enhance the antitumor activity of miR-7.
Collapse
Affiliation(s)
- Yanna Wang
- Department of Infectious Diseases, Yantai Hospital For Infectious Diseases, Yantai, Shandong 264001, P.R. China
| | - Qiaoling Wang
- Department of Infectious Diseases, Yantai Hospital For Infectious Diseases, Yantai, Shandong 264001, P.R. China
| | - Jiqing Song
- Nursing Department of Yantai Yeda Hospital, Yantai, Shandong 264006, P.R. China
| |
Collapse
|
84
|
Iyama K, Matsuse M, Mitsutake N, Rogounovitch T, Saenko V, Suzuki K, Ashizawa M, Ookouchi C, Suzuki S, Mizunuma H, Fukushima T, Suzuki S, Yamashita S. Identification of Three Novel Fusion Oncogenes, SQSTM1/NTRK3, AFAP1L2/RET, and PPFIBP2/RET, in Thyroid Cancers of Young Patients in Fukushima. Thyroid 2017; 27:811-818. [PMID: 28351223 DOI: 10.1089/thy.2016.0673] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND The BRAFV600E mutation is the most frequent genetic abnormality in adult papillary thyroid carcinomas (PTCs). On the other hand, various chromosomal rearrangements are more prevalent in childhood and adolescent PTCs. The aim of the present study was to identify novel rearrangements in PTCs from young patients. METHODS Among 63 postoperative specimens of childhood and adolescent PTCs, which had been discovered by the thyroid ultrasound screening program in Fukushima, nine samples without prevalent known oncogenes, BRAFV600E, RAS, RET/PTC1, RET/PTC3, and ETV6/NTRK3, were analyzed in the current study by quantitative real-time reverse transcription polymerase chain reaction to screen for novel fusion genes by comparing transcript expression between extracellular and kinase domains of ALK, NTRK1, NTRK3, and RET. RESULTS Of the above nine samples, five samples were suspected to harbor a fusion, and using subsequent 5' rapid amplification of cDNA end (RACE), two already reported fusion oncogenes, STRN/ALK and TPR/NTRK1, and three novel fusions, SQSTM1/NTRK3, AFAP1L2/RET, and PPFIBP2/RET, were identified. Functional analyses of these three chimeric genes were performed, and their transforming abilities were confirmed through the activation of mitogen-activated protein kinase (MAPK). CONCLUSIONS Three novel fusion oncogenes have been identified in young PTC patients in Fukushima, suggesting that rare fusions may be present among the cases negative for known oncogenes in this age group and that such rearrangements can play a significant role in thyroid carcinogenesis.
Collapse
Affiliation(s)
- Keita Iyama
- 1 Department of Radiation Medical Sciences, Atomic Bomb Disease Institute , Nagasaki University, Nagasaki, Japan
- 2 Nagasaki University Graduate School of Biomedical Sciences , Nagasaki, Japan
| | - Michiko Matsuse
- 1 Department of Radiation Medical Sciences, Atomic Bomb Disease Institute , Nagasaki University, Nagasaki, Japan
| | - Norisato Mitsutake
- 1 Department of Radiation Medical Sciences, Atomic Bomb Disease Institute , Nagasaki University, Nagasaki, Japan
| | - Tatiana Rogounovitch
- 1 Department of Radiation Medical Sciences, Atomic Bomb Disease Institute , Nagasaki University, Nagasaki, Japan
| | - Vladimir Saenko
- 3 Department of Radiation Molecular Epidemiology, Atomic Bomb Disease Institute , Nagasaki University, Nagasaki, Japan
| | - Keiji Suzuki
- 1 Department of Radiation Medical Sciences, Atomic Bomb Disease Institute , Nagasaki University, Nagasaki, Japan
| | - Mai Ashizawa
- 4 Department of Thyroid and Endocrinology, Fukushima Medical University , Fukushima, Japan
| | - Chiyo Ookouchi
- 4 Department of Thyroid and Endocrinology, Fukushima Medical University , Fukushima, Japan
| | - Satoshi Suzuki
- 4 Department of Thyroid and Endocrinology, Fukushima Medical University , Fukushima, Japan
| | - Hiroshi Mizunuma
- 4 Department of Thyroid and Endocrinology, Fukushima Medical University , Fukushima, Japan
| | - Toshihiko Fukushima
- 4 Department of Thyroid and Endocrinology, Fukushima Medical University , Fukushima, Japan
| | - Shinichi Suzuki
- 4 Department of Thyroid and Endocrinology, Fukushima Medical University , Fukushima, Japan
| | - Shunichi Yamashita
- 1 Department of Radiation Medical Sciences, Atomic Bomb Disease Institute , Nagasaki University, Nagasaki, Japan
- 3 Department of Radiation Molecular Epidemiology, Atomic Bomb Disease Institute , Nagasaki University, Nagasaki, Japan
| |
Collapse
|
85
|
Duan XC, Wang W, Feng DX, Yin J, Zuo G, Chen DD, Chen ZQ, Li HY, Wang Z, Chen G. Roles of autophagy and endoplasmic reticulum stress in intracerebral hemorrhage-induced secondary brain injury in rats. CNS Neurosci Ther 2017; 23:554-566. [PMID: 28544790 DOI: 10.1111/cns.12703] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 03/23/2017] [Accepted: 04/11/2017] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVES This study aimed to evaluate the roles of autophagy and endoplasmic reticulum (ER) stress in intracerebral hemorrhage (ICH)-induced secondary brain injury (SBI) in rats. METHODS Autophagy inducer (rapamycin) and inhibitor (3-methyladenine), as well as ER stress activator (tunicamycin, TM) and inhibitor (tauroursodeoxycholic acid, TUDCA), were used. Bafilomycin A1, an inhibitor of autophagosome-lysosome fusion, was used to assess autophagic flux. RESULTS Autophagy and ER stress were enhanced in the week after ICH. At 6 hours after ICH, autophagy was excessive, while the autophagic flux was damaged at 72 hours and return to be intact at 7 days after ICH. At 6 hours after ICH, ER stress induction by TM could enhance autophagy and lead to caspase 12-mediated apoptosis and neuronal degeneration, which was further aggravated by autophagy induction. At 7 days after ICH, ER stress inhibition by TUDCA still could suppress ICH-induced SBI. And, the effects of TUDCA were enhanced by autophagy induction. CONCLUSIONS At 6 hours after ICH, excessive autophagy may participate in ER stress-induced brain injury; at 7 days after ICH, autophagy could enhance the protection of ER stress inhibitor possibly via clearing up the cell rubbish generated due to the early-stage damaged autophagic flux.
Collapse
Affiliation(s)
- Xiao-Chun Duan
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wei Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | | | - Jia Yin
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Gang Zuo
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Dong-Dong Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhou-Qing Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hai-Ying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
86
|
Yin Y, Sun G, Li E, Kiselyov K, Sun D. ER stress and impaired autophagy flux in neuronal degeneration and brain injury. Ageing Res Rev 2017; 34:3-14. [PMID: 27594375 DOI: 10.1016/j.arr.2016.08.008] [Citation(s) in RCA: 169] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 08/31/2016] [Indexed: 12/12/2022]
Abstract
Autophagy is a highly controlled lysosome-mediated function in eukaryotic cells to eliminate damaged or aged long-lived proteins and organelles. It is required for restoring cellular homeostasis in cell survival under multiple stresses. Autophagy is known to be a double-edged sword because too much activation or inhibition of autophagy can disrupt homeostatic degradation of protein and organelles within the brain and play a role in neuronal cell death. Many factors affect autophagy flux function in the brain, including endoplasmic reticulum (ER) stress, oxidative stress, and aging. Newly emerged research indicates that altered autophagy flux functionality is involved in neurodegeneration of the aged brain, chronic neurological diseases, and after traumatic and ischemic brain injuries. In search to identify neuroprotective agents that may reduce oxidative stress and stimulate autophagy, one particular neuroprotective agent docosahexaenoic acid (DHA) presents unique functions in reducing ER and oxidative stress and modulating autophagy. This review will summarize the recent findings on changes of autophagy in aging, neurodegenerative diseases, and brain injury after trauma or ischemic strokes. Discussion of DHA functions is focused on modulating ER stress and autophagy in regard to its neuroprotection and anti-tumor functions.
Collapse
Affiliation(s)
- Yan Yin
- Department of Neurology, The Second Hospital of Dalian Medical University, Dalian 116023, PR China; Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, United States.
| | - George Sun
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, United States
| | - Eric Li
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, United States
| | - Kirill Kiselyov
- Department of Biological Science, University of Pittsburgh, Pittsburgh, PA 15213, United States
| | - Dandan Sun
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, United States; Veterans Affairs Pittsburgh Health Care System, Geriatric Research, Education and Clinical Center, Pittsburgh, PA 15213, United States.
| |
Collapse
|
87
|
Haga S, Yimin, Ozaki M. Relevance of FXR-p62/SQSTM1 pathway for survival and protection of mouse hepatocytes and liver, especially with steatosis. BMC Gastroenterol 2017; 17:9. [PMID: 28086800 PMCID: PMC5237313 DOI: 10.1186/s12876-016-0568-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 12/27/2016] [Indexed: 01/12/2023] Open
Abstract
Background Liver injury and regeneration involve complicated processes and are affected by various physio-pathological conditions. Surgically, severe liver injury after surgical resection often leads to fatal liver failure, especially with some underlying pathological conditions such as steatosis. Therefore, protection from the injury of hepatocytes and liver is a serious concern in various clinical settings. Methods We studied the effects of the farnesoid X receptor (FXR) on cell survival and steatosis in mouse hepatocytes (AML12 mouse liver cells) and investigated their molecular mechanisms. We next studied whether or not FXR improves liver injury, regeneration and steatosis in a mouse model of partial hepatectomy (PH) with steatosis. Results An FXR-specific agonist, GW4064, induced expressions of the p62/SQSTM1 gene and protein in AML12 mouse liver cells. Because we previously reported p62/SQSTM1 as a key molecule for antioxidation and cell survival in hepatocytes, we next examined the activation of nuclear factor erythroid 2-related factor-2 (Nrf2) and induction of the antioxidant molecules by GW4064. GW4064 activated Nrf2 and subsequently induced antioxidant molecules (Nrf2, catalase, HO-1, and thioredoxin). We also examined expressions of pro-survival and cell protective molecules associated with p62/SQSTM1. Expectedly, GW4064 induced phosphorylation of Akt, expression of the anti-apoptotic
molecules (Bcl-xL and Bcl-2), and reduced harmful hepatic molecules (Fas ligand and Fas). GW4064 promoted
hepatocyte survival, which was cancelled by p62/SQSTM1 siRNA. These findings suggest the potential relevance of the FXR-p62/SQSTM1 pathway for the survival and protection of hepatocytes. Furthermore, GW4064 induced the expression of small heterodimer partners (SHP) and suppressed liver X receptor (LXR)-induced steatosis in hepatocytes, expecting the in vivo protective effect of FXR on liver injury especially with steatosis. In the hepatectomy model of db/db mice with fatty liver, pre-treatment by GW4064 significantly reduced post-PH liver injury (serum levels of LDH, AST & ALT and histological study) and improved steatosis. The key molecules, p62/SQSTM1, Nrf2 and SHP were upregulated in fatty liver tissue by GW4064 treatment. Conclusions The present study is the first to demonstrate the relevance of FXR-p62/SQSTM1 and -SHP in the protection against injury of hepatocytes and post-PH liver, especially with steatosis. Electronic supplementary material The online version of this article (doi:10.1186/s12876-016-0568-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sanae Haga
- Department of Biological Response and Regulation, Faculty of Health Sciences, Hokkaido University, N-12, W-5, Kita-ku, Sapporo, Hokkaido, 060-0812, Japan
| | - Yimin
- Department of Advanced Medicine, Graduate School of Medicine, Hokkaido University, N-15, W-7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Michitaka Ozaki
- Department of Biological Response and Regulation, Faculty of Health Sciences, Hokkaido University, N-12, W-5, Kita-ku, Sapporo, Hokkaido, 060-0812, Japan.
| |
Collapse
|
88
|
Roosen DA, Cookson MR. LRRK2 at the interface of autophagosomes, endosomes and lysosomes. Mol Neurodegener 2016; 11:73. [PMID: 27927216 PMCID: PMC5142374 DOI: 10.1186/s13024-016-0140-1] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 12/03/2016] [Indexed: 02/07/2023] Open
Abstract
Over the past 20 years, substantial progress has been made in identifying the underlying genetics of Parkinson's disease (PD). Of the known genes, LRRK2 is a major genetic contributor to PD. However, the exact function of LRRK2 remains to be elucidated. In this review, we discuss how familial forms of PD have led us to hypothesize that alterations in endomembrane trafficking play a role in the pathobiology of PD. We will discuss the major observations that have been made to elucidate the role of LRRK2 in particular, including LRRK2 animal models and high-throughput proteomics approaches. Taken together, these studies strongly support a role of LRRK2 in vesicular dynamics. We also propose that targeting these pathways may not only be beneficial for developing therapeutics for LRRK2-driven PD, but also for other familial and sporadic cases.
Collapse
Affiliation(s)
- Dorien A. Roosen
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bldg. 35, 35 Convent Drive, Bethesda, MD 20892-3707 USA
- School of Pharmacy, University of Reading, Whiteknights, Reading, RG6 6AP UK
| | - Mark R. Cookson
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bldg. 35, 35 Convent Drive, Bethesda, MD 20892-3707 USA
| |
Collapse
|
89
|
Höhn A, Weber D, Jung T, Ott C, Hugo M, Kochlik B, Kehm R, König J, Grune T, Castro JP. Happily (n)ever after: Aging in the context of oxidative stress, proteostasis loss and cellular senescence. Redox Biol 2016; 11:482-501. [PMID: 28086196 PMCID: PMC5228102 DOI: 10.1016/j.redox.2016.12.001] [Citation(s) in RCA: 238] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 11/30/2016] [Accepted: 12/01/2016] [Indexed: 12/14/2022] Open
Abstract
Aging is a complex phenomenon and its impact is becoming more relevant due to the rising life expectancy and because aging itself is the basis for the development of age-related diseases such as cancer, neurodegenerative diseases and type 2 diabetes. Recent years of scientific research have brought up different theories that attempt to explain the aging process. So far, there is no single theory that fully explains all facets of aging. The damage accumulation theory is one of the most accepted theories due to the large body of evidence found over the years. Damage accumulation is thought to be driven, among others, by oxidative stress. This condition results in an excess attack of oxidants on biomolecules, which lead to damage accumulation over time and contribute to the functional involution of cells, tissues and organisms. If oxidative stress persists, cellular senescence is a likely outcome and an important hallmark of aging. Therefore, it becomes crucial to understand how senescent cells function and how they contribute to the aging process. This review will cover cellular senescence features related to the protein pool such as morphological and molecular hallmarks, how oxidative stress promotes protein modifications, how senescent cells cope with them by proteostasis mechanisms, including antioxidant enzymes and proteolytic systems. We will also highlight the nutritional status of senescent cells and aged organisms (including human clinical studies) by exploring trace elements and micronutrients and on their importance to develop strategies that might increase both, life and health span and postpone aging onset.
Collapse
Affiliation(s)
- Annika Höhn
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany
| | - Daniela Weber
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; NutriAct - Competence Cluster Nutrition Research Berlin-Potsdam, 14558 Nuthetal, Germany
| | - Tobias Jung
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; German Center for Cardiovascular Research (DZHK), 10117 Berlin, Germany
| | - Christiane Ott
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; German Center for Cardiovascular Research (DZHK), 10117 Berlin, Germany
| | - Martin Hugo
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany
| | - Bastian Kochlik
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; NutriAct - Competence Cluster Nutrition Research Berlin-Potsdam, 14558 Nuthetal, Germany
| | - Richard Kehm
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany
| | - Jeannette König
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany
| | - Tilman Grune
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany; German Center for Cardiovascular Research (DZHK), 10117 Berlin, Germany; NutriAct - Competence Cluster Nutrition Research Berlin-Potsdam, 14558 Nuthetal, Germany
| | - José Pedro Castro
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany; Faculty of Medicine, Department of Biomedicine, University of Porto, 4200-319, Portugal; Institute for Innovation and Health Research (I3S), Aging and Stress Group, R. Alfredo Allen, 4200-135 Porto, Portugal.
| |
Collapse
|
90
|
Takanezawa Y, Nakamura R, Sone Y, Uraguchi S, Kiyono M. Atg5-dependent autophagy plays a protective role against methylmercury-induced cytotoxicity. Toxicol Lett 2016; 262:135-141. [DOI: 10.1016/j.toxlet.2016.09.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 08/19/2016] [Accepted: 09/17/2016] [Indexed: 12/19/2022]
|
91
|
Inhibition of autophagy enhances heat-induced apoptosis in human non-small cell lung cancer cells through ER stress pathways. Arch Biochem Biophys 2016; 607:55-66. [DOI: 10.1016/j.abb.2016.08.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 08/17/2016] [Accepted: 08/22/2016] [Indexed: 12/11/2022]
|
92
|
Violacein induces death of RAS-mutated metastatic melanoma by impairing autophagy process. Tumour Biol 2016; 37:14049-14058. [PMID: 27502397 DOI: 10.1007/s13277-016-5265-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 07/15/2016] [Indexed: 12/22/2022] Open
Abstract
Treatment of metastatic melanoma still remains a challenge, since in advanced stage it is refractory to conventional treatments. Most patients with melanoma have either B-RAF or N-RAS mutations, and these oncogenes lead to activation of the RAS-RAF-MEK-ERK and AKT signal pathway, keeping active the proliferation and survival pathways in the cell. Therefore, the identification of small molecules that block metastatic cell proliferation and induce cell death is needed. Violacein, a pigment produced by Chromobacterium violaceum found in Amazon River, has been used by our group as a biotool for scrutinizing signaling pathways associated with proliferation, survival, aggressiveness, and resistance of cancer cells. In the present study, we demonstrate that violacein diminished the viability of RAS- and RAF-mutated melanoma cells (IC50 value ∼500 nM), and more important, this effect was not abolished after treatment medium removal. Furthermore, violacein was able to reduce significantly the invasion capacity of metastatic melanoma cells in 3D culture. In the molecular context, we have shown for the first time that violacein causes a strong drop on histone deacetylase 6 expression, a proliferating activator, in melanoma cells. Besides, an inhibition of AXL and AKT was detected. All these molecular events propitiate an inhibition of autophagy, and consequently, melanoma cell death by apoptosis.
Collapse
|
93
|
Law ME, Ferreira RB, Davis BJ, Higgins PJ, Kim JS, Castellano RK, Chen S, Luesch H, Law BK. CUB domain-containing protein 1 and the epidermal growth factor receptor cooperate to induce cell detachment. Breast Cancer Res 2016; 18:80. [PMID: 27495374 PMCID: PMC4974783 DOI: 10.1186/s13058-016-0741-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 07/22/2016] [Indexed: 01/01/2023] Open
Abstract
Background While localized malignancies often respond to available therapies, most disseminated cancers are refractory. Novel approaches, therefore, are needed for the treatment of metastatic disease. CUB domain-containing protein1 (CDCP1) plays an important role in metastasis and drug resistance; the mechanism however, is poorly understood. Methods Breast cancer cell lines were engineered to stably express EGFR, CDCP1 or phosphorylation site mutants of CDCP1. These cell lines were used for immunoblot analysis or affinity purification followed by immunoblot analysis to assess protein phosphorylation and/or protein complex formation with CDCP1. Kinase activity was evaluated using phosphorylation site-specific antibodies and immunoblot analysis in in vitro kinase assays. Protein band excision and mass spectrometry was utilized to further identify proteins complexed with CDCP1 or ΔCDCP1, which is a mimetic of the cleaved form of CDCP1. Cell detachment was assessed using cell counting. Results This paper reports that CDCP1 forms ternary protein complexes with Src and EGFR, facilitating Src activation and Src-dependent EGFR transactivation. Importantly, we have discovered that a class of compounds termed Disulfide bond Disrupting Agents (DDAs) blocks CDCP1/EGFR/Src ternary complex formation and downstream signaling. CDCP1 and EGFR cooperate to induce detachment of breast cancer cells from the substratum and to disrupt adherens junctions. Analysis of CDCP1-containing complexes using proteomics techniques reveals that CDCP1 associates with several proteins involved in cell adhesion, including adherens junction and desmosomal cadherins, and cytoskeletal elements. Conclusions Together, these results suggest that CDCP1 may facilitate loss of adhesion by promoting activation of EGFR and Src at sites of cell-cell and cell-substratum contact. Electronic supplementary material The online version of this article (doi:10.1186/s13058-016-0741-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mary E Law
- Department of Pharmacology and Therapeutics, University of Florida, Acad. Res. Bldg., Room R5-210, 1200 Newell Drive, P.O. Box 100267, Gainesville, FL, 32610, USA.,UF-Health Cancer Center, University of Florida, Gainesville, FL, 32610, USA
| | - Renan B Ferreira
- Department of Chemistry, University of Florida, Gainesville, FL, 32611, USA
| | - Bradley J Davis
- Department of Pharmacology and Therapeutics, University of Florida, Acad. Res. Bldg., Room R5-210, 1200 Newell Drive, P.O. Box 100267, Gainesville, FL, 32610, USA.,UF-Health Cancer Center, University of Florida, Gainesville, FL, 32610, USA
| | - Paul J Higgins
- Center for Cell Biology and Cancer Research, Albany Medical College, Albany, NY, 12208, USA
| | - Jae-Sung Kim
- Department of Surgery, University of Florida, Gainesville, FL, 32610, USA
| | | | - Sixue Chen
- Department of Biology, Interdisciplinary Center for Biotechnology, University of Florida, Gainesville, FL, 32611, USA
| | - Hendrik Luesch
- Department of Medicinal Chemistry, University of Florida, Gainesville, FL, 32610, USA.,Center for Natural Products, Drug Discovery and Development (CNPD3), University of Florida, Gainesville, FL, 32610, USA
| | - Brian K Law
- Department of Pharmacology and Therapeutics, University of Florida, Acad. Res. Bldg., Room R5-210, 1200 Newell Drive, P.O. Box 100267, Gainesville, FL, 32610, USA. .,UF-Health Cancer Center, University of Florida, Gainesville, FL, 32610, USA. .,Center for Natural Products, Drug Discovery and Development (CNPD3), University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
94
|
Nacarelli T, Azar A, Sell C. Mitochondrial stress induces cellular senescence in an mTORC1-dependent manner. Free Radic Biol Med 2016; 95:133-54. [PMID: 27016071 DOI: 10.1016/j.freeradbiomed.2016.03.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 03/07/2016] [Accepted: 03/12/2016] [Indexed: 11/25/2022]
Abstract
Although mitochondrial stress is a key determinant of cellular homeostasis, the intracellular mechanisms by which this stress is communicated to the nucleus and its impact on cell fate decisions are not well defined. In this study, we report that activation of mTORC1 signaling triggered by mitochondrial-generated reactive oxygen species (ROS) results in activation of the senescence program. We show that exposure of human fibroblasts to nucleoside analogs commonly used in antiretroviral therapies, and known to induce mitochondrial dysfunction, increases mitochondrial ROS and leads to a rise in intracellular ROS concomitant with activation of mTORC1. In this setting, it appears that mTORC1 activates senescence through HDM2 phosphorylation, facilitating a p53-mediated response. Inhibition of mTORC1 by rapamycin decreases HDM2 phosphorylation and blocks activation of the senescence program in human cells. In addition, decreasing mitochondrial ROS directly blocks mTORC1 signaling and prevents the onset of senescence. Consistent with these results, both total and mitochondrial-specific ROS increased in cells undergoing replicative senescence along with ribosomal p70 phosphorylation. The results reveal a novel link between mitochondrial dysfunction, mTORC1 signaling, and the senescence program.
Collapse
Affiliation(s)
- Timothy Nacarelli
- Drexel University College of Medicine, 245 N 15th Street, Philadelphia, PA 19102, United States
| | - Ashley Azar
- Drexel University College of Medicine, 245 N 15th Street, Philadelphia, PA 19102, United States
| | - Christian Sell
- Drexel University College of Medicine, 245 N 15th Street, Philadelphia, PA 19102, United States.
| |
Collapse
|
95
|
Liebl MP, Hoppe T. It's all about talking: two-way communication between proteasomal and lysosomal degradation pathways via ubiquitin. Am J Physiol Cell Physiol 2016; 311:C166-78. [PMID: 27225656 DOI: 10.1152/ajpcell.00074.2016] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Selective degradation of proteins requires a fine-tuned coordination of the two major proteolytic pathways, the ubiquitin-proteasome system (UPS) and autophagy. Substrate selection and proteolytic activity are defined by a plethora of regulatory cofactors influencing each other. Both proteolytic pathways are initiated by ubiquitylation to mark substrate proteins for degradation, although the size and/or topology of the modification are different. In this context E3 ubiquitin ligases, ensuring the covalent attachment of activated ubiquitin to the substrate, are of special importance. The regulation of E3 ligase activity, competition between different E3 ligases for binding E2 conjugation enzymes and substrates, as well as their interplay with deubiquitylating enzymes (DUBs) represent key events in the cross talk between the UPS and autophagy. The coordination between both degradation routes is further influenced by heat shock factors and ubiquitin-binding proteins (UBPs) such as p97, p62, or optineurin. Mutations in enzymes and ubiquitin-binding proteins or a general decline of both proteolytic systems during aging result in accumulation of damaged and aggregated proteins. Thus further mechanistic understanding of how UPS and autophagy communicate might allow therapeutic intervention especially against age-related diseases.
Collapse
Affiliation(s)
- Martina P Liebl
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Thorsten Hoppe
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| |
Collapse
|
96
|
Krishnan VS, White Z, McMahon CD, Hodgetts SI, Fitzgerald M, Shavlakadze T, Harvey AR, Grounds MD. A Neurogenic Perspective of Sarcopenia: Time Course Study of Sciatic Nerves From Aging Mice. J Neuropathol Exp Neurol 2016; 75:464-78. [DOI: 10.1093/jnen/nlw019] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
|
97
|
Chen K, Zeng J, Xiao H, Huang C, Hu J, Yao W, Yu G, Xiao W, Xu H, Ye Z. Regulation of glucose metabolism by p62/SQSTM1 through HIF1α. J Cell Sci 2016; 129:817-30. [PMID: 26743088 PMCID: PMC4760374 DOI: 10.1242/jcs.178756] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 12/23/2015] [Indexed: 12/16/2022] Open
Abstract
The signaling adaptor sequestosome 1 (SQSTM1)/p62 is frequently overexpressed in tumors and plays an important role in the regulation of tumorigenesis. Although great progress has been made, biological roles of p62 and relevant molecular mechanisms responsible for its pro-tumor activity remain largely unknown. Here, we show that p62 knockdown reduces cell growth and the expression of glycolytic genes in a manner that depends on HIF1α activity in renal cancer cells. Knockdown of p62 decreases HIF1α levels and transcriptional activity by regulating mTORC1 activity and NF-κB nuclear translocation. Furthermore, p62 interacts directly with the von Hippel-Lindau (VHL) E3 ligase complex to modulate the stability of HIF1α. Mechanistically, p62 binds to the VHL complex and competes with HIF1α. Expression of p62 inhibits the interaction of DCNL1 (also known as DCUN1D1) with CUL2 and attenuates the neddylation of CUL2, and thus downregulates the VHL E3 ligase complex activity. Functionally, HIF1α expression is required for p62-induced glucose uptake, lactate production and soft agar colony growth. Taken together, our findings demonstrate that p62 is a crucial positive regulator of HIF1α, which is a facilitating factor in p62-enhanced tumorigenesis.
Collapse
Affiliation(s)
- Ke Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Jin Zeng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Haibing Xiao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Chunhua Huang
- College of Basic Medicine Science, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
| | - Junhui Hu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Weimin Yao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Gan Yu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Wei Xiao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Hua Xu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Zhangqun Ye
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| |
Collapse
|
98
|
Xie WY, Zhou XD, Li Q, Chen LX, Ran DH. Acid-induced autophagy protects human lung cancer cells from apoptosis by activating ER stress. Exp Cell Res 2015; 339:270-9. [DOI: 10.1016/j.yexcr.2015.11.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Revised: 10/22/2015] [Accepted: 11/06/2015] [Indexed: 02/04/2023]
|
99
|
Eino A, Kageyama S, Uemura T, Annoh H, Saito T, Narita I, Waguri S, Komatsu M. Sqstm1-GFP knock-in mice reveal dynamic actions of Sqstm1 during autophagy and under stress conditions in living cells. J Cell Sci 2015; 128:4453-61. [PMID: 26483381 PMCID: PMC4712822 DOI: 10.1242/jcs.180174] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 10/09/2015] [Indexed: 01/12/2023] Open
Abstract
Sqstm1 serves as a signaling hub and receptor for selective autophagy. Consequently, dysregulation of Sqstm1 causes imbalances in signaling pathways and disrupts proteostasis, thereby contributing to the development of human diseases. Environmental stresses influence the level of Sqstm1 by altering its expression and/or autophagic degradation, and also changes the localization of Sqstm1, making it difficult to elucidate the actions and roles of this protein. In this study, we developed knock-in mice expressing Sqstm1 fused to GFP (Sqstm1-GFP(KI/+)). Using these Sqstm1-GFP(KI/+) mice, we revealed for the first time the dynamics of endogenous Sqstm1 in living cells. Sqstm1-GFP was translocated to a restricted area of LC3-positive structures, which primarily correspond to the inside of autophagosomes, and then degraded. Moreover, exposure to arsenite induced expression of Sqstm1-GFP, followed by accumulation of the fusion protein in large aggregates that were degraded by autophagy. Furthermore, suppression of autophagy in Sqstm1-GFP(KI/+) mouse livers caused accumulation of Sqstm1-GFP and formation of GFP-positive aggregate structures, leading to severe hepatic failure. These results indicate that Sqstm1-GFP(KI/+) mice are a useful tool for analyzing Sqstm1 in living cells and intact animals.
Collapse
Affiliation(s)
- Atsushi Eino
- Department of Biochemistry, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata 951-8510, Japan Division of Clinical Nephrology and Rheumatology, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata 951-8510, Japan
| | - Shun Kageyama
- Department of Biochemistry, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata 951-8510, Japan
| | - Takefumi Uemura
- Department of Anatomy and Histology, Fukushima Medical University School of Medicine, Hikarigaoka, Fukushima 960-1295, Japan
| | - Hiromichi Annoh
- Department of Anatomy and Histology, Fukushima Medical University School of Medicine, Hikarigaoka, Fukushima 960-1295, Japan
| | - Tetsuya Saito
- Department of Biochemistry, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata 951-8510, Japan
| | - Ichiei Narita
- Division of Clinical Nephrology and Rheumatology, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata 951-8510, Japan
| | - Satoshi Waguri
- Department of Anatomy and Histology, Fukushima Medical University School of Medicine, Hikarigaoka, Fukushima 960-1295, Japan
| | - Masaaki Komatsu
- Department of Biochemistry, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata 951-8510, Japan
| |
Collapse
|
100
|
Katsuragi Y, Ichimura Y, Komatsu M. p62/SQSTM1 functions as a signaling hub and an autophagy adaptor. FEBS J 2015; 282:4672-8. [PMID: 26432171 DOI: 10.1111/febs.13540] [Citation(s) in RCA: 634] [Impact Index Per Article: 63.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 09/10/2015] [Accepted: 09/28/2015] [Indexed: 12/12/2022]
Abstract
p62/SQSTM1 is a stress-inducible cellular protein that is conserved among metazoans but not in plants and fungi. p62/SQSTM1 has multiple domains that mediate its interactions with various binding partners and it serves as a signaling hub for diverse cellular events such as amino acid sensing and the oxidative stress response. In addition, p62/SQSTM1 functions as a selective autophagy receptor for degradation of ubiqutinated substrates. In the present review, we describe the current knowledge about p62 with regard to mammalian target of rapamycin complex 1 activation, the Keap1-Nrf2 pathway and selective autophagy.
Collapse
Affiliation(s)
- Yoshinori Katsuragi
- Department of Biochemistry, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yoshinobu Ichimura
- Department of Biochemistry, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Masaaki Komatsu
- Department of Biochemistry, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|