1
|
Zinnah KMA, Munna AN, Park SY. Optimizing autophagy modulation for enhanced TRAIL-mediated therapy: Unveiling the superiority of late-stage inhibition over early-stage inhibition to overcome therapy resistance in cancer. Basic Clin Pharmacol Toxicol 2025; 136:e14110. [PMID: 39668304 DOI: 10.1111/bcpt.14110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 10/13/2024] [Accepted: 11/12/2024] [Indexed: 12/14/2024]
Abstract
Autophagy is a vital mechanism that eliminates large cytoplasmic components via lysosomal degradation to maintain cellular homeostasis. The role of autophagy in cancer treatment has been studied extensively. Autophagy primarily prevents tumour initiation by maintaining genomic stability and preventing cellular inflammation. However, autophagy also supports cancer cell survival and growth by providing essential nutrients for therapeutic resistance. Thus, autophagy has emerged as a promising strategy for overcoming resistance and enhancing anti-cancer therapy. Inhibiting autophagy significantly improves the sensitivity of lung, colorectal, breast, liver and prostate cancer cells to tumour necrosis factor-related apoptosis-inducing ligand (TRAIL). This review investigates the intricate interplay between autophagy modulation and TRAIL-based therapy, specifically focussing on comparing the efficacy of late-stage autophagy inhibition versus early-stage inhibition in overcoming cancer resistance. We expose the distinctive advantages of late-stage autophagy inhibition by exploring the mechanisms underlying autophagy's impact on TRAIL sensitivity. Current preclinical and clinical investigations are inspected, showing the potential of targeting late-stage autophagy for sensitizing resistant cancer cells to TRAIL-induced apoptosis. This review emphasizes the significance of optimizing autophagy modulation to enhance TRAIL-mediated therapy and overcome the challenge of treatment resistance in cancer. We offer insights and recommendations for guiding the development of potential therapeutic strategies aimed at overcoming the challenges posed by treatment-resistant cancers.
Collapse
Affiliation(s)
- Kazi Mohammad Ali Zinnah
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Iksan, South Korea
- Faculty of Biotechnology and Genetic Engineering, Department of Animal and Fish Biotechnology, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Ali Newaz Munna
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Iksan, South Korea
| | - Sang-Youel Park
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Iksan, South Korea
| |
Collapse
|
2
|
Thylur Puttalingaiah R, Dean MJ, Zheng L, Philbrook P, Wyczechowska D, Kayes T, Del Valle L, Danos D, Sanchez-Pino MD. Excess Potassium Promotes Autophagy to Maintain the Immunosuppressive Capacity of Myeloid-Derived Suppressor Cells Independent of Arginase 1. Cells 2024; 13:1736. [PMID: 39451254 PMCID: PMC11505641 DOI: 10.3390/cells13201736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 10/10/2024] [Accepted: 10/17/2024] [Indexed: 10/26/2024] Open
Abstract
Potassium ions (K+) are critical electrolytes that regulate multiple functions in immune cells. Recent studies have shown that the elevated concentration of extracellular potassium in the tumor interstitial fluid limits T cell effector function and suppresses the anti-tumor capacity of tumor-associated macrophages (TAMs). The effect of excess potassium on the biology of myeloid-derived suppressor cells (MDSCs), another important immune cell component of the tumor microenvironment (TME), is unknown. Here, we present data showing that increased concentrations of potassium chloride (KCl), as the source of K+ ions, facilitate autophagy by increasing the expression of the autophagosome marker LC3β. Simultaneously, excess potassium ions significantly decrease the expression of arginase I (Arg I) and inducible nitric oxide synthase (iNOS) without reducing the ability of MDSCs to suppress T cell proliferation. Further investigation reveals that excess K+ ions decrease the expression of the transcription factor C/EBP-β and alter the expression of phosphorylated kinases. While excess K+ ions downregulated the expression levels of phospho-AMPKα (pAMPKα), it increased the levels of pAKT and pERK. Additionally, potassium increased mitochondrial respiration as measured by the oxygen consumption rate (OCR). Interestingly, all these alterations induced by K+ ions were abolished by the autophagy inhibitor 3-methyladenine (3-MA). Our results suggest that hyperosmotic stress caused by excess K+ ions regulate the mitochondrial respiration and signaling pathways in MDSCs to trigger the process of autophagy to support MDSCs' immunosuppressive function by mechanisms independent of Arg I and iNOS. Overall, our in vitro and ex vivo findings offer valuable insights into the adaptations of MDSCs within the K+ ion-rich TME, which has important implications for MDSCs-targeted therapies.
Collapse
Affiliation(s)
- Ramesh Thylur Puttalingaiah
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (M.J.D.); (L.Z.); (P.P.); (D.W.); (T.K.); (L.D.V.); (D.D.)
| | - Matthew J. Dean
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (M.J.D.); (L.Z.); (P.P.); (D.W.); (T.K.); (L.D.V.); (D.D.)
| | - Liqin Zheng
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (M.J.D.); (L.Z.); (P.P.); (D.W.); (T.K.); (L.D.V.); (D.D.)
| | - Phaethon Philbrook
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (M.J.D.); (L.Z.); (P.P.); (D.W.); (T.K.); (L.D.V.); (D.D.)
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Dorota Wyczechowska
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (M.J.D.); (L.Z.); (P.P.); (D.W.); (T.K.); (L.D.V.); (D.D.)
| | - Timothy Kayes
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (M.J.D.); (L.Z.); (P.P.); (D.W.); (T.K.); (L.D.V.); (D.D.)
| | - Luis Del Valle
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (M.J.D.); (L.Z.); (P.P.); (D.W.); (T.K.); (L.D.V.); (D.D.)
| | - Denise Danos
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (M.J.D.); (L.Z.); (P.P.); (D.W.); (T.K.); (L.D.V.); (D.D.)
- Department of Interdisciplinary Oncology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
- School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Maria Dulfary Sanchez-Pino
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (M.J.D.); (L.Z.); (P.P.); (D.W.); (T.K.); (L.D.V.); (D.D.)
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
- Department of Interdisciplinary Oncology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| |
Collapse
|
3
|
Dass D, Banerjee A, Dhotre K, Sonawane V, More A, Mukherjee A. HSV-2 Manipulates Autophagy through Interferon Pathway: A Strategy for Viral Survival. Viruses 2024; 16:1383. [PMID: 39339859 PMCID: PMC11437441 DOI: 10.3390/v16091383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/14/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024] Open
Abstract
Autophagy, an evolutionarily conserved cellular process, influences the regulation of viral infections. While the existing understanding indicates that Herpes Simplex Virus type 2 (HSV-2) maintains a basal level of autophagy to support its viral yield, the precise pathways governing the induction of autophagy during HSV-2 infection remain unknown. Therefore, this study aims to explore the role of type I interferons (IFN-I) in modulating autophagy during HSV-2 infection and to decode the associated signaling pathways. Our findings revealed an interplay wherein IFN-I regulates the autophagic response during HSV-2 infection. Additionally, we investigated the cellular pathways modulated during this complex process. Exploring the intricate network of signaling events involved in autophagy induction during HSV-2 infection holds promising therapeutic implications. Identifying these pathways advances our understanding of host-virus interactions and holds the foundation for developing targeted therapeutic strategies against HSV-2. The insight gained from this study provides a platform for exploring potential therapeutic targets to restrict HSV-2 infections, addressing a crucial need in antiviral research.
Collapse
Affiliation(s)
| | | | | | | | | | - Anupam Mukherjee
- Division of Virology, ICMR-National Institute of Translational Virology and AIDS Research, Pune 411026, India; (D.D.); (A.B.); (K.D.); (V.S.); (A.M.)
| |
Collapse
|
4
|
Wang Q, Yang Z, Li Q, Zhang W, Kang P. Lithium prevents glucocorticoid-induced osteonecrosis of the femoral head by regulating autophagy. J Cell Mol Med 2024; 28:e18385. [PMID: 38801405 PMCID: PMC11129728 DOI: 10.1111/jcmm.18385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 04/17/2024] [Accepted: 04/27/2024] [Indexed: 05/29/2024] Open
Abstract
Autophagy may play an important role in the occurrence and development of glucocorticoid-induced osteonecrosis of the femoral head (GC-ONFH). Lithium is a classical autophagy regulator, and lithium can also activate osteogenic pathways, making it a highly promising therapeutic agent for GC-ONFH. We aimed to evaluate the potential therapeutic effect of lithium on GC-ONFH. For in vitro experiments, primary osteoblasts of rats were used for investigating the underlying mechanism of lithium's protective effect on GC-induced autophagy levels and osteogenic activity dysfunction. For in vivo experiments, a rat model of GC-ONFH was used for evaluating the therapeutic effect of oral lithium on GC-ONFH and underlying mechanism. Findings demonstrated that GC over-activated the autophagy of osteoblasts and reduced their osteogenic activity. Lithium reduced the over-activated autophagy of GC-treated osteoblasts through PI3K/AKT/mTOR signalling pathway and increased their osteogenic activity. Oral lithium reduced the osteonecrosis rates in a rat model of GC-ONFH, and restrained the increased expression of autophagy related proteins in bone tissues through PI3K/AKT/mTOR signalling pathway. In conclusion, lithium can restrain over-activated autophagy by activating PI3K/AKT/mTOR signalling pathway and up-regulate the expression of genes for bone formation both in GC induced osteoblasts and in a rat model of GC-ONFH. Lithium may be a promising therapeutic agent for GC-ONFH. However, the role of autophagy in the pathogenesis of GC-ONFH remains controversial. Studies are still needed to further explore the role of autophagy in the pathogenesis of GC-ONFH, and the efficacy of lithium in the treatment of GC-ONFH and its underlying mechanisms.
Collapse
Affiliation(s)
- Qiuru Wang
- Department of Orthopedic Surgery, West China HospitalSichuan UniversityChengduChina
| | - Zhouyuan Yang
- Department of Orthopedic Surgery, West China HospitalSichuan UniversityChengduChina
| | - Qianhao Li
- Department of Orthopedic Surgery, West China HospitalSichuan UniversityChengduChina
| | - Wanli Zhang
- Public Laboratory Technology Center, West China HospitalSichuan UniversityChengduChina
| | - Pengde Kang
- Department of Orthopedic Surgery, West China HospitalSichuan UniversityChengduChina
| |
Collapse
|
5
|
Wang P, Zhang H, Guo K, Liu C, Chen S, Pu B, Chen S, Feng T, Jiao H, Gao C. Rapamycin inhibits B16 melanoma cell viability invitro and invivo by inducing autophagy and inhibiting the mTOR/p70‑S6k pathway. Oncol Lett 2024; 27:140. [PMID: 38385108 PMCID: PMC10877231 DOI: 10.3892/ol.2024.14273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 01/19/2024] [Indexed: 02/23/2024] Open
Abstract
Rapamycin is an immunosuppressant that has been shown to prevent tumor growth following organ transplantation. However, its exact mode of antitumor action remains unknown. The present study used the B16-F10 (B16) murine melanoma model to explore the antitumor mechanism of rapamycin, and it was revealed that rapamycin reduced B16 cell viability in vitro and in vivo. In addition, in vitro and in vivo, the results of western blotting showed that rapamycin reduced Bcl2 expression, and enhanced the protein expression levels of cleaved caspase 3 and Bax, indicating that it can induce the apoptosis of B16 melanoma cells. Furthermore, the results of cell cycle analysis and western blotting showed that rapamycin induced B16 cell cycle arrest in the G1 phase, based on the reduction in the protein expression levels of CDK1, cyclin D1 and CDK4, as well as the increase in the percentage of cells in G1 phase. Rapamycin also significantly increased the number of autophagosomes in B16 melanoma cells, as determined by transmission electron microscopy. Furthermore, the results of RT-qPCR and western blotting showed that rapamycin upregulated the protein expression levels of microtubule-associated protein light chain 3 (LC3) and Beclin-1, while downregulating the expression of p62 in vitro and in vivo, thus indicating that rapamycin could trigger cellular autophagy. The present study revealed that rapamycin in combination with chloroquine (CQ) further increased LC3 expression compared with that in the CQ group, suggesting that rapamycin induced an increase in autophagy in B16 cells. Furthermore, the results of western blotting showed that rapamycin blocked the phosphorylation of p70 ribosomal S6 kinase (p70-S6k) and mammalian target of rapamycin (mTOR) proteins in vitro and in vivo, thus suggesting that rapamycin may exert its antitumor effect by inhibiting the phosphorylation of the mTOR/p70-S6k pathway. In conclusion, rapamycin may inhibit tumor growth by inducing cellular G1 phase arrest and apoptosis. In addition, rapamycin may exert its antitumor effects by inducing the autophagy of B16 melanoma cells in vitro and in vivo, and the mTOR/p70-S6k signaling pathway may be involved in this process.
Collapse
Affiliation(s)
- Penghui Wang
- Department of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan 570100, P.R. China
| | - Haifang Zhang
- Hainan Institute for Drug Control, Haikou, Hainan 570216, P.R. China
| | - Kaikai Guo
- Department of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan 570100, P.R. China
| | - Chun Liu
- Hainan Institute for Drug Control, Haikou, Hainan 570216, P.R. China
| | - Shimin Chen
- Department of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan 570100, P.R. China
| | - Baopeng Pu
- Department of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan 570100, P.R. China
| | - Sirun Chen
- Hainan Medical University Press, Hainan Medical University, Haikou, Hainan 570100, P.R. China
| | - Tong Feng
- School of Pharmacy, Hainan Medical University, Haikou, Hainan 570100, P.R. China
| | - Hanyi Jiao
- Department of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan 570100, P.R. China
| | - Chang Gao
- Department of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan 570100, P.R. China
| |
Collapse
|
6
|
Nguyen NTK, Vo TH, Lin SH, Le NQK. A computational predictor for autophagy-related proteins using interpretable machine learning and genetic algorithm. GENE REPORTS 2024; 34:101876. [DOI: 10.1016/j.genrep.2023.101876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
7
|
Hong JM, Munna AN, Moon JH, Kim JH, Seol JW, Eo SK, Park SY. Antiviral activity of prion protein against Japanese encephalitis virus infection in vitro and in vivo. Virus Res 2023; 338:199249. [PMID: 37858731 PMCID: PMC10598702 DOI: 10.1016/j.virusres.2023.199249] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 10/05/2023] [Accepted: 10/16/2023] [Indexed: 10/21/2023]
Abstract
Flaviviruses are a major cause of viral diseases worldwide, for which effective treatments have yet to be discovered. The prion protein (PrPc) is abundantly expressed in brain cells and has been shown to play a variety of roles, including neuroprotection, cell homeostasis, and regulation of cellular signaling. However, it is still unclear whether PrPc can protect against flaviviruses. In this study, we investigated the role of PrPc in regulating autophagy flux and its potential antiviral activity during Japanese encephalitis virus (JEV) infection. Our in vivo experiment showed that JEV was more lethal to the PrPc knocked out mice which was further supported by histological analysis, western blot and rtPCR results from infected mice brain samples. Role of PrPc against viral propagation in vitro was verified through cell survival study, protein expression and RNA replication analysis, and adenoviral vector assay by overexpressing PrPc. Further analysis indicated that after virus entry, PrPc inhibited autophagic flux that prevented JEV replication inside the host cell. Our results from in vivo and in vitro investigations demonstrate that prion protein effectively inhibited JEV propagation by regulating autophagy flux which is used by JEV to release its genetic material and replication after entering the host cell, suggesting that prion protein may be a promising therapeutic target for flavivirus infection.
Collapse
Affiliation(s)
- Jeong-Min Hong
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, 79, Gobong-ro, Iksan, Jeonbuk 54596, South Korea
| | - Ali Newaz Munna
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, 79, Gobong-ro, Iksan, Jeonbuk 54596, South Korea
| | - Ji-Hong Moon
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, 79, Gobong-ro, Iksan, Jeonbuk 54596, South Korea
| | - Jong-Hoon Kim
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, 79, Gobong-ro, Iksan, Jeonbuk 54596, South Korea
| | - Jae-Won Seol
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, 79, Gobong-ro, Iksan, Jeonbuk 54596, South Korea
| | - Seong-Kug Eo
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, 79, Gobong-ro, Iksan, Jeonbuk 54596, South Korea
| | - Sang-Youel Park
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, 79, Gobong-ro, Iksan, Jeonbuk 54596, South Korea.
| |
Collapse
|
8
|
Ferreira‐Marques M, Carvalho A, Franco AC, Leal A, Botelho M, Carmo‐Silva S, Águas R, Cortes L, Lucas V, Real AC, López‐Otín C, Nissan X, de Almeida LP, Cavadas C, Aveleira CA. Ghrelin delays premature aging in Hutchinson-Gilford progeria syndrome. Aging Cell 2023; 22:e13983. [PMID: 37858983 PMCID: PMC10726901 DOI: 10.1111/acel.13983] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/22/2023] [Accepted: 08/25/2023] [Indexed: 10/21/2023] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a rare and fatal genetic condition that arises from a single nucleotide alteration in the LMNA gene, leading to the production of a defective lamin A protein known as progerin. The accumulation of progerin accelerates the onset of a dramatic premature aging phenotype in children with HGPS, characterized by low body weight, lipodystrophy, metabolic dysfunction, skin, and musculoskeletal age-related dysfunctions. In most cases, these children die of age-related cardiovascular dysfunction by their early teenage years. The absence of effective treatments for HGPS underscores the critical need to explore novel safe therapeutic strategies. In this study, we show that treatment with the hormone ghrelin increases autophagy, decreases progerin levels, and alleviates other cellular hallmarks of premature aging in human HGPS fibroblasts. Additionally, using a HGPS mouse model (LmnaG609G/G609G mice), we demonstrate that ghrelin administration effectively rescues molecular and histopathological progeroid features, prevents progressive weight loss in later stages, reverses the lipodystrophic phenotype, and extends lifespan of these short-lived mice. Therefore, our findings uncover the potential of modulating ghrelin signaling offers new treatment targets and translational approaches that may improve outcomes and enhance the quality of life for patients with HGPS and other age-related pathologies.
Collapse
Affiliation(s)
- Marisa Ferreira‐Marques
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- CIBB – Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
- Faculty of PharmacyUniversity of CoimbraCoimbraPortugal
| | - André Carvalho
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
| | - Ana Catarina Franco
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- CIBB – Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
- Faculty of PharmacyUniversity of CoimbraCoimbraPortugal
| | - Ana Leal
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
| | - Mariana Botelho
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
| | - Sara Carmo‐Silva
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- CIBB – Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
| | - Rodolfo Águas
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
| | - Luísa Cortes
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- CIBB – Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
| | - Vasco Lucas
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
| | - Ana Carolina Real
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
| | - Carlos López‐Otín
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de OncologíaUniversidad de OviedoOviedoSpain
| | - Xavier Nissan
- CECS, I‐StemCorbeil‐EssonnesFrance
- INSERM U861, I‐StemCorbeil‐EssonnesFrance
- UEVE U861, I‐StemCorbeil‐EssonnesFrance
| | - Luís Pereira de Almeida
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- CIBB – Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
- Faculty of PharmacyUniversity of CoimbraCoimbraPortugal
| | - Cláudia Cavadas
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- CIBB – Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
- Faculty of PharmacyUniversity of CoimbraCoimbraPortugal
| | - Célia A. Aveleira
- CNC – Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- CIBB – Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
- MIA‐Portugal – Multidisciplinar Institute of AgeingUniversity of CoimbraCoimbraPortugal
| |
Collapse
|
9
|
Chueh KS, Lu JH, Juan TJ, Chuang SM, Juan YS. The Molecular Mechanism and Therapeutic Application of Autophagy for Urological Disease. Int J Mol Sci 2023; 24:14887. [PMID: 37834333 PMCID: PMC10573233 DOI: 10.3390/ijms241914887] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/25/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
Autophagy is a lysosomal degradation process known as autophagic flux, involving the engulfment of damaged proteins and organelles by double-membrane autophagosomes. It comprises microautophagy, chaperone-mediated autophagy (CMA), and macroautophagy. Macroautophagy consists of three stages: induction, autophagosome formation, and autolysosome formation. Atg8-family proteins are valuable for tracking autophagic structures and have been widely utilized for monitoring autophagy. The conversion of LC3 to its lipidated form, LC3-II, served as an indicator of autophagy. Autophagy is implicated in human pathophysiology, such as neurodegeneration, cancer, and immune disorders. Moreover, autophagy impacts urological diseases, such as interstitial cystitis /bladder pain syndrome (IC/BPS), ketamine-induced ulcerative cystitis (KIC), chemotherapy-induced cystitis (CIC), radiation cystitis (RC), erectile dysfunction (ED), bladder outlet obstruction (BOO), prostate cancer, bladder cancer, renal cancer, testicular cancer, and penile cancer. Autophagy plays a dual role in the management of urologic diseases, and the identification of potential biomarkers associated with autophagy is a crucial step towards a deeper understanding of its role in these diseases. Methods for monitoring autophagy include TEM, Western blot, immunofluorescence, flow cytometry, and genetic tools. Autophagosome and autolysosome structures are discerned via TEM. Western blot, immunofluorescence, northern blot, and RT-PCR assess protein/mRNA levels. Luciferase assay tracks flux; GFP-LC3 transgenic mice aid study. Knockdown methods (miRNA and RNAi) offer insights. This article extensively examines autophagy's molecular mechanism, pharmacological regulation, and therapeutic application involvement in urological diseases.
Collapse
Affiliation(s)
- Kuang-Shun Chueh
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, No. 100, Shih-Chuan 1st Road, San-min District, Kaohsiung 80708, Taiwan;
- Department of Urology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
- Department of Urology, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Jian-He Lu
- Center for Agricultural, Forestry, Fishery, Livestock and Aquaculture Carbon Emission Inventory and Emerging Compounds (CAFEC), General Research Service Center, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan;
| | - Tai-Jui Juan
- Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan;
- Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan
| | - Shu-Mien Chuang
- Department of Urology, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Yung-Shun Juan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, No. 100, Shih-Chuan 1st Road, San-min District, Kaohsiung 80708, Taiwan;
- Department of Urology, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| |
Collapse
|
10
|
Annangi B, Villacorta A, Vela L, Tavakolpournegari A, Marcos R, Hernández A. Effects of true-to-life PET nanoplastics using primary human nasal epithelial cells. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2023; 100:104140. [PMID: 37137422 DOI: 10.1016/j.etap.2023.104140] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/14/2023] [Accepted: 04/29/2023] [Indexed: 05/05/2023]
Abstract
Since inhalation is a relevant exposure route, studies using appropriate micro/nanoplastic (MNPLs) models, representative targeted cells, and relevant biomarkers of effect are required. We have used lab-made polyethylene terephthalate (PET)NPLs obtained from PET plastic water bottles. Human primary nasal epithelial cells (HNEpCs) were used as a model of the first barrier of the respiratory system. Cell internalization and intracellular reactive oxygen species (iROS) induction, as well as the effects on mitochondria functionality and in the modulation of the autophagy pathway, were evaluated. The data indicated significant cellular uptake and increased levels of iROS. Furthermore, a loss of mitochondrial membrane potential was observed in the exposed cells. Regarding the effects on the autophagy pathway, PETNPLs exposure significantly increases LC3-II protein expression levels. PETNPLs exposure also induced significant increases in the expression of p62. This is the first study showing that true-to-life PETNPLs can alter the autophagy pathway in HNEpCs.
Collapse
Affiliation(s)
- Balasubramanyam Annangi
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Aliro Villacorta
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Facultad de Recursos Naturales Renovables, Universidad Arturo Prat, Iquique, Chile
| | - Lourdes Vela
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Faculty of Health Sciences Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Alireza Tavakolpournegari
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Ricard Marcos
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.
| | - Alba Hernández
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.
| |
Collapse
|
11
|
Zinnah KMA, Munna AN, Seol JW, Park BY, Park SY. An Antidepressant Drug Increased TRAIL Receptor-2 Expression and Sensitized Lung Cancer Cells to TRAIL-induced Apoptosis. Anticancer Agents Med Chem 2023; 23:2225-2236. [PMID: 37859313 PMCID: PMC10788920 DOI: 10.2174/0118715206262252231004110310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/01/2023] [Accepted: 09/07/2023] [Indexed: 10/21/2023]
Abstract
BACKGROUND TRAIL has emerged as a promising therapeutic target due to its ability to selectively induce apoptosis in cancer cells while sparing normal cells. Autophagy, a highly regulated cellular recycling mechanism, is known to play a cell survival role by providing a required environment for the cell. Recent studies suggest that autophagy plays a significant role in increasing TRAIL resistance in certain cancer cells. Thus, regulating autophagy in TRAIL-mediated cancer therapy is crucial for its role in cancer treatment. OBJECTIVE Our study explored whether the antidepressant drug desipramine could enhance the ability of TRAIL to kill cancer cells by inhibiting autophagy. METHODS The effect of desipramine on TRAIL sensitivity was examined in various lung cancer cell lines. Cell viability was measured by morphological analysis, trypan blue exclusion, and crystal violet staining. Flow cytometry analysis was carried out to measure apoptosis with annexin V-PI stained cells. Western blotting, rtPCR, and immunocytochemistry were carried out to measure autophagy and death receptor expression. TEM was carried out to detect autophagy inhibition. RESULTS Desipramine treatment increased the TRAIL sensitivity in all lung cancer cell lines. Mechanistically, desipramine treatment induced death receptor expression to increase TRAIL sensitivity. This effect was confirmed when the genetic blockade of DR5 reduced the effect of desipramine in enhanced TRAIL-mediated cell death. Further investigation revealed that desipramine treatment increased the LC3 and p62 levels, indicating the inhibition of lysosomal degradation of autophagy. Notably, TRAIL, in combination with either desipramine or the autophagy inhibitor chloroquine, exhibited enhanced cytotoxicity compared to TRAIL treatment alone. CONCLUSION Our findings revealed the potential of desipramine to induce TRAIL-mediated cell death by autophagy impairment. This discovery suggests its therapeutic potential for inducing TRAIL-mediated cell death by increasing the expression of death receptors, which is caused by impairing autophagy.
Collapse
Affiliation(s)
- Kazi Mohammad Ali Zinnah
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Gobong ro, Iksan, Jeonbuk, 54596, South Korea
- Department of Animal and Fish Biotechnology, Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
| | - Ali Newaz Munna
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Gobong ro, Iksan, Jeonbuk, 54596, South Korea
| | - Jae-Won Seol
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Gobong ro, Iksan, Jeonbuk, 54596, South Korea
| | - Byung-Yong Park
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Gobong ro, Iksan, Jeonbuk, 54596, South Korea
| | - Sang-Youel Park
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Gobong ro, Iksan, Jeonbuk, 54596, South Korea
| |
Collapse
|
12
|
Volpe AR, Carmignani M, Cesare P. Hydroalcoholic extract of Buxus sempervirens shows antiproliferative effect on melanoma, colorectal carcinoma and prostate cancer cells by affecting the autophagic flow. Front Pharmacol 2023; 14:1073338. [PMID: 36891266 PMCID: PMC9986284 DOI: 10.3389/fphar.2023.1073338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 02/09/2023] [Indexed: 02/22/2023] Open
Abstract
Buxus sempervirens (European Box, Buxaceae, boxwood) has been used in folk medicine to treat rheumatism, arthritis, fever, malaria and skin ulceration while, in recent years, interest has grown on possible employment of boxwood extracts in cancer therapy. We studied the effect of hydroalcoholic extract from dried leaves of Buxus sempervirens (BSHE) on four human cell lines (BMel melanoma cells, HCT116 colorectal carcinoma cells, PC3 prostate cancer cells, and HS27 skin fibroblasts) to ascertain its possible antineoplastic activity. This extract inhibited proliferation of all cell lines in different degree as shown, after 48 h-exposure and MTS assay, by the values of GR50 (normalized growth rate inhibition50) that were 72, 48, 38, and 32 μg/mL for HS27, HCT116, PC3 and BMel cells, respectively. At the above GR50 concentrations, 99% of all studied cells remained vital showing accumulation of acidic vesicles in the cytoplasm, mainly around nuclei, whereas a higher extract concentration (125 μg/mL) was cytotoxic causing, after 48 h-exposure, death of all BMel and HCT116 cells. Immunofluorescence showed microtubule-associated light chain three protein (LC3, a marker for autophagy) to be localized on the above acidic vesicles when cells were treated for 48 h with BSHE (GR50 concentrations). Western blot analysis revealed, in all treated cells, a significant increase (2.2-3.3 times at 24 h) of LC3II, i.e., the phosphatidylethanolamine conjugate of the cytoplasmic form LC3I that is recruited in autophagosome membranes during autophagy. Such increase was accompanied, in all cell lines treated for 24 h or 48 h with BSHE, by a significant increment (2.5-3.4 times at 24 h) of p62, an autophagic cargo protein undergoing degradation during the autophagic process. Therefore, BSHE appeared to promote autophagic flow with its following blockade and consequent accumulation of autophagosome or autolysosomes. The antiproliferative effects of BSHE also involved cell cycle regulators such as p21 (HS27, BMel and HCT116 cells) and cyclin B1 (HCT116, BMel and PC3 cells) whereas, among apoptosis markers, BSHE only decreased (30%-40% at 48 h) the expression of the antiapoptotic protein survivin. It was concluded that BSHE impairs autophagic flow with arrest of proliferation and death in both fibroblasts and cancer cells, being the latter much more sensitive to these effects.
Collapse
Affiliation(s)
- Anna Rita Volpe
- Section of Pharmacology and Toxicology, Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Marco Carmignani
- Section of Pharmacology and Toxicology, Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Patrizia Cesare
- Section of Pharmacology and Toxicology, Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
13
|
Xiao W, Wang J, Wang X, Cai S, Guo Y, Ye L, Li D, Hu A, Jin S, Yuan B, Zhou Y, Li Q, Tong Q, Zheng L. Therapeutic targeting of the USP2-E2F4 axis inhibits autophagic machinery essential for zinc homeostasis in cancer progression. Autophagy 2022; 18:2615-2635. [PMID: 35253629 PMCID: PMC9629121 DOI: 10.1080/15548627.2022.2044651] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Macroautophagy/autophagy is a conserved cellular process associated with tumorigenesis and aggressiveness, while mechanisms regulating expression of autophagic machinery genes in cancers still remain elusive. Herein, we identified E2F4 (E2F transcription factor 4) as a novel transcriptional activator of cytoprotective autophagy crucial for zinc homeostasis in cancer cells. Gain- and loss-of-function studies showed that E2F4 promoted autophagy in a cell cycle-dependent manner, resulting in facilitated degradation of MT (metallothionein) proteins, elevated distribution of Zn2+ within autophagosomes, decreased labile intracellular zinc ions, and increased growth, invasion, and metastasis of gastric cancer cells. Mechanistically, E2F4 directly regulated the transcription of ATG2A (autophagy related 2A) and ULK2 (unc-51 like autophagy activating kinase 2), leading to autophagic degradation of MT1E, MT1M, and MT1X, while USP2 (ubiquitin specific peptidase 2) stabilized E2F4 protein to induce its transactivation via physical interaction and deubiquitination in cancer cells. Rescue experiments revealed that USP2 harbored oncogenic properties via E2F4-facilitated autophagy and zinc homeostasis. Emetine, a small chemical inhibitor of autophagy, was able to block interaction between UPS2 and E2F4, increase labile intracellular zinc ions, and suppress tumorigenesis and aggressiveness. In clinical gastric cancer specimens, both USP2 and E2F4 were upregulated and associated with poor outcome of patients. These findings indicate that therapeutic targeting of the USP2-E2F4 axis inhibits autophagic machinery essential for zinc homeostasis in cancer progression.Abbreviations: 3-MA: 3-methyladenine; ANOVA: analysis of variance; ATG2A: autophagy related 2A; ATG5: autophagy related 5; ATP: adenosine triphosphate; BECN1: beclin 1; BiFC: bimolecular fluorescence complementation; CCND1: cyclin D1; CDK: cyclin dependent kinase; ChIP: chromatin immunoprecipitation; CHX: cycloheximide; Co-IP: co-immunoprecipitation; DAPI: 4',6-diamidino-2-phenylindole; E2F4: E2F transcription factor 4; eATP: extracellular adenosine triphosphate; EBSS: Earle's balanced salt solution; FP: first progression; FRET: fluorescence resonance energy transfer; FUCCI: fluorescent ubiquitination-based cell cycle indicator; GFP: green fluorescent protein; GST: glutathione S-transferase; HA: hemagglutinin; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MDM2: MDM2 proto-oncogene; MKI67/Ki-67: marker of proliferation Ki-67; MT: metallothionein; MT1E: metallothionein 1E; MT1M: metallothionein 1M; MT1X: metallothionein 1X; MTT: 3-(4,5-dimethyltriazol-2-yl)-2,5-diphenyl tetrazolium bromide; OS: overall survival; PECAM1/CD31: platelet and endothelial cell adhesion molecule 1; PIK3C3: phosphatidylinositol 3-kinase catalytic subunit type 3; qPCR: quantitative PCR; RFP: red fluorescent protein; SQSTM1/p62: sequestosome 1; UBXN1: UBX domain protein 1; Ub: ubiquitin; ULK2: unc-51 like autophagy activating kinase 2; USP14: ubiquitin specific peptidase 14; USP2: ubiquitin specific peptidase 2; USP5: ubiquitin specific peptidase 5; USP7: ubiquitin specific peptidase 7; ZnCl2: zinc chloride.
Collapse
Affiliation(s)
- Wenjing Xiao
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Jianqun Wang
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xiaojing Wang
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China.,Clinical Center of Human Genomic Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Shuang Cai
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yanhua Guo
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Lin Ye
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Dan Li
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Anpei Hu
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Shikai Jin
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Boling Yuan
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yi Zhou
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Qilan Li
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Qiangsong Tong
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China.,Clinical Center of Human Genomic Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Liduan Zheng
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China.,Clinical Center of Human Genomic Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
14
|
Jeong YH, Kim TI, Oh YC, Ma JY. Selaginella tamariscina Inhibits Glutamate-Induced Autophagic Cell Death by Activating the PI3K/AKT/mTOR Signaling Pathways. Int J Mol Sci 2022; 23:ijms231911445. [PMID: 36232743 PMCID: PMC9569781 DOI: 10.3390/ijms231911445] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/20/2022] [Accepted: 09/23/2022] [Indexed: 11/21/2022] Open
Abstract
Glutamate-induced neural toxicity in autophagic neuron death is partially mediated by increased oxidative stress. Therefore, reducing oxidative stress in the brain is critical for treating or preventing neurodegenerative diseases. Selaginella tamariscina is a traditional medicinal plant for treating gastrointestinal bleeding, hematuria, leucorrhea, inflammation, chronic hepatitis, gout, and hyperuricemia. We investigate the inhibitory effects of Selaginella tamariscina ethanol extract (STE) on neurotoxicity and autophagic cell death in glutamate-exposed HT22 mouse hippocampal cells. STE significantly increased cell viability and mitochondrial membrane potential and decreased the expression of reactive oxygen species, lactate dehydrogenase release, and cell apoptosis in glutamate-exposed HT22 cells. In addition, while glutamate induced the excessive activation of mitophagy, STE attenuated glutamate-induced light chain (LC) 3 II and Beclin-1 expression and increased p62 expression. Furthermore, STE strongly enhanced the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) phosphorylation activation. STE strongly inhibited glutamate-induced autophagy by activating the PI3K/Akt/mTOR signaling pathway. In contrast, the addition of LY294002, a PI3K/Akt inhibitor, remarkably suppressed cell viability and p-Akt and p62 expression, while markedly increasing the expression of LC3 II and Beclin-1. Our findings indicate that autophagy inhibition by activating PI3K/Akt/mTOR phosphorylation levels could be responsible for the neuroprotective effects of STE on glutamate neuronal damage.
Collapse
Affiliation(s)
| | | | - You-Chang Oh
- Correspondence: (Y.-C.O.); (J.Y.M.); Tel.: +82-53-940-3882 (Y.-C.O.); +82-53-940-3812 (J.Y.M.)
| | - Jin Yeul Ma
- Correspondence: (Y.-C.O.); (J.Y.M.); Tel.: +82-53-940-3882 (Y.-C.O.); +82-53-940-3812 (J.Y.M.)
| |
Collapse
|
15
|
Hwang SH, Kim HG, Lee M. Paradoxical downregulation of LPAR3 exerts tumor-promoting activity through autophagy induction in Ras-transformed cells. BMC Cancer 2022; 22:969. [PMID: 36088312 PMCID: PMC9463806 DOI: 10.1186/s12885-022-10053-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 09/02/2022] [Indexed: 11/23/2022] Open
Abstract
Background Lysophosphatidic acid receptor 3 (LPAR3) is coupled to Gαi/o and Gα11/q signaling. Previously, we reported that LPAR3 is highly methylated in carcinogen-induced transformed cells. Here, we demonstrate that LPAR3 exhibits malignant transforming activities, despite being downregulated in transformed cells. Methods The LPAR3 knockout (KO) in NIH 3 T3 and Bhas 42 cells was established using the CRISPR/Cas9 system. Both RT-PCR and DNA sequencing were performed to confirm the KO of LPAR3. The cellular effects of LPAR3 KO were further examined by WST-1 assay, immunoblotting analysis, transwell migration assay, colony formation assay, wound scratch assday, in vitro cell transformation assay, and autophagy assay. Results In v-H-ras-transformed cells (Ras-NIH 3 T3) with LPAR3 downregulation, ectopic expression of LPAR3 significantly enhanced the migration. In particular, LPAR3 knockout (KO) in Bhas 42 (v-Ha-ras transfected Balb/c 3 T3) and NIH 3 T3 cells caused a decrease in cell survival, transformed foci, and colony formation. LPAR3 KO led to the robust accumulation of LC3-II and autophagosomes and inhibition of autophagic flux by disrupting autophagosome fusion with lysosome. Conversely, autolysosome maturation proceeded normally in Ras-NIH 3 T3 cells upon LPAR3 downregulation. Basal phosphorylation of MEK and ERK markedly increased in Ras-NIH 3 T3 cells, whereas being significantly lower in LPAR3 KO cells, suggesting that increased MEK signaling is involved in autophagosome–lysosome fusion in Ras-NIH 3 T3 cells. Conclusions Paradoxical downregulation of LPAR3 exerts cooperative tumor-promoting activity with MEK activation through autophagy induction in Ras-transformed cells. Our findings have implications for the development of cancer chemotherapeutic approaches. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-10053-0.
Collapse
|
16
|
Dengler F, Sternberg F, Grages M, Kästner SBR, Verhaar N. Adaptive mechanisms in no flow vs. low flow ischemia in equine jejunum epithelium: Different paths to the same destination. Front Vet Sci 2022; 9:947482. [PMID: 36157182 PMCID: PMC9493374 DOI: 10.3389/fvets.2022.947482] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/17/2022] [Indexed: 01/18/2023] Open
Abstract
Intestinal ischemia reperfusion injury (IRI) is a frequent complication of equine colic. Several mechanisms may be involved in adaptation of the intestinal epithelium to IRI and might infer therapeutic potential, including hypoxia-inducible factor (HIF) 1α, AMP-activated protein kinase (AMPK), nuclear factor-erythroid 2-related factor 2 (NRF2), and induction of autophagy. However, the mechanisms supporting adaptation and thus cellular survival are not completely understood yet. We investigated the activation of specific adaptation mechanisms in both no and low flow ischemia and reperfusion simulated in equine jejunum epithelium in vivo. We found an activation of HIF1α in no and low flow ischemia as indicated by increased levels of HIF1α target genes and phosphorylation of AMPKα tended to increase during ischemia. Furthermore, the protein expression of the autophagy marker LC3B in combination with decreased expression of nuclear-encoded mitochondrial genes indicates an increased rate of mitophagy in equine intestinal IRI, possibly preventing damage by mitochondria-derived reactive oxygen species (ROS). Interestingly, ROS levels were increased only shortly after the onset of low flow ischemia, which may be explained by an increased antioxidative defense, although NFR2 was not activated in this setup. In conclusion, we could demonstrate that a variety of adaptation mechanisms manipulating different aspects of cellular homeostasis are activated in IRI irrespective of the ischemia model, and that mitophagy might be an important factor for epithelial survival following small intestinal ischemia in horses that should be investigated further.
Collapse
Affiliation(s)
- Franziska Dengler
- Department of Biochemical Sciences, Institute of Physiology, Pathophysiology and Biophysics, University of Veterinary Medicine, Vienna, Austria
- *Correspondence: Franziska Dengler
| | - Felix Sternberg
- Department of Biochemical Sciences, Institute of Physiology, Pathophysiology and Biophysics, University of Veterinary Medicine, Vienna, Austria
| | - Marei Grages
- Clinic for Horses, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Sabine BR Kästner
- Clinic for Horses, University of Veterinary Medicine Hannover, Hannover, Germany
- Small Animal Clinic, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Nicole Verhaar
- Clinic for Horses, University of Veterinary Medicine Hannover, Hannover, Germany
| |
Collapse
|
17
|
Matsuo I, Kawamura N, Ohnuki Y, Suita K, Ishikawa M, Matsubara T, Mototani Y, Ito A, Hayakawa Y, Nariyama M, Morii A, Kiyomoto K, Tsunoda M, Gomi K, Okumura S. Role of TLR4 signaling on Porphyromonas gingivalis LPS-induced cardiac dysfunction in mice. PLoS One 2022; 17:e0258823. [PMID: 35648750 PMCID: PMC9159598 DOI: 10.1371/journal.pone.0258823] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 05/14/2022] [Indexed: 12/02/2022] Open
Abstract
Oral infections, particularly periodontitis, are a well-established risk factor for cardiovascular diseases, although the molecular mechanisms involved remain elusive. The aims of the present study were to investigate the effects of lipopolysaccharide derived from Porphyromonas gingivalis (PG-LPS) on cardiac function in mice, and to elucidate the underlying mechanisms. Mice (C57BL/6) were injected with PG-LPS (0.8 mg/kg/day) with or without an inhibitor of Toll-like receptor 4 (TLR4) signaling (TAK-242, 0.8 mg/kg/day) for 4 weeks. Left ventricular ejection function was significantly decreased at 1 week (from 67 ± 0.5 to 58 ± 1.2%) and remained low at 4 weeks (57 ± 1.0%). The number of apoptotic myocytes was increased (approximately 7.4-fold), the area of fibrosis was increased (approximately 3.3-fold) and the number of 8-hydroxydeoxyguanosine-positive myocytes, a sensitive indicator of oxidative DNA damage, was increased (approximately 7.6-fold) at 4 weeks in the heart of PG-LPS treated mice. However, levels of various serum pro-inflammatory cytokines in PG-LPS-treated mice were similar to those in control mice. The impairment of cardiac function in PG-LPS-treated mice appears to involve activation of TLR4-NADPH oxidase (NOX) 4 signaling, leading to abundant production of reactive oxygen species and Ca2+ leakage from sarcoplastic reticulumn induced by calmodulin kinase II (CaMKII)-mediated phosphorylation of phospholamban (at Thr-17) and ryanodine receptor 2 (at Ser-2448). Pharmacological inhibition of TLR4 with TAK-242 attenuated the changes in cardiac function in PG-LPS-treated mice. Our results indicate that TLR4-NOX4 signaling may be a new therapeutic target for treatment of cardiovascular diseases in patients with periodontitis.
Collapse
Affiliation(s)
- Ichiro Matsuo
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Naoya Kawamura
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yoshiki Ohnuki
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Kenji Suita
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Misao Ishikawa
- Department of Oral Anatomy, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Takehiro Matsubara
- Division of BioBank, Center for Comprehensive Genomic Medicine, Okayama University Hospital, Okayama, Japan
| | - Yasumasa Mototani
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Aiko Ito
- Department of Orthodontics, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yoshio Hayakawa
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Dental Anesthesiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Megumi Nariyama
- Department of Pediatric Dentistry, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Akinaka Morii
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Kenichi Kiyomoto
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Michinori Tsunoda
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Kazuhiro Gomi
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Satoshi Okumura
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- * E-mail:
| |
Collapse
|
18
|
Chiu CC, Chen YC, Bow YD, Chen JYF, Liu W, Huang JL, Shu ED, Teng YN, Wu CY, Chang WT. diTFPP, a Phenoxyphenol, Sensitizes Hepatocellular Carcinoma Cells to C2-Ceramide-Induced Autophagic Stress by Increasing Oxidative Stress and ER Stress Accompanied by LAMP2 Hypoglycosylation. Cancers (Basel) 2022; 14:cancers14102528. [PMID: 35626132 PMCID: PMC9139631 DOI: 10.3390/cancers14102528] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/14/2022] [Accepted: 05/16/2022] [Indexed: 12/29/2022] Open
Abstract
Simple Summary Chemotherapy is the major treatment modality for advanced or unresectable hepatocellular carcinoma (HCC). Unfortunately, chemoresistance carries a poor prognosis in HCC patients. Exogenous ceramide, a sphingolipid, has been well documented to exert anticancer effects; however, recent reports showed ceramide resistance, which limits the development of the ceramide-based cancer treatment diTFPP, a novel phenoxyphenol compound that has been shown to sensitize HCC cells to ceramide treatment. Here, we further clarified the mechanism underlying diTFPP-mediated sensitization of HCC to C2-ceramide-induced stresses, including oxidative stress, ER stress, and autophagic stress, especially the modulation of LAMP2 glycosylation, the lysosomal membrane protein that is crucial for autophagic fusion. This study may shed light on the mechanism of ceramide resistance and help in the development of adjuvants for ceramide-based cancer therapeutics. Abstract Hepatocellular carcinoma (HCC), the most common type of liver cancer, is the leading cause of cancer-related mortality worldwide. Chemotherapy is the major treatment modality for advanced or unresectable HCC; unfortunately, chemoresistance results in a poor prognosis for HCC patients. Exogenous ceramide, a sphingolipid, has been well documented to exert anticancer effects. However, recent reports suggest that sphingolipid metabolism in ceramide-resistant cancer cells favors the conversion of exogenous ceramides to prosurvival sphingolipids, conferring ceramide resistance to cancer cells. However, the mechanism underlying ceramide resistance remains unclear. We previously demonstrated that diTFPP, a novel phenoxyphenol compound, enhances the anti-HCC effect of C2-ceramide. Here, we further clarified that treatment with C2-ceramide alone increases the protein level of CERS2, which modulates sphingolipid metabolism to favor the conversion of C2-ceramide to prosurvival sphingolipids in HCC cells, thus activating the unfolded protein response (UPR), which further initiates autophagy and the reversible senescence-like phenotype (SLP), ultimately contributing to C2-ceramide resistance in these cells. However, cotreatment with diTFPP and ceramide downregulated the protein level of CERS2 and increased oxidative and endoplasmic reticulum (ER) stress. Furthermore, insufficient LAMP2 glycosylation induced by diTFPP/ceramide cotreatment may cause the failure of autophagosome–lysosome fusion, eventually lowering the threshold for triggering cell death in response to C2-ceramide. Our study may shed light on the mechanism of ceramide resistance and help in the development of adjuvants for ceramide-based cancer therapeutics.
Collapse
Affiliation(s)
- Chien-Chih Chiu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.C.); (Y.-C.C.); (J.Y.-F.C.); (W.L.); (E.-D.S.); (C.-Y.W.)
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan
- The Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yen-Chun Chen
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.C.); (Y.-C.C.); (J.Y.-F.C.); (W.L.); (E.-D.S.); (C.-Y.W.)
| | - Yung-Ding Bow
- Ph.D. Program in Life Sciences, College of Life Science, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Jeff Yi-Fu Chen
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.C.); (Y.-C.C.); (J.Y.-F.C.); (W.L.); (E.-D.S.); (C.-Y.W.)
| | - Wangta Liu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.C.); (Y.-C.C.); (J.Y.-F.C.); (W.L.); (E.-D.S.); (C.-Y.W.)
| | - Jau-Ling Huang
- Department of Bioscience Technology, College of Health Science, Chang Jung Christian University, Tainan 711, Taiwan;
| | - En-De Shu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.C.); (Y.-C.C.); (J.Y.-F.C.); (W.L.); (E.-D.S.); (C.-Y.W.)
| | - Yen-Ni Teng
- Department of Biological Sciences and Technology, National University of Tainan, Tainan 700, Taiwan;
| | - Chang-Yi Wu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.C.); (Y.-C.C.); (J.Y.-F.C.); (W.L.); (E.-D.S.); (C.-Y.W.)
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan
| | - Wen-Tsan Chang
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Correspondence: ; Tel.: +886-7-312-1101 (ext. 7651); Fax: +886-7-312-6992
| |
Collapse
|
19
|
He YQ, Qiao YL, Xu S, Jiao WE, Yang R, Kong YG, Tao ZZ, Chen SM. Allergen induces CD11c+ dendritic cell autophagy to aggravate allergic rhinitis through promoting immune imbalance. Int Immunopharmacol 2022; 106:108611. [DOI: 10.1016/j.intimp.2022.108611] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/27/2022] [Accepted: 02/04/2022] [Indexed: 12/14/2022]
|
20
|
Coenzyme Q 0 Inhibits NLRP3 Inflammasome Activation through Mitophagy Induction in LPS/ATP-Stimulated Macrophages. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4266214. [PMID: 35035661 PMCID: PMC8759827 DOI: 10.1155/2022/4266214] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 12/04/2021] [Accepted: 12/16/2021] [Indexed: 12/20/2022]
Abstract
Coenzyme Q (CoQ) analogs with a variable number of isoprenoid units have exhibited as anti-inflammatory as well as antioxidant molecules. Using novel quinone derivative CoQ0 (2,3-dimethoxy-5-methyl-1,4-benzoquinone, zero side chain isoprenoid), we studied its molecular activities against LPS/ATP-induced inflammation and redox imbalance in murine RAW264.7 macrophages. CoQ0's non- or subcytotoxic concentration suppressed the NLRP3 inflammasome and procaspase-1 activation, followed by downregulation of IL1β expression in LPS/ATP-stimulated RAW264.7 macrophages. Similarly, treatment of CoQ0 led to LC3-I/II accumulation and p62/SQSTM1 activation. An increase in the Beclin-1/Bcl-2 ratio and a decrease in the expression of phosphorylated PI3K/AKT, p70 S6 kinase, and mTOR showed that autophagy was activated. Besides, CoQ0 increased Parkin protein to recruit damaged mitochondria and induced mitophagy in LPS/ATP-stimulated RAW264.7 macrophages. CoQ0 inhibited LPS/ATP-stimulated ROS generation in RAW264.7 macrophages. Notably, when LPS/ATP-stimulated RAW264.7 macrophages were treated with CoQ0, Mito-TEMPO (a mitochondrial ROS inhibitor), or N-acetylcysteine (NAC, a ROS inhibitor), there was a significant reduction of LPS/ATP-stimulated NLRP3 inflammasome activation and IL1β expression. Interestingly, treatment with CoQ0 or Mito-TEMPO, but not NAC, significantly increased LPS/ATP-induced LC3-II accumulation indicating that mitophagy plays a key role in the regulation of CoQ0-inhibited NLRP3 inflammasome activation. Nrf2 knockdown significantly decreased IL1β expression in LPS/ATP-stimulated RAW264.7 macrophages suggesting that CoQ0 inhibited ROS-mediated NLRP3 inflammasome activation and IL1β expression was suppressed due to the Nrf2 activation. Hence, this study showed that CoQ0 might be a promising candidate for the therapeutics of inflammatory disorders due to its effective anti-inflammatory as well as antioxidant properties.
Collapse
|
21
|
Theofani E, Semitekolou M, Samitas K, Mais A, Galani IE, Triantafyllia V, Lama J, Morianos I, Stavropoulos A, Jeong S, Andreakos E, Razani B, Rovina N, Xanthou G. TFEB signaling attenuates NLRP3-driven inflammatory responses in severe asthma. Allergy 2022; 77:2131-2146. [PMID: 35038351 DOI: 10.1111/all.15221] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 12/20/2021] [Accepted: 12/26/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND NLRP3-driven inflammatory responses by circulating and lung-resident monocytes are critical drivers of asthma pathogenesis. Autophagy restrains NLRP3-induced monocyte activation in asthma models. Yet, the effects of autophagy and its master regulator, transcription factor EB (TFEB), on monocyte responses in human asthma remain unexplored. Here, we investigated whether activation of autophagy and TFEB signaling suppress inflammatory monocyte responses in asthmatic individuals. METHODS Peripheral blood CD14+ monocytes from asthmatic patients (n = 83) and healthy controls (n = 46) were stimulated with LPS/ATP to induce NLRP3 activation with or without the autophagy inducer, rapamycin. ASC specks, caspase-1 activation, IL-1β and IL-18 levels, mitochondrial function, ROS release, and mTORC1 signaling were examined. Autophagy was evaluated by LC3 puncta formation, p62/SQSTM1 degradation and TFEB activation. In a severe asthma (SA) model, we investigated the role of NLRP3 signaling using Nlrp3-/- mice and/or MCC950 administration, and the effects of TFEB activation using myeloid-specific TFEB-overexpressing mice or administration of the TFEB activator, trehalose. RESULTS We observed increased NLRP3 inflammasome activation, concomitant with impaired autophagy in circulating monocytes that correlated with asthma severity. SA patients also exhibited mitochondrial dysfunction and ROS accumulation. Autophagy failed to inhibit NLRP3-driven monocyte responses, due to defective TFEB activation and excessive mTORC1 signaling. NLRP3 blockade restrained inflammatory cytokine release and linked airway disease. TFEB activation restored impaired autophagy, attenuated NLRP3-driven pulmonary inflammation, and ameliorated SA phenotype. CONCLUSIONS Our studies uncover a crucial role for TFEB-mediated reprogramming of monocyte inflammatory responses, raising the prospect that this pathway can be therapeutically harnessed for the management of SA.
Collapse
Affiliation(s)
- Efthymia Theofani
- Cellular Immunology Laboratory Center for Basic Research Biomedical Research Foundation of the Academy of Athens (BRFAA) Athens Greece
- 1st Department of Respiratory Medicine Medical School ‘Sotiria’ Athens Chest Diseases HospitalNational Kapodistrian University of Athens Athens Greece
| | - Maria Semitekolou
- Cellular Immunology Laboratory Center for Basic Research Biomedical Research Foundation of the Academy of Athens (BRFAA) Athens Greece
| | - Konstantinos Samitas
- Cellular Immunology Laboratory Center for Basic Research Biomedical Research Foundation of the Academy of Athens (BRFAA) Athens Greece
- 7th Respiratory Clinic and Asthma Center of the ‘Sotiria’ Athens Chest Hospital Athens Greece
| | - Annie Mais
- Cellular Immunology Laboratory Center for Basic Research Biomedical Research Foundation of the Academy of Athens (BRFAA) Athens Greece
| | - Ioanna E. Galani
- Laboratory of Immunobiology Center for Clinical, Experimental Surgery and Translational Research BRFAA Athens Greece
| | - Vasiliki Triantafyllia
- Laboratory of Immunobiology Center for Clinical, Experimental Surgery and Translational Research BRFAA Athens Greece
| | - Joanna Lama
- Cellular Immunology Laboratory Center for Basic Research Biomedical Research Foundation of the Academy of Athens (BRFAA) Athens Greece
| | - Ioannis Morianos
- Cellular Immunology Laboratory Center for Basic Research Biomedical Research Foundation of the Academy of Athens (BRFAA) Athens Greece
| | - Athanasios Stavropoulos
- Laboratory of Immunobiology Center for Clinical, Experimental Surgery and Translational Research BRFAA Athens Greece
| | - Se‐Jin Jeong
- Department of Medicine Cardiovascular Division, and Department of Pathology & Immunology Washington University School of Medicine St. Louis Missouri USA
| | - Evangelos Andreakos
- Laboratory of Immunobiology Center for Clinical, Experimental Surgery and Translational Research BRFAA Athens Greece
| | - Babak Razani
- Department of Medicine Cardiovascular Division, and Department of Pathology & Immunology Washington University School of Medicine St. Louis Missouri USA
- John Cochran VA Medical Center St. Louis Missouri USA
| | - Nikoletta Rovina
- 1st Department of Respiratory Medicine Medical School ‘Sotiria’ Athens Chest Diseases HospitalNational Kapodistrian University of Athens Athens Greece
| | - Georgina Xanthou
- Cellular Immunology Laboratory Center for Basic Research Biomedical Research Foundation of the Academy of Athens (BRFAA) Athens Greece
| |
Collapse
|
22
|
Locatelli AG, Cenci S. Autophagy and longevity: Evolutionary hints from hyper-longevous mammals. Front Endocrinol (Lausanne) 2022; 13:1085522. [PMID: 36605941 PMCID: PMC9807614 DOI: 10.3389/fendo.2022.1085522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/02/2022] [Indexed: 12/24/2022] Open
Abstract
Autophagy is a fundamental multi-tasking adaptive cellular degradation and recycling strategy. Following its causal implication in age-related decline, autophagy is currently among the most broadly studied and challenged mechanisms within aging research. Thanks to these efforts, new cellular nodes interconnected with this phylogenetically ancestral pathway and unexpected roles of autophagy-associated genetic products are unveiled daily, yet the history of functional adaptations of autophagy along its evolutive trail is poorly understood and documented. Autophagy is traditionally studied in canonical and research-wise convenient model organisms such as yeast and mice. However, unconventional animal models endowed with extended longevity and exemption from age-related diseases offer a privileged perspective to inquire into the role of autophagy in the evolution of longevity. In this mini review we retrace the appearance and functions evolved by autophagy in eukaryotic cells and its protective contribution in the pathophysiology of aging.
Collapse
Affiliation(s)
- Andrea G. Locatelli
- Age Related Diseases, San Raffaele Scientific Institute, Division of Genetics and Cell Biology, Milano, Italy
- *Correspondence: Andrea G. Locatelli, ; Simone Cenci,
| | - Simone Cenci
- Age Related Diseases, San Raffaele Scientific Institute, Division of Genetics and Cell Biology, Milano, Italy
- University Vita-Salute San Raffaele, Milano, Italy
- *Correspondence: Andrea G. Locatelli, ; Simone Cenci,
| |
Collapse
|
23
|
Yeom H, Hwang SH, Kim HG, Lee M. Increase in the sensitivity to PLX4720 through inhibition of transcription factor EB-dependent autophagy in BRAF inhibitor-resistant cells. Toxicol Res 2022; 38:35-44. [PMID: 35070939 PMCID: PMC8748576 DOI: 10.1007/s43188-021-00109-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 09/08/2021] [Accepted: 09/22/2021] [Indexed: 01/03/2023] Open
Abstract
Long-term treatment with oncogenic BRAF inhibitors confers resistance to BRAF inhibitor monotherapy. In this study, a combination treatment strategy with autophagy inhibitors was proposed to increase the sensitivity of BRAF mutant containing A375P melanoma cells that have developed resistance to BRAF inhibitors. We found that the A375P/Multi-drug resistance (A375P/Mdr) cells, which are resistant to both BRAF inhibitors and MEK inhibitors, exhibited a higher basal autophagic flux compared to their parental A375P cells, as determined by tandem mRFP-GFP-tagged LC3 imaging assay and LC3 conversion. In addition, transcription factor EB (TFEB), which acts as a transcription factor regulating the transcription of autophagy-related genes, was much more localized in the nucleus in A375P/Mdr cells than in A375P cells, indicating that the increase in basal autophagic flux was TFEB-dependent. In particular, the overexpression of an activated form of TFEB (TFEBAA) caused a modest increase in PLX4720 resistance in A375P/Mdr cells. Interestingly, treatment with early stage autophagy inhibitors reversed BRAF inhibitor-induced resistance, whereas late autophagy inhibition did not. In contrast, inhibition of ER stress by 4-phenylbutyric acid suppressed basal autophagic flux. Moreover, ER stress inhibition significantly remarkably inhibited the nuclear localization of TFEB, resulting in an increase in the sensitivity of A375P/Mdr cells to PLX4720. Taken together, these results suggest that autophagy may be an important mechanism of acquired resistance to BRAF inhibitors. Thus, targeting autophagy may be suitable for the treatment of tumors resistant to BRAF inhibitor.
Collapse
Affiliation(s)
- Hojin Yeom
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, 119 Academy-ro, Yeonsu-gu, Incheon, 22012 Republic of Korea
| | - Sung-Hee Hwang
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, 119 Academy-ro, Yeonsu-gu, Incheon, 22012 Republic of Korea
| | - Hye-Gyo Kim
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, 119 Academy-ro, Yeonsu-gu, Incheon, 22012 Republic of Korea
| | - Michael Lee
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, 119 Academy-ro, Yeonsu-gu, Incheon, 22012 Republic of Korea
| |
Collapse
|
24
|
Selvan SR, Brichetti JA, Thurber DB, Botting GM, Bertenshaw GP. Functional Profiling of Head and Neck/Esophageal Squamous Cell Carcinoma to Predict Cetuximab Response. Cancer Biother Radiopharm 2021. [PMID: 34846938 DOI: 10.1089/cbr.2021.0283] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Background: Cetuximab, an epidermal growth factor receptor (EGFR)-targeting antibody, remains the only Food and Drug Administration-approved targeted therapy for squamous cell carcinoma (SCC) of head and neck/esophagus. However, in clinical trials, cetuximab only benefited a subset of patients and frequently caused toxicity. Predicting which patients respond to cetuximab remains unsolved. The authors sought to identify predictive biomarkers in EGFR signaling and autophagy pathways, which may be impacted by cetuximab under certain treatment conditions. Methods: In vitro responses of SCC cell lines to cetuximab under various nutrient conditions were assessed by WST-8 growth assay. Functional profiles of several EGFR signaling biomarkers were investigated by Luminex-based assays and corroborated with immunoblots. Autophagy markers were analyzed with immunoblots. Results: In vitro growth response assays identified cetuximab responder and nonresponder cell lines. Optimal growth conditions and growth factors enhanced responses, and even reversed nonresponsiveness in some cell lines. Strong correlation was found between response in growth assays (reference assay) and dynamic changes in p-Erk1/2 and LC3-II (index assays). Conclusions: This study indicates that nutrient modification may enhance cetuximab response in SCC patients. Biomarker results strengthen the hypothesis that dynamic biomarkers can be used to predict patient response to cetuximab. Future studies are warranted to test in more complex samples including patient-derived tumor tissues.
Collapse
|
25
|
Shen T, Li X, Jin B, Loor JJ, Aboragah A, Ju L, Fang Z, Yu H, Chen M, Zhu Y, Ouyang H, Song Y, Wang Z, Du X, Liu G. Free fatty acids impair autophagic activity and activate nuclear factor kappa B signaling and NLR family pyrin domain containing 3 inflammasome in calf hepatocytes. J Dairy Sci 2021; 104:11973-11982. [PMID: 34454753 DOI: 10.3168/jds.2021-20273] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 07/12/2021] [Indexed: 12/14/2022]
Abstract
Free fatty acids (FFA)-induced hepatic inflammation agravates liver injury and metabolic dysfunction in dairy cows with ketosis or fatty liver. Under stressful conditions, autophagy is generally considered as a cell protection mechanism, but whether the FFA-induced inflammatory and stress effect on hepatocytes involves an autophagy response is not well known. Thus, the objective of this study was to investigate the effects of FFA on autophagy and the role of autophagy in the activation of NF-κB (nuclear factor kappa B) signaling and NLRP3 (NLR family pyrin domain containing 3) inflammasome in calf hepatocytes. Calf hepatocytes were isolated from 3 healthy Holstein female new-born calves (1 d of age, 30-40 kg) and exposed to various concentrations of FFA (0, 0.3, 0.6, or 1.2 mM) after treatment with or without the autophagy inhibitor chloroquine (CQ) or the autophagy activator rapamycin. Expression of autophagy markers, LC3 (microtubule-associated protein 1 light chain 3) and p62 (sequestosome 1), NF-κB signaling, and NLRP3 inflammasome-related molecules were analyzed via western blot and quantitative real-time PCR. Results revealed that 0.6 and 1.2 mM FFA activated NF-κB signaling and NLRP3 inflammasome as indicated by an elevated ratio of p-NF-κB/NF-κB, protein abundance of NLRP3 and CASP1 (caspase 1), activity of CASP1, and mRNA abundance of IL1B and IL18. In addition, hepatocyte treated with 0.6 and 1.2 mM FFA or autophagy inhibitor CQ displayed increased protein abundance of p62 and LC3-II. Moreover, there was no difference in protein abundance of p62 and LC3-II between calf hepatocytes treated with 1.2 mM FFA and 1.2 mM FFA plus CQ, indicating that FFA inhibits autophagic activity in calf hepatocytes. Treatment with CQ led to overactivation of NF-κB signaling and NLRP3 inflammasome. Furthermore, CQ plus 1.2 mM FFA aggravated FFA-induced inflammation. In contrast, induction of autophagy by rapamycin ameliorated the FFA-activated NF-κB signaling and NLRP3 inflammasome as demonstrated by a lower ratio of p-NF-κB/NF-κB, protein abundance of NLRP3 and CASP1, activity of CASP1, and mRNA abundance of IL1B and IL18. Overall, inhibition of autophagy exacerbated, whereas induction of autophagy alleviated, FFA-induced inflammatory processes in calf hepatocytes, suggesting that impairment of autophagy might be partly responsible for hepatic inflammation and subsequent liver injury in dairy cows with ketosis or fatty liver. As such, regulation of autophagy may be an effective therapeutic strategy for controlling overt inflammatory responses in vivo.
Collapse
Affiliation(s)
- Taiyu Shen
- Key Laboratory of Zoonoses Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, Jilin Province, 130062, China
| | - Xinwei Li
- Key Laboratory of Zoonoses Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, Jilin Province, 130062, China
| | - Bo Jin
- Key Laboratory of Zoonoses Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, Jilin Province, 130062, China
| | - Juan J Loor
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - Ahmad Aboragah
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - Lingxue Ju
- Key Laboratory of Zoonoses Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, Jilin Province, 130062, China
| | - Zhiyuan Fang
- Key Laboratory of Zoonoses Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, Jilin Province, 130062, China
| | - Hao Yu
- Key Laboratory of Zoonoses Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, Jilin Province, 130062, China
| | - Meng Chen
- Key Laboratory of Zoonoses Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, Jilin Province, 130062, China
| | - Yiwei Zhu
- Key Laboratory of Zoonoses Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, Jilin Province, 130062, China
| | - Hongsheng Ouyang
- Jilin Provincial Key Laboratory of Animal Embryo Engineering, College of Animal Sciences, Jilin University, 5333 Xi'an Road, Changchun, 130062, China
| | - Yuxiang Song
- Key Laboratory of Zoonoses Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, Jilin Province, 130062, China
| | - Zhe Wang
- Key Laboratory of Zoonoses Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, Jilin Province, 130062, China
| | - Xiliang Du
- Key Laboratory of Zoonoses Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, Jilin Province, 130062, China.
| | - Guowen Liu
- Key Laboratory of Zoonoses Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xi'an Road, Changchun, Jilin Province, 130062, China.
| |
Collapse
|
26
|
Yeom H, Hwang SH, Han BI, Lee M. Differential Sensitivity of Wild-Type and BRAF-Mutated Cells to Combined BRAF and Autophagy Inhibition. Biomol Ther (Seoul) 2021; 29:434-444. [PMID: 33551379 PMCID: PMC8255135 DOI: 10.4062/biomolther.2020.203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/15/2021] [Accepted: 01/19/2021] [Indexed: 01/07/2023] Open
Abstract
BRAF inhibitors are insufficient monotherapies for BRAF-mutated cancer; therefore, we investigated which inhibitory pathway would yield the most effective therapeutic approach when targeted in combination with BRAF inhibition. The oncogenic BRAF inhibitor, PLX4720, increased basal autophagic flux in BRAF-mutated cells compared to wild-type (WT) BRAF cells. Interestingly, early autophagy inhibition improved the effectiveness of PLX4720 regardless of BRAF mutation, whereas late autophagy inhibition did not. Although ATG5 knockout led to PLX4720 resistance in both WT and BRAF-mutated cells, the MEK inhibitor trametinib exhibited a synergistic effect on PLX4720 sensitivity in WT BRAF cells but not in BRAF-mutated cells. Conversely, the prolonged inhibition of endoplasmic reticulum (ER) stress reduced basal autophagy in BRAF-mutated cells, thereby increasing PLX4720 sensitivity. Taken together, our results suggest that the combined inhibition of ER stress and BRAF may simultaneously suppress both pro-survival ER stress and autophagy, and may therefore be suitable for treatment of BRAF-mutated tumors whose autophagy is increased by chronic ER stress. Similarly, for WT BRAF tumors, therapies targeting MEK signaling may be a more effective treatment strategy. Together, this study presents a rational combination treatment strategy to improve the efficacy of BRAF inhibitors depending on BRAF mutation status.
Collapse
Affiliation(s)
- Hojin Yeom
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Sung-Hee Hwang
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Byeal-I Han
- Institute for New Drug Development, Incheon National University, Incheon 22012, Republic of Korea
| | - Michael Lee
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea.,Institute for New Drug Development, Incheon National University, Incheon 22012, Republic of Korea
| |
Collapse
|
27
|
Sannino S, Yates ME, Schurdak ME, Oesterreich S, Lee AV, Wipf P, Brodsky JL. Unique integrated stress response sensors regulate cancer cell susceptibility when Hsp70 activity is compromised. eLife 2021; 10:64977. [PMID: 34180400 PMCID: PMC8275131 DOI: 10.7554/elife.64977] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 06/27/2021] [Indexed: 12/11/2022] Open
Abstract
Molecular chaperones, such as Hsp70, prevent proteotoxicity and maintain homeostasis. This is perhaps most evident in cancer cells, which overexpress Hsp70 and thrive even when harboring high levels of misfolded proteins. To define the response to proteotoxic challenges, we examined adaptive responses in breast cancer cells in the presence of an Hsp70 inhibitor. We discovered that the cells bin into distinct classes based on inhibitor sensitivity. Strikingly, the most resistant cells have higher autophagy levels, and autophagy was maximally activated only in resistant cells upon Hsp70 inhibition. In turn, resistance to compromised Hsp70 function required the integrated stress response transducer, GCN2, which is commonly associated with amino acid starvation. In contrast, sensitive cells succumbed to Hsp70 inhibition by activating PERK. These data reveal an unexpected route through which breast cancer cells adapt to proteotoxic insults and position GCN2 and autophagy as complementary mechanisms to ensure survival when proteostasis is compromised.
Collapse
Affiliation(s)
- Sara Sannino
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, United States
| | - Megan E Yates
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Magee-Women Research Institute, Pittsburgh, United States.,Integrative Systems Biology Program, University of Pittsburgh, Pittsburgh, United States.,Medical Scientist Training Program, University of Pittsburgh School of Medicine, Pittsburgh, United States
| | - Mark E Schurdak
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, United States.,University of Pittsburgh Drug Discovery Institute, Pittsburgh, United States
| | - Steffi Oesterreich
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Magee-Women Research Institute, Pittsburgh, United States.,Integrative Systems Biology Program, University of Pittsburgh, Pittsburgh, United States.,Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, United States
| | - Adrian V Lee
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Magee-Women Research Institute, Pittsburgh, United States.,Integrative Systems Biology Program, University of Pittsburgh, Pittsburgh, United States.,Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, United States
| | - Peter Wipf
- Department of Chemistry, University of Pittsburgh, Pittsburgh, United States
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, United States
| |
Collapse
|
28
|
Zhu Q, Liu Z, Wang Y, Song E, Song Y. Endoplasmic reticulum stress manipulates autophagic response that antagonizes polybrominated diphenyl ethers quinone induced cytotoxicity in microglial BV2 cells. JOURNAL OF HAZARDOUS MATERIALS 2021; 411:124958. [PMID: 33450633 DOI: 10.1016/j.jhazmat.2020.124958] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/13/2020] [Accepted: 12/22/2020] [Indexed: 06/12/2023]
Abstract
Polybrominated diphenyl ethers (PBDEs) were widely used as flame retardants. Previously, we reported that their quinone-type metabolite (PBDEQ) induced selective autophagy, but its biological consequences remain obscure. Here, we illustrated the possible link of PBDEQ-induced autophagy with endoplasmic reticulum (ER) stress and cytotoxicity in microglial BV2 cells. We found PBDEQ increased the formation of autophagosomes, promoted autophagic degradation, suggesting an improved autophagy flux in BV2 cells. Interestingly, both pharmacologic autophagy inhibitors and autophagy-related 5 gene small interfering RNA (ATG5 siRNA) aggravated the cytotoxicity of PBDEQ, suggesting the antagonizing role of autophagy. PBDEQ induced ER stress and activated protein kinase R-like ER kinase (PERK)-eukaryotic translation initiation factor 2α (eIF2α)-activating transcription factor 4 (ATF4)-C/EBP homologous protein (CHOP) axis of classic unfolded protein response (UPR) pathway, whilst ER stress inhibitor blocked PBDEQ-induced autophagy. Moreover, N-acetyl-L-cysteine (NAC) alleviated PBDEQ-induced activation of ER stress and autophagy, suggesting reactive oxygen species (ROS) were involved in regulating PBDEQ-induced ER stress and autophagy. Taken together, our results demonstrate a new mechanism of PBDEQ-associated toxicity.
Collapse
Affiliation(s)
- Qiushuang Zhu
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing 400715, China
| | - Zixuan Liu
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing 400715, China
| | - Yuting Wang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing 400715, China
| | - Erqun Song
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing 400715, China
| | - Yang Song
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing 400715, China; State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China.
| |
Collapse
|
29
|
Jiang Y, Fang B, Xu B. PCR array analysis identified hyperproliferation but not autophagy or apoptosis in fibrous epulis. J Clin Lab Anal 2021; 35:e23784. [PMID: 33934404 PMCID: PMC8183928 DOI: 10.1002/jcla.23784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/08/2021] [Accepted: 03/11/2021] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND The pathogenesis of fibrous epulis is still quite unclear. Our recent genome-wide RNA sequencing analysis revealed that in fibrous epulis, RAS-PI3K-AKT-NF-κB pathway regulates the expression of Bcl-2 family and IAP family genes, leading to increased proliferation and the inhibition of apoptosis. The PI3K/AKT signaling pathway can promote autophagy in human gingival fibroblasts; therefore, the purpose of the present study was to identify whether autophagy is involved in the pathogenesis of fibrous epulis. METHODS Differentially expressed genes (DEGs) between fibrous epulis lesions and normal gingival tissues were identified using the PCR array. The expression levels of eighteen autophagy-related (ATG) family genes, twelve B-cell lymphoma 2 (Bcl-2) family genes, and eleven cysteine-dependent aspartate-directed protease (caspase) family genes were validated using quantitative real-time PCR (qRT-PCR). Autophagy induction was determined by measuring microtubule-associated protein light chain 3 (LC3) conversion (LC3-I to LC3-II) by immunoblot analysis. RESULTS The PCR array identified six upregulated genes, whereas no genes were expressed at significantly lower levels. The upregulated genes were BCL2, BCL2L1, CXCR4, HSP90AA1, HSPA8, and IGF1, which all belong to the "regulation of autophagy" group but not the "autophagy machinery components" group. qRT-PCR verified that the expression levels of BCL2, BCL2L1 (also known as BCL-XL), and BCL2L2 (also known as BCL-W) were significantly increased in fibrous epulis. No LC3-I to LC3-II conversion was observed. CONCLUSIONS The present study reveals that in fibrous epulis, Bcl-2 and Bcl-xL coordinately mediate gingival cell escape from apoptosis, leading to uncontrolled proliferation. Moreover, ATG family genes are not activated, and autophagy is not involved in this process.
Collapse
Affiliation(s)
- Yangyang Jiang
- Department of OrthodonticsShanghai Ninth People’s HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Bing Fang
- Department of OrthodonticsShanghai Ninth People’s HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Bin Xu
- Department of StomatologyHwa Mei HospitalUniversity of Chinese Academy of SciencesNingboChina
| |
Collapse
|
30
|
Zhang X, Lv S, Zhang W, Jia Q, Wang L, Ding Y, Yuan P, Zhu Y, Liu L, Li Y, Zhang J. Shenmai injection improves doxorubicin cardiotoxicity via miR-30a/Beclin 1. Biomed Pharmacother 2021; 139:111582. [PMID: 33895525 DOI: 10.1016/j.biopha.2021.111582] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 04/01/2021] [Accepted: 04/02/2021] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Shenmai Injection (SMI) has been widely used in the treatment of cardiovascular diseases and can reduce side effects when combined with chemotherapy drugs. However, the potential protective mechanism of SMI on the cardiotoxicity caused by anthracyclines has not been clear. METHODS We used network pharmacology methods to collect the compound components in SMI and myocardial injury targets, constructed a 'drug-disease' target interaction network relationship diagram, and screened the core targets to predict the potential mechanism of SMI in treating cardiotoxicity of anthracyclines. In addition, the rat model of doxorubicin cardiotoxicity was induced by injecting doxorubicin through the tail vein. The rats were randomized in the model group, miR-30a agomir group, SMI low-dose group, SMI high-dose group,and the control group. The cardiac ultrasound was used to evaluate the structure and function of the rat heart. HE staining was used to observe the pathological changes of the rat myocardium. Transmission electron microscopy was used to observe myocardial autophagosomes. The expression of miR-30a and Beclin 1 mRNA in the rat myocardium was detected by RT-qPCR. Western Blot detected the expression of LC3-II/LC3-I and p62 protein. RESULTS The network pharmacological analysis found that SMI could act synergistically through multiple targets and multiple pathways, which might exert a myocardial protective effect through PI3K-Akt signaling pathways and cancer microRNAs. In vivo, compared with the control group, the treatment group could improve the cardiac structure and function, and reduce myocardial pathological damage and the number of autophagosomes. The expression of miR-30a in the myocardium of rats in miR-30a agomir group and SMI group increased (P < 0.01),Beclin 1 mRNA was decreased (P < 0.01),LC3-Ⅱ/LC3-I protein was decreased (P < 0.01 or P < 0.05),and p62 protein was increased (P < 0.01 or P < 0.05). CONCLUSIONS SMI has the characteristics of multi-component, multi-target, and multi-pathway. It can inhibit myocardial excessive autophagy by regulating the expression of miR-30a/Beclin 1 and alleviate the myocardial injury induced by doxorubicin.
Collapse
Affiliation(s)
- Xiaonan Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Shichao Lv
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Tianjin Key Laboratory of Traditional Research of TCM Prescription and Syndrome, Tianjin 300193, China
| | - Wanqin Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Qiujin Jia
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Lirong Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Yuejia Ding
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Peng Yuan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Yaping Zhu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Longtao Liu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China.
| | - Yanyang Li
- Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China.
| | - Junping Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.
| |
Collapse
|
31
|
Chen S, Zhang X, Nie Y, Li H, Chen W, Lin W, Chen F, Xie Q. African Swine Fever Virus Protein E199L Promotes Cell Autophagy through the Interaction of PYCR2. Virol Sin 2021; 36:196-206. [PMID: 33830435 PMCID: PMC8027715 DOI: 10.1007/s12250-021-00375-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 02/01/2021] [Indexed: 11/27/2022] Open
Abstract
African swine fever virus (ASFV), as a member of the large DNA viruses, may regulate autophagy and apoptosis by inhibiting programmed cell death. However, the function of ASFV proteins has not been fully elucidated, especially the role of autophagy in ASFV infection. One of three Pyrroline-5-carboxylate reductases (PYCR), is primarily involved in conversion of glutamate to proline. Previous studies have shown that depletion of PYCR2 was related to the induction of autophagy. In the present study, we found for the first time that ASFV E199L protein induced a complete autophagy process in Vero and HEK-293T cells. Through co-immunoprecipitation coupled with mass spectrometry (CoIP-MS) analysis, we firstly identified that E199L interact with PYCR2 in vitro. Importantly, our work provides evidence that E199L down-regulated the expression of PYCR2, resulting in autophagy activation. Overall, our results demonstrate that ASFV E199L protein induces complete autophagy through interaction with PYCR2 and down-regulate the expression level of PYCR2, which provide a valuable reference for the role of autophagy during ASFV infection and contribute to the functional clues of PYCR2.
Collapse
Affiliation(s)
- Sheng Chen
- College of Animal Science, South China Agricultural University & Lingnan Guangdong Laboratory of Modern Agriculture & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Guangzhou, 510642, China.,South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou, 510642, China.,Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangzhou, 510642, China
| | - Xinheng Zhang
- College of Animal Science, South China Agricultural University & Lingnan Guangdong Laboratory of Modern Agriculture & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Guangzhou, 510642, China.,South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou, 510642, China.,Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangzhou, 510642, China
| | - Yu Nie
- College of Animal Science, South China Agricultural University & Lingnan Guangdong Laboratory of Modern Agriculture & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Guangzhou, 510642, China.,South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou, 510642, China.,Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangzhou, 510642, China
| | - Hongxin Li
- College of Animal Science, South China Agricultural University & Lingnan Guangdong Laboratory of Modern Agriculture & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Guangzhou, 510642, China.,South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou, 510642, China.,Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangzhou, 510642, China
| | - Weiguo Chen
- College of Animal Science, South China Agricultural University & Lingnan Guangdong Laboratory of Modern Agriculture & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Guangzhou, 510642, China.,South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou, 510642, China.,Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangzhou, 510642, China
| | - Wencheng Lin
- College of Animal Science, South China Agricultural University & Lingnan Guangdong Laboratory of Modern Agriculture & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Guangzhou, 510642, China.,South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou, 510642, China.,Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangzhou, 510642, China
| | - Feng Chen
- College of Animal Science, South China Agricultural University & Lingnan Guangdong Laboratory of Modern Agriculture & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Guangzhou, 510642, China.,South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou, 510642, China.,Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangzhou, 510642, China
| | - Qingmei Xie
- College of Animal Science, South China Agricultural University & Lingnan Guangdong Laboratory of Modern Agriculture & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Guangzhou, 510642, China. .,South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, Guangzhou, 510642, China. .,Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangzhou, 510642, China.
| |
Collapse
|
32
|
Wu JH, Tung SY, Ho CC, Su LH, Gan SW, Liao JY, Cho CC, Lin BC, Chiu PW, Pan YJ, Kao YY, Liu YC, Sun CH. A myeloid leukemia factor homolog involved in encystation-induced protein metabolism in Giardia lamblia. Biochim Biophys Acta Gen Subj 2021; 1865:129859. [PMID: 33581251 DOI: 10.1016/j.bbagen.2021.129859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/14/2021] [Accepted: 01/25/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Giardia lamblia differentiates into resistant cysts as an established model for dormancy. Myeloid leukemia factor (MLF) proteins are important regulators of cell differentiation. Giardia possesses a MLF homolog which was up-regulated during encystation and localized to unknown cytosolic vesicles named MLF vesicles (MLFVs). METHODS We used double staining for visualization of potential factors with role in protein metabolism pathway and a strategy that employed a deletion mutant, CDK2m3, to test the protein degradation pathway. We also explored whether autophagy or proteasomal degradation are regulators of Giardia encystation by treatment with MG132, rapamycin, or chloroquine. RESULTS Double staining of MLF and ISCU or CWP1 revealed no overlap between their vesicles. The aberrant CDK2m3 colocalized with MLFVs and formed complexes with MLF. MG132 increased the number of CDK2m3-localized vesicles and its protein level. We further found that MLF colocalized and interacted with a FYVE protein and an ATG8-like (ATG8L) protein, which were up-regulated during encystation and their expression induced Giardia encystation. The addition of MG132, rapamycin, or chloroquine, increased their levels and the number of their vesicles, and inhibited the cyst formation. MLF and FYVE were detected in exosomes released from culture. CONCLUSIONS The MLFVs are not mitosomes or encystation-specific vesicles, but are related with degradative pathway for CDK2m3. MLF, FYVE, and ATG8L play a positive role in encystation and function in protein clearance pathway, which is important for encystation and coordinated with Exosomes. GENERAL SIGNIFICANCE MLF, FYVE, and ATG8L may be involved an encystation-induced protein metabolism during Giardia differentiation.
Collapse
Affiliation(s)
- Jui-Hsuan Wu
- Department of Tropical Medicine and Parasitology, College of Medicine, National Taiwan University, Taipei 100, Taiwan, ROC
| | - Szu-Yu Tung
- Department of Tropical Medicine and Parasitology, College of Medicine, National Taiwan University, Taipei 100, Taiwan, ROC
| | - Chun-Che Ho
- Department of Tropical Medicine and Parasitology, College of Medicine, National Taiwan University, Taipei 100, Taiwan, ROC
| | - Li-Hsin Su
- Department of Tropical Medicine and Parasitology, College of Medicine, National Taiwan University, Taipei 100, Taiwan, ROC
| | - Soo-Wah Gan
- Department of Tropical Medicine and Parasitology, College of Medicine, National Taiwan University, Taipei 100, Taiwan, ROC
| | - Jo-Yu Liao
- Department of Tropical Medicine and Parasitology, College of Medicine, National Taiwan University, Taipei 100, Taiwan, ROC
| | - Chao-Cheng Cho
- Department of Tropical Medicine and Parasitology, College of Medicine, National Taiwan University, Taipei 100, Taiwan, ROC
| | - Bo-Chi Lin
- Department of Tropical Medicine and Parasitology, College of Medicine, National Taiwan University, Taipei 100, Taiwan, ROC
| | - Pei-Wei Chiu
- Department of Tropical Medicine and Parasitology, College of Medicine, National Taiwan University, Taipei 100, Taiwan, ROC
| | - Yu-Jiao Pan
- Department of Tropical Medicine and Parasitology, College of Medicine, National Taiwan University, Taipei 100, Taiwan, ROC
| | - Yu-Yun Kao
- Department of Tropical Medicine and Parasitology, College of Medicine, National Taiwan University, Taipei 100, Taiwan, ROC
| | - Yu-Chen Liu
- Department of Tropical Medicine and Parasitology, College of Medicine, National Taiwan University, Taipei 100, Taiwan, ROC
| | - Chin-Hung Sun
- Department of Tropical Medicine and Parasitology, College of Medicine, National Taiwan University, Taipei 100, Taiwan, ROC.
| |
Collapse
|
33
|
The apoptotic and autophagic effects of cast Au-Pt, and differently manufactured Co-Cr and cp-Ti on three-dimensional oral mucosal model. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 120:111672. [PMID: 33545837 DOI: 10.1016/j.msec.2020.111672] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 10/19/2020] [Accepted: 10/20/2020] [Indexed: 12/26/2022]
Abstract
The application of digitally manufactured dental metals has aroused the attention on their biocompatibilities. Three-dimensional oral mucosal model (3D OMM) would provide excellent assessments to the biocompatibility. In the current study, we set to measure metal ion release levels in the extracts of cast gold-platinum alloy (Au-Pt), differently manufactured cobalt-chromium alloy (Co-Cr) and commercially pure titanium (cp-Ti). We further tested two scaffold materials of 3D OMM to determine the better one for the succedent work. Lastly, we evaluated the apoptotic and autophagic effects of cast Au-Pt, and differently manufactured Co-Cr and cp-Ti on mucosal cells based on 3D OMM. We found that, in the construction of 3D OMM, Matrigel showed better performance than bovine acellular dermal matrix. Thus, Matrigel was chosen to construct the 3D OMM in the succedent studies. The results of ion release and biological assessments showed that, firstly, cast Au-Pt and cp-Ti triggered less early apoptotic cells and ion release than cast Co-Cr, implying better chemical stability and biocompatibility of them; secondly, digitally manufactured (including CAD/CAM milling and SLM) Co-Cr showed significantly lower ion release levels and lesser early apoptotic effects on 3D OMM as compared to the cast one. Although cast cp-Ti released much more ions than CAD/CAM milling one, manufacturing methods had no impact on apoptotic effect of cp-Ti. Therefore, we believe that digital methods possess same or even better chemical stability and biocompatibility than conventional casting one. Thirdly, although increased autophagic levels are observed in all test groups, so far there is no evidence that the test metals trigger different levels of autophagy as compared to each other. In addition, correlation analysis indicates that Co, W, and Mn appear to be the potential inducements for the apoptotic and autophagic effects of Co-Cr.
Collapse
|
34
|
Fang S, Wan X, Zou X, Sun S, Hao X, Liang C, Zhang Z, Zhang F, Sun B, Li H, Yu B. Arsenic trioxide induces macrophage autophagy and atheroprotection by regulating ROS-dependent TFEB nuclear translocation and AKT/mTOR pathway. Cell Death Dis 2021; 12:88. [PMID: 33462182 PMCID: PMC7814005 DOI: 10.1038/s41419-020-03357-1] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 01/30/2023]
Abstract
Inducing autophagy and inhibiting apoptosis may provide a therapeutic treatment for atherosclerosis (AS). For the treatment of progressive AS, arsenic trioxide (ATO) has been used to coat vascular stents. However, the effect of ATO on autophagy of macrophages is still unknown. Therefore, the aims of this study were to characterize the effects and the mechanism of actions of ATO on autophagy in macrophages. Our results showed that ATO-induced activation of autophagy was an earlier event than ATO-induced inhibition of the expression of apoptosis markers in macrophages and foam cells. Nuclear transcription factor EB (TFEB) prevents atherosclerosis by activating macrophage autophagy and promoting lysosomal biogenesis. Here, we report that ATO triggered the nuclear translocation of TFEB, which in turn promoted autophagy and autophagosome-lysosome fusion. Both the latter events were prevented by TFEB knockdown. Moreover, ATO decreased the p-AKT and p-mTOR in the PI3K/AKT/mTOR signaling pathway, thus inducing autophagy. Correspondingly, treatment with the autophagy inhibitor 3-methyladenine (3-MA) abolished the autophagy-inducing effects of ATO. Meanwhile, PI3K inhibitor (LY294002) and mTOR inhibitor (rapamycin) cooperated with ATO to induce autophagy. Furthermore, reactive oxygen species (ROS) were generated in macrophages after treatment with ATO. The ROS scavenger N-acetyl-1-cysteine (NAC) abolished ATO-induced nuclear translocation of TFEB, as well as changes in key molecules of the AKT/mTOR signaling pathway and downstream autophagy. More importantly, ATO promoted autophagy in the aorta of ApoE-/- mice and reduced atherosclerotic lesions in early AS, which were reversed by 3-MA treatment. In summary, our data indicated that ATO promoted ROS induction, which resulted in nuclear translocation of TFEB and inhibition of the PI3K/AKT/mTOR pathway. These actions ultimately promoted macrophage autophagy and reduced atherosclerotic lesions at early stages. These findings may provide a new perspective for the clinical treatment of early-stage atherosclerosis and should be further studied.
Collapse
Affiliation(s)
- Shaohong Fang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China
| | - Xin Wan
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, China
| | - Xiaoyi Zou
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, China
| | - Song Sun
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, China
| | - Xinran Hao
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, China
| | - Chenchen Liang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China
| | - Zhenming Zhang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China
| | - Fangni Zhang
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, China
| | - Bo Sun
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, China
| | - Hulun Li
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China.
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, China.
| | - Bo Yu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China.
| |
Collapse
|
35
|
Sarmah DT, Bairagi N, Chatterjee S. Tracing the footsteps of autophagy in computational biology. Brief Bioinform 2020; 22:5985288. [PMID: 33201177 PMCID: PMC8293817 DOI: 10.1093/bib/bbaa286] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 09/29/2020] [Accepted: 09/30/2020] [Indexed: 12/11/2022] Open
Abstract
Autophagy plays a crucial role in maintaining cellular homeostasis through the degradation of unwanted materials like damaged mitochondria and misfolded proteins. However, the contribution of autophagy toward a healthy cell environment is not only limited to the cleaning process. It also assists in protein synthesis when the system lacks the amino acids’ inflow from the extracellular environment due to diet consumptions. Reduction in the autophagy process is associated with diseases like cancer, diabetes, non-alcoholic steatohepatitis, etc., while uncontrolled autophagy may facilitate cell death. We need a better understanding of the autophagy processes and their regulatory mechanisms at various levels (molecules, cells, tissues). This demands a thorough understanding of the system with the help of mathematical and computational tools. The present review illuminates how systems biology approaches are being used for the study of the autophagy process. A comprehensive insight is provided on the application of computational methods involving mathematical modeling and network analysis in the autophagy process. Various mathematical models based on the system of differential equations for studying autophagy are covered here. We have also highlighted the significance of network analysis and machine learning in capturing the core regulatory machinery governing the autophagy process. We explored the available autophagic databases and related resources along with their attributes that are useful in investigating autophagy through computational methods. We conclude the article addressing the potential future perspective in this area, which might provide a more in-depth insight into the dynamics of autophagy.
Collapse
Affiliation(s)
| | - Nandadulal Bairagi
- Centre for Mathematical Biology and Ecology, Department of Mathematics, Jadavpur University, Kolkata, India
| | - Samrat Chatterjee
- Translational Health Science and Technology Institute, Faridabad, India
| |
Collapse
|
36
|
Machado-Oliveira G, Ramos C, Marques ARA, Vieira OV. Cell Senescence, Multiple Organelle Dysfunction and Atherosclerosis. Cells 2020; 9:E2146. [PMID: 32977446 PMCID: PMC7598292 DOI: 10.3390/cells9102146] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/19/2020] [Accepted: 09/20/2020] [Indexed: 01/10/2023] Open
Abstract
Atherosclerosis is an age-related disorder associated with long-term exposure to cardiovascular risk factors. The asymptomatic progression of atherosclerotic plaques leads to major cardiovascular diseases (CVD), including acute myocardial infarctions or cerebral ischemic strokes in some cases. Senescence, a biological process associated with progressive structural and functional deterioration of cells, tissues and organs, is intricately linked to age-related diseases. Cell senescence involves coordinated modifications in cellular compartments and has been demonstrated to contribute to different stages of atheroma development. Senescence-based therapeutic strategies are currently being pursued to treat and prevent CVD in humans in the near-future. In addition, distinct experimental settings allowed researchers to unravel potential approaches to regulate anti-apoptotic pathways, facilitate excessive senescent cell clearance and eventually reverse atherogenesis to improve cardiovascular function. However, a deeper knowledge is required to fully understand cellular senescence, to clarify senescence and atherogenesis intertwining, allowing researchers to establish more effective treatments and to reduce the cardiovascular disorders' burden. Here, we present an objective review of the key senescence-related alterations of the major intracellular organelles and analyze the role of relevant cell types for senescence and atherogenesis. In this context, we provide an updated analysis of therapeutic approaches, including clinically relevant experiments using senolytic drugs to counteract atherosclerosis.
Collapse
Affiliation(s)
- Gisela Machado-Oliveira
- CEDOC, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (C.R.); (A.R.A.M.)
| | | | | | - Otília V. Vieira
- CEDOC, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisboa, Portugal; (C.R.); (A.R.A.M.)
| |
Collapse
|
37
|
Ma Z, Li J, Lin K, Ramachandran M, Zhang D, Showalter M, De Souza C, Lindstrom A, Solano LN, Jia B, Urayama S, Duan Y, Fiehn O, Lin TY, Li M, Li Y. Pharmacophore hybridisation and nanoscale assembly to discover self-delivering lysosomotropic new-chemical entities for cancer therapy. Nat Commun 2020; 11:4615. [PMID: 32934241 PMCID: PMC7493904 DOI: 10.1038/s41467-020-18399-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 08/18/2020] [Indexed: 01/01/2023] Open
Abstract
Integration of the unique advantages of the fields of drug discovery and drug delivery is invaluable for the advancement of drug development. Here we propose a self-delivering one-component new-chemical-entity nanomedicine (ONN) strategy to improve cancer therapy through incorporation of the self-assembly principle into drug design. A lysosomotropic detergent (MSDH) and an autophagy inhibitor (Lys05) are hybridised to develop bisaminoquinoline derivatives that can intrinsically form nanoassemblies. The selected BAQ12 and BAQ13 ONNs are highly effective in inducing lysosomal disruption, lysosomal dysfunction and autophagy blockade and exhibit 30-fold higher antiproliferative activity than hydroxychloroquine used in clinical trials. These single-drug nanoparticles demonstrate excellent pharmacokinetic and toxicological profiles and dramatic antitumour efficacy in vivo. In addition, they are able to encapsulate and deliver additional drugs to tumour sites and are thus promising agents for autophagy inhibition-based combination therapy. Given their transdisciplinary advantages, these BAQ ONNs have enormous potential to improve cancer therapy.
Collapse
Affiliation(s)
- Zhao Ma
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA, 95817, USA
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmacy, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Jin Li
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA, 95817, USA
| | - Kai Lin
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA, 95817, USA
| | - Mythili Ramachandran
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA, 95817, USA
| | - Dalin Zhang
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA, 95817, USA
| | - Megan Showalter
- West Coast Metabolomics Center, University of California Davis, Davis, CA, 95616, USA
| | - Cristabelle De Souza
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA, 95817, USA
| | - Aaron Lindstrom
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA, 95817, USA
| | - Lucas N Solano
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA, 95817, USA
| | - Bei Jia
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA, 95817, USA
| | - Shiro Urayama
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Yuyou Duan
- Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China
| | - Oliver Fiehn
- West Coast Metabolomics Center, University of California Davis, Davis, CA, 95616, USA
| | - Tzu-Yin Lin
- Division of Hematology and Oncology, Department of Internal Medicine, University of California Davis, Sacramento, CA, 95817, USA
| | - Minyong Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmacy, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- State Key Laboratory of Microbial Technology, Shandong University, Jinan, 250100, Shandong, China
| | - Yuanpei Li
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA, 95817, USA.
| |
Collapse
|
38
|
López-Pérez Ó, Badiola JJ, Bolea R, Ferrer I, Llorens F, Martín-Burriel I. An Update on Autophagy in Prion Diseases. Front Bioeng Biotechnol 2020; 8:975. [PMID: 32984276 PMCID: PMC7481332 DOI: 10.3389/fbioe.2020.00975] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 07/27/2020] [Indexed: 12/18/2022] Open
Abstract
Autophagy is a dynamic intracellular mechanism involved in protein and organelle turnover through lysosomal degradation. When properly regulated, autophagy supports normal cellular and developmental processes, whereas defects in autophagic degradation have been associated with several pathologies, including prion diseases. Prion diseases, or transmissible spongiform encephalopathies (TSE), are a group of fatal neurodegenerative disorders characterized by the accumulation of the pathological misfolded isoform (PrPSc) of the physiological cellular prion protein (PrPc) in the central nervous system. Autophagic vacuoles have been described in experimental models of TSE and in the natural disease in humans. The precise connection of this process with prion-related neuropathology, or even whether autophagy is completely beneficial or pathogenic during neurodegeneration, is poorly understood. Thus, the biological role of autophagy in these diseases is still open to debate. During the last years, researchers have used a wide range of morphological, genetic and biochemical methods to monitor and manipulate the autophagic pathway and thus determine the specific role of this process in TSE. It has been suggested that PrPc could play a crucial role in modulating the autophagic pathway in neuronal cells, and the presence of abnormal autophagic activity has been frequently observed in several models of TSE both in vitro and in vivo, as well as in human prion diseases. Altogether, these findings suggest that autophagy is implicated in prion neuropathology and points to an impairment or failure of the process, potentially contributing to the pathogenesis of the disease. Additionally, autophagy is now emerging as a host defense response in controlling prion infection that plays a protective role by facilitating the clearance of aggregation-prone proteins accumulated within neurons. Since autophagy is one of the pathways of PrPSc degradation, and drug-induced stimulation of autophagic flux (the dynamic process of autophagic degradation activity) produces anti-prion effects, new treatments based on its activation have been tested to develop therapeutic strategies for prion diseases. In this review, we summarize previous and recent findings concerning the role of autophagy in TSE.
Collapse
Affiliation(s)
- Óscar López-Pérez
- Laboratorio de Genética Bioquímica (LAGENBIO), Instituto Agroalimentario de Aragón-IA2, Instituto de Investigación Sanitaria Aragón-IISA, Universidad de Zaragoza, Zaragoza, Spain.,Centro de Encefalopatías y Enfermedades Transmisibles Emergentes (CEETE), Instituto Agroalimentario de Aragón-IA2, Instituto de Investigación Sanitaria Aragón-IISA, Universidad de Zaragoza, Zaragoza, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto Carlos III, L'Hospitalet de Llobregat, Barcelona, Spain.,Instituto de Investigación Biomédica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Juan José Badiola
- Centro de Encefalopatías y Enfermedades Transmisibles Emergentes (CEETE), Instituto Agroalimentario de Aragón-IA2, Instituto de Investigación Sanitaria Aragón-IISA, Universidad de Zaragoza, Zaragoza, Spain
| | - Rosa Bolea
- Centro de Encefalopatías y Enfermedades Transmisibles Emergentes (CEETE), Instituto Agroalimentario de Aragón-IA2, Instituto de Investigación Sanitaria Aragón-IISA, Universidad de Zaragoza, Zaragoza, Spain
| | - Isidro Ferrer
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto Carlos III, L'Hospitalet de Llobregat, Barcelona, Spain.,Instituto de Investigación Biomédica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.,Departamento de Patología y Terapéutica Experimental, Universidad de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Franc Llorens
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto Carlos III, L'Hospitalet de Llobregat, Barcelona, Spain.,Instituto de Investigación Biomédica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.,Department of Neurology, Clinical Dementia Center and National Reference Center for CJD Surveillance, University Medical School, Göttingen, Germany
| | - Inmaculada Martín-Burriel
- Laboratorio de Genética Bioquímica (LAGENBIO), Instituto Agroalimentario de Aragón-IA2, Instituto de Investigación Sanitaria Aragón-IISA, Universidad de Zaragoza, Zaragoza, Spain.,Centro de Encefalopatías y Enfermedades Transmisibles Emergentes (CEETE), Instituto Agroalimentario de Aragón-IA2, Instituto de Investigación Sanitaria Aragón-IISA, Universidad de Zaragoza, Zaragoza, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto Carlos III, Zaragoza, Spain
| |
Collapse
|
39
|
Yagisawa Y, Suita K, Ohnuki Y, Ishikawa M, Mototani Y, Ito A, Matsuo I, Hayakawa Y, Nariyama M, Umeki D, Saeki Y, Amitani Y, Nakamura Y, Tomonari H, Okumura S. Effects of occlusal disharmony on cardiac fibrosis, myocyte apoptosis and myocyte oxidative DNA damage in mice. PLoS One 2020; 15:e0236547. [PMID: 32716920 PMCID: PMC7384634 DOI: 10.1371/journal.pone.0236547] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 07/08/2020] [Indexed: 12/05/2022] Open
Abstract
Occlusal disharmony leads to morphological changes in the hippocampus and osteopenia of the lumbar vertebra and long bones in mice, and causes stress. Various types of stress are associated with increased incidence of cardiovascular disease, but the relationship between occlusal disharmony and cardiovascular disease remain poorly understood. Therefore, in this work, we examined the effects of occlusal disharmony on cardiac homeostasis in bite-opening (BO) mice, in which a 0.7 mm space was introduced by cementing a suitable applicance onto the mandibular incisior. We first examined the effects of BO on the level of serum corticosterone, a key biomarker for stress, and on heart rate variability at 14 days after BO treatment, compared with baseline. BO treatment increased serum corticosterone levels by approximately 3.6-fold and the low frequency/high frequency ratio, an index of sympathetic nervous activity, was significantly increased by approximately 4-fold by the BO treatment. We then examined the effects of BO treatment on cardiac homeostasis in mice treated or not treated with the non-selective β-blocker propranolol for 2 weeks. Cardiac function was significantly decreased in the BO group compared to the control group, but propranolol ameliorated the dysfunction. Cardiac fibrosis, myocyte apoptosis and myocyte oxidative DNA damage were significantly increased in the BO group, but propranolol blocked these changes. The BO-induced cardiac dysfunction was associated with increased phospholamban phosphorylation at threonine-17 and serine-16, as well as inhibition of Akt/mTOR signaling and autophagic flux. These data suggest that occlusal disharmony might affect cardiac homeostasis via alteration of the autonomic nervous system.
Collapse
Affiliation(s)
- Yuka Yagisawa
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Orthodontics, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Kenji Suita
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yoshiki Ohnuki
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Misao Ishikawa
- Department of Oral Anatomy, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yasumasa Mototani
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Aiko Ito
- Department of Orthodontics, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Ichiro Matsuo
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yoshio Hayakawa
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Dental Anesthesiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Megumi Nariyama
- Department of Pediatric Dentistry, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Daisuke Umeki
- Department of Orthodontics, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yasutake Saeki
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yasuharu Amitani
- Department of Mathematics, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yoshiki Nakamura
- Department of Orthodontics, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Hiroshi Tomonari
- Department of Orthodontics, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Satoshi Okumura
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| |
Collapse
|
40
|
Lee HJ, Kang SJ, Woo Y, Hahn TW, Ko HJ, Jung YJ. TLR7 Stimulation With Imiquimod Induces Selective Autophagy and Controls Mycobacterium tuberculosis Growth in Mouse Macrophages. Front Microbiol 2020; 11:1684. [PMID: 32765474 PMCID: PMC7380068 DOI: 10.3389/fmicb.2020.01684] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 06/29/2020] [Indexed: 12/16/2022] Open
Abstract
Autophagy is a lysosomal self-digestion pathway that maintains internal homeostasis inside cells and critical process by which the innate immune system eliminates intracellular bacteria. In this study, we showed that stimulation of toll-like receptor 7 (TLR7) with imiquimod (IMQ) triggered autophagic cell death in macrophages by enhancing the generation of reactive oxygen species (ROS) via the p38- or MEK/ERK1/2-mediated signaling pathway in the early phase. IMQ significantly increased mitochondrial ROS and targeted autophagosomes to the mitochondria. Stimulation of TLR7 with IMQ enhanced the expression of BNIP3, which was localized to mitochondria and interacted with beclin-1, leading to mitophagy. In addition, IMQ substantially induced NO production through the GSK-3β-mediated signaling pathway, which led to autophagy in the late stage. We further examined whether the induction of autophagy by IMQ effectively eliminated intracellular microbes. Macrophages were infected with a virulent Mycobacterium tuberculosis (Mtb) strain, H37Rv, and then treated with IMQ. IMQ suppressed intracellular Mtb growth by inducing autophagy in a dose-dependent manner and increased NO production. Inhibition of autophagy using 3-methyladenine (3-MA) prevented autophagosome formation and control of intracellular Mtb growth in macrophages. These findings revealed a novel mechanism by which IMQ induces selective autophagy to promote intracellular killing machinery against Mtb infection in macrophages.
Collapse
Affiliation(s)
- Hyo-Ji Lee
- Department of Biological Sciences, Kangwon National University, Chuncheon, South Korea.,Institute of Life Sciences, Kangwon National University, Chuncheon, South Korea
| | - Su-Jin Kang
- Department of Biological Sciences, Kangwon National University, Chuncheon, South Korea
| | - Yunseo Woo
- Department of Biological Sciences, Kangwon National University, Chuncheon, South Korea.,Institute of Life Sciences, Kangwon National University, Chuncheon, South Korea
| | - Tae-Wook Hahn
- College of Veterinary Medicine, Kangwon National University, Chuncheon, South Korea
| | - Hyun-Jeong Ko
- College of Pharmacy, Kangwon National University, Chuncheon, South Korea
| | - Yu-Jin Jung
- Department of Biological Sciences, Kangwon National University, Chuncheon, South Korea.,Institute of Life Sciences, Kangwon National University, Chuncheon, South Korea
| |
Collapse
|
41
|
Feng Q, Bian X, Liu X, Wang Y, Zhou H, Ma X, Quan C, Yao Y, Zheng Z. Intracellular expression of arginine deiminase activates the mitochondrial apoptosis pathway by inhibiting cytosolic ferritin and inducing chromatin autophagy. BMC Cancer 2020; 20:665. [PMID: 32677906 PMCID: PMC7367323 DOI: 10.1186/s12885-020-07133-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 07/02/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Based on its low toxicity, arginine starvation therapy has the potential to cure malignant tumors that cannot be treated surgically. The Arginine deiminase (ADI) gene has been identified to be an ideal cancer-suppressor gene. ADI expressed in the cytosol displays higher oncolytic efficiency than ADI-PEG20 (Pegylated Arginine Deiminase by PEG 20,000). However, it is still unknown whether cytosolic ADI has the same mechanism of action as ADI-PEG20 or other underlying cellular mechanisms. METHODS The interactions of ADI with other protein factors were screened by yeast hybrids, and verified by co-immunoprecipitation and immunofluorescent staining. The effect of ADI inhibiting the ferritin light-chain domain (FTL) in mitochondrial damage was evaluated by site-directed mutation and flow cytometry. Control of the mitochondrial apoptosis pathway was analyzed by Western Blotting and real-time PCR experiments. The effect of p53 expression on cancer cells death was assessed by siTP53 transfection. Chromatin autophagy was explored by immunofluorescent staining and Western Blotting. RESULTS ADI expressed in the cytosol inhibited the activity of cytosolic ferritin by interacting with FTL. The inactive mutant of ADI still induced apoptosis in certain cell lines of ASS- through mitochondrial damage. Arginine starvation also generated an increase in the expression of p53 and p53AIP1, which aggravated the cellular mitochondrial damage. Chromatin autophagy appeared at a later stage of arginine starvation. DNA damage occurred along with the entire arginine starvation process. Histone 3 (H3) was found in autophagosomes, which implies that cancer cells attempted to utilize the arginine present in histones to survive during arginine starvation. CONCLUSIONS Mitochondrial damage is the major mechanism of cell death induced by cytosolic ADI. The process of chromatophagy does not only stimulate cancer cells to utilize histone arginine but also speeds up cancer cell death at a later stage of arginine starvation.
Collapse
Affiliation(s)
- Qingyuan Feng
- Department of Otorhinolaryngology Head and Neck Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xuzhao Bian
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Xuan Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Ying Wang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Huiting Zhou
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Xiaojing Ma
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Chunju Quan
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Yi Yao
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zhongliang Zheng
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China.
| |
Collapse
|
42
|
Activation of autophagic flux via LKB1/AMPK/mTOR axis against xenoestrogen Bisphenol-A exposure in primary rat hepatocytes. Food Chem Toxicol 2020; 141:111314. [DOI: 10.1016/j.fct.2020.111314] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 03/07/2020] [Accepted: 03/30/2020] [Indexed: 12/25/2022]
|
43
|
Vicencio E, Cordero EM, Cortés BI, Palominos S, Parra P, Mella T, Henrríquez C, Salazar N, Monasterio G, Cafferata EA, Murgas P, Vernal R, Cortez C. Aggregatibacter Actinomycetemcomitans Induces Autophagy in Human Junctional Epithelium Keratinocytes. Cells 2020; 9:E1221. [PMID: 32423042 PMCID: PMC7290389 DOI: 10.3390/cells9051221] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 05/04/2020] [Accepted: 05/07/2020] [Indexed: 12/23/2022] Open
Abstract
The adverse environmental conditions found in the periodontium during periodontitis pathogenesis stimulate local autophagy responses, mainly due to a continuous inflammatory response against the dysbiotic subgingival microbiome. The junctional epithelium represents the main site of the initial interaction between the host and the dysbiotic biofilm. Here, we investigated the role of autophagy in junctional epithelium keratinocytes (JEKs) in response to Aggregatibacter actinomycetemcomitans or its purified lipopolysaccharides (LPS). Immunofluorescence confocal analysis revealed an extensive nuclear translocation of transcription factor EB (TFEB) and consequently, an increase in autophagy markers and LC3-turnover assessed by immunoblotting and qRT-PCR. Correspondingly, challenged JEKs showed a punctuate cytosolic profile of LC3 protein contrasting with the diffuse distribution observed in untreated controls. Three-dimensional reconstructions of confocal images displayed a close association between intracellular bacteria and LC3-positive vesicles. Similarly, a close association between autophagic vesicles and the protein p62 was observed in challenged JEKs, indicating that p62 is the main adapter protein recruited during A. actinomycetemcomitans infection. Finally, the pharmacological inhibition of autophagy significantly increased the number of bacteria-infected cells as well as their death, similar to treatment with LPS. Our results indicate that A. actinomycetemcomitans infection induces autophagy in JEKs, and this homeostatic process has a cytoprotective effect on the host cells during the early stages of infection.
Collapse
Affiliation(s)
- Emiliano Vicencio
- Center for Genomics and Bioinformatics, Faculty of Science, Universidad Mayor, Camino la Pirámide 5750, Huechuraba 8580745, Chile; (E.V.); (E.M.C.); (S.P.); (P.P.); (T.M.); (C.H.); (N.S.)
- Center for Integrative Biology, Faculty of Science, Universidad Mayor, Camino la Pirámide 5750, Huechuraba 8580745, Chile; (B.I.C.); (P.M.)
| | - Esteban M. Cordero
- Center for Genomics and Bioinformatics, Faculty of Science, Universidad Mayor, Camino la Pirámide 5750, Huechuraba 8580745, Chile; (E.V.); (E.M.C.); (S.P.); (P.P.); (T.M.); (C.H.); (N.S.)
- Parasitology Section, Instituto de Salud Pública de Chile, Avenida Marathon 1000, Ñuñoa 7780050, Chile
| | - Bastián I. Cortés
- Center for Integrative Biology, Faculty of Science, Universidad Mayor, Camino la Pirámide 5750, Huechuraba 8580745, Chile; (B.I.C.); (P.M.)
| | - Sebastián Palominos
- Center for Genomics and Bioinformatics, Faculty of Science, Universidad Mayor, Camino la Pirámide 5750, Huechuraba 8580745, Chile; (E.V.); (E.M.C.); (S.P.); (P.P.); (T.M.); (C.H.); (N.S.)
- School of Dentistry, Faculty of Science, Universidad Mayor, Avenida Libertador Bernardo O’higgins 2013, Huechuraba 8580745, Chile
| | - Pedro Parra
- Center for Genomics and Bioinformatics, Faculty of Science, Universidad Mayor, Camino la Pirámide 5750, Huechuraba 8580745, Chile; (E.V.); (E.M.C.); (S.P.); (P.P.); (T.M.); (C.H.); (N.S.)
- School of Dentistry, Faculty of Science, Universidad Mayor, Avenida Libertador Bernardo O’higgins 2013, Huechuraba 8580745, Chile
| | - Tania Mella
- Center for Genomics and Bioinformatics, Faculty of Science, Universidad Mayor, Camino la Pirámide 5750, Huechuraba 8580745, Chile; (E.V.); (E.M.C.); (S.P.); (P.P.); (T.M.); (C.H.); (N.S.)
- School of Medical Technology, Faculty of Science, Universidad Mayor, Camino la Pirámide 5750, Huechuraba 8580745, Chile
| | - Constanza Henrríquez
- Center for Genomics and Bioinformatics, Faculty of Science, Universidad Mayor, Camino la Pirámide 5750, Huechuraba 8580745, Chile; (E.V.); (E.M.C.); (S.P.); (P.P.); (T.M.); (C.H.); (N.S.)
- School of Medical Technology, Faculty of Science, Universidad Mayor, Camino la Pirámide 5750, Huechuraba 8580745, Chile
| | - Nelda Salazar
- Center for Genomics and Bioinformatics, Faculty of Science, Universidad Mayor, Camino la Pirámide 5750, Huechuraba 8580745, Chile; (E.V.); (E.M.C.); (S.P.); (P.P.); (T.M.); (C.H.); (N.S.)
- School of Medical Technology, Faculty of Science, Universidad Mayor, Camino la Pirámide 5750, Huechuraba 8580745, Chile
| | - Gustavo Monasterio
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Sergio Livingstone Pohlhammer 943, Independencia 8380492, Chile; (G.M.); (E.A.C.)
| | - Emilio A. Cafferata
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Sergio Livingstone Pohlhammer 943, Independencia 8380492, Chile; (G.M.); (E.A.C.)
- Department of Periodontology, School of Dentistry, Universidad Científica del Sur, Av. Paseo de la República 5544, Lima 15074, Peru
| | - Paola Murgas
- Center for Integrative Biology, Faculty of Science, Universidad Mayor, Camino la Pirámide 5750, Huechuraba 8580745, Chile; (B.I.C.); (P.M.)
| | - Rolando Vernal
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Sergio Livingstone Pohlhammer 943, Independencia 8380492, Chile; (G.M.); (E.A.C.)
| | - Cristian Cortez
- Center for Genomics and Bioinformatics, Faculty of Science, Universidad Mayor, Camino la Pirámide 5750, Huechuraba 8580745, Chile; (E.V.); (E.M.C.); (S.P.); (P.P.); (T.M.); (C.H.); (N.S.)
- School of Dentistry, Faculty of Science, Universidad Mayor, Avenida Libertador Bernardo O’higgins 2013, Huechuraba 8580745, Chile
- School of Medical Technology, Faculty of Science, Universidad Mayor, Camino la Pirámide 5750, Huechuraba 8580745, Chile
| |
Collapse
|
44
|
Akpinar HA, Kahraman H, Yaman I. Ochratoxin A Sequentially Activates Autophagy and the Ubiquitin-Proteasome System. Toxins (Basel) 2019; 11:E615. [PMID: 31653047 PMCID: PMC6891609 DOI: 10.3390/toxins11110615] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/12/2019] [Accepted: 10/23/2019] [Indexed: 12/21/2022] Open
Abstract
Ochratoxin A (OTA) is a carcinogenic mycotoxin, which is produced by Aspergillus and Penicillium genera of fungi and commonly contaminates food and feed. We and others have previously shown that OTA causes sustained activation of PI3K/AKT and MAPK/ERK1-2 signaling pathways in different cell types and animal models. Given the close relationship between cellular signaling activity and protein stability, we were curious whether increased PI3K/AKT and MAPK/ERK1-2 signaling may be the result of OTA-stimulated alterations in proteolytic activity. We show that both of the major proteolytic systems, autophagy, and the ubiquitin-proteasome system (UPS), are activated upon OTA exposure in human kidney proximal tubule HK-2 and mouse embryonic fibroblast (MEF) cells. OTA stimulates transient autophagic activity at early time points of treatment but autophagic activity subsides after 6 h even in the sustained presence of OTA. Interestingly, OTA exposure also results in increased cell death in wild-type MEF cells but not in autophagy-halted Atg5-deficient cells, suggesting that autophagy exerts a pro-death effect on OTA-induced cytotoxicity. In addition, prolonged OTA exposure decreased ubiquitinated protein levels by increasing proteasomal activity. Using purified and cellular proteasomes, we observed enhanced chymotrypsin-, caspase-, and trypsin-like activities of the 26S but not the 20S proteasome in the presence of OTA. However, in the cellular context, increased proteasomal activity depended on prior induction of autophagy. Our results suggest that autophagy and subsequent UPS activation are responsible for sustained activation of PI3K/AKT and MAPK/ERK1-2 pathways through regulating the levels of critical phosphatases VHR/DUSP3, DUSP4, and PHLPP, which are known to be involved in OTA toxicity and carcinogenicity.
Collapse
Affiliation(s)
- Hafize Aysin Akpinar
- Molecular Toxicology and Cancer Research Laboratory, Department of Molecular Biology and Genetics, Bogazici University, Bebek-Istanbul 34342, Turkey.
| | - Hilal Kahraman
- Molecular Toxicology and Cancer Research Laboratory, Department of Molecular Biology and Genetics, Bogazici University, Bebek-Istanbul 34342, Turkey.
| | - Ibrahim Yaman
- Molecular Toxicology and Cancer Research Laboratory, Department of Molecular Biology and Genetics, Bogazici University, Bebek-Istanbul 34342, Turkey.
- Center for Life Sciences and Technologies, Bogazici University, Bebek-Istanbul 34342, Turkey.
| |
Collapse
|
45
|
Dodig S, Čepelak I, Pavić I. Hallmarks of senescence and aging. Biochem Med (Zagreb) 2019; 29:030501. [PMID: 31379458 PMCID: PMC6610675 DOI: 10.11613/bm.2019.030501] [Citation(s) in RCA: 192] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 06/10/2019] [Indexed: 12/12/2022] Open
Abstract
The complex process of biological aging, as an intrinsic feature of living beings, is the result of genetic and, to a greater extent, environmental factors and time. For many of the changes taking place in the body during aging, three factors are important: inflammation, immune aging and senescence (cellular aging, biological aging). Senescence is an irreversible form of long-term cell-cycle arrest, caused by excessive intracellular or extracellular stress or damage. The purpose of this cell-cycles arrest is to limit the proliferation of damaged cells, to eliminate accumulated harmful factors and to disable potential malignant cell transformation. As the biological age does not have to be in accordance with the chronological age, it is important to find specific hallmarks and biomarkers that could objectively determine the rate of age of a person. These biomarkers might be a valuable measure of physiological, i.e. biological age. Biomarkers should meet several criteria. For example, they have to predict the rate of aging, monitor a basic process that underlies the aging process, be able to be tested repeatedly without harming the person. In addition, biomarkers have to be indicators of biological processes, pathogenic processes or pharmacological responses to therapeutic intervention. It is considered that the telomere length is the weak biomarker (with poor predictive accuracy), and there is currently no reliable biomarker that meets all the necessary criteria.
Collapse
Affiliation(s)
- Slavica Dodig
- Department of Medical Biochemistry and Hematology, Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia
| | - Ivana Čepelak
- Department of Medical Biochemistry and Hematology, Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia
| | - Ivan Pavić
- Department of Pulmonology, Allergology and Immunology, Children’s Hospital Zagreb; School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
46
|
Kong Y, Huang T, Zhang H, Zhang Q, Ren J, Guo X, Fan H, Liu L. The lncRNA NEAT1/miR-29b/Atg9a axis regulates IGFBPrP1-induced autophagy and activation of mouse hepatic stellate cells. Life Sci 2019; 237:116902. [PMID: 31610195 DOI: 10.1016/j.lfs.2019.116902] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 09/13/2019] [Accepted: 09/20/2019] [Indexed: 12/22/2022]
Abstract
AIMS Insulin-like growth factor binding protein-related protein 1 (IGFBPrP1) promotes hepatic stellate cell (HSC) autophagy and activation. However, the underlying mechanism remains unknown. Noncoding RNAs (ncRNAs) including long noncoding RNAs (lncRNAs) and microRNAs (miRNAs), have received increasing attention. We aimed to investigate the roles of the lncRNA nuclear enriched abundant transcript 1 (NEAT1), miR-29b, and autophagy related protein 9a (Atg9a), and their relationships with each other during IGFBPrP1-induced HSC autophagy and activation. MAIN METHODS Levels of NEAT1, miR-29b, Atg9a, and autophagy were detected in adenovirus-mediated IGFBPrP1 (AdIGFBPrP1)-treated mouse liver tissue and immortalized mouse hepatic stellate cell line JS1 transfected with either AdIGFBPrP1 or siIGFBPrP1. In AdIGFBPrP1-treated JS1 cells, autophagy and activation were detected after altering NEAT1, miR-29b, or Atg9a levels. In AdIGFBPrP1-treated JS1 cells, relationships among NEAT1, miR-29b, and Atg9a were explored using dual-luciferase reporter assays, Western blot, qRT-PCR, and immunofluorescence. KEY FINDINGS IGFBPrP1 increased levels of NEAT1, Atg9a, and autophagy while decreasing the level of miR-29b in mouse liver tissues and mouse HSCs. Moreover, NEAT1 increased HSC autophagy and activation while miR-29b decreased both processes. Atg9a also participated in IGFBPrP1-induced HSC autophagy and activation. Importantly, NEAT1, miR-29b, and Atg9a formed a NEAT1/miR-29b/Atg9a regulatory axis for IGFBPrP1-induced HSC autophagy and activation. SIGNIFICANCE Our study unveiled the new NEAT1/miR-29b/Atg9a regulatory axis involved in IGFBPrP1-induced mouse HSC autophagy and activation. The study thus provides new insights in the pathogenesis and potential therapeutic strategies of liver fibrosis.
Collapse
Affiliation(s)
- Yangyang Kong
- Department of Gastroenterology and Hepatology, The First Clinical Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Tingjuan Huang
- Department of Gastroenterology and Hepatology, The First Clinical Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Haiyan Zhang
- Department of Gastroenterology and Hepatology, The First Clinical Hospital of Shanxi Medical University, Taiyuan, 030001, China; Experimental Center of Science and Research, The First Clinical Hospital of Shanxi Medical University, Taiyuan, 030001, China; Key Laboratory of Cell Physiology, Provincial Department of the Ministry of Education, Shanxi Medical University, Taiyuan, 030001, China
| | - Qianqian Zhang
- Department of Gastroenterology and Hepatology, The First Clinical Hospital of Shanxi Medical University, Taiyuan, 030001, China; Experimental Center of Science and Research, The First Clinical Hospital of Shanxi Medical University, Taiyuan, 030001, China; Key Laboratory of Cell Physiology, Provincial Department of the Ministry of Education, Shanxi Medical University, Taiyuan, 030001, China
| | - Junjie Ren
- Department of Gastroenterology and Hepatology, The First Clinical Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Xiaohong Guo
- Department of Gastroenterology and Hepatology, The First Clinical Hospital of Shanxi Medical University, Taiyuan, 030001, China; Experimental Center of Science and Research, The First Clinical Hospital of Shanxi Medical University, Taiyuan, 030001, China; Key Laboratory of Cell Physiology, Provincial Department of the Ministry of Education, Shanxi Medical University, Taiyuan, 030001, China
| | - Huiqin Fan
- Department of Gastroenterology and Hepatology, The First Clinical Hospital of Shanxi Medical University, Taiyuan, 030001, China; Experimental Center of Science and Research, The First Clinical Hospital of Shanxi Medical University, Taiyuan, 030001, China; Key Laboratory of Cell Physiology, Provincial Department of the Ministry of Education, Shanxi Medical University, Taiyuan, 030001, China
| | - Lixin Liu
- Department of Gastroenterology and Hepatology, The First Clinical Hospital of Shanxi Medical University, Taiyuan, 030001, China; Experimental Center of Science and Research, The First Clinical Hospital of Shanxi Medical University, Taiyuan, 030001, China; Key Laboratory of Cell Physiology, Provincial Department of the Ministry of Education, Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
47
|
Torp MK, Yang K, Ranheim T, Husø Lauritzen K, Alfsnes K, Vinge LE, Aukrust P, Stensløkken KO, Yndestad A, Sandanger Ø. Mammalian Target of Rapamycin (mTOR) and the Proteasome Attenuates IL-1β Expression in Primary Mouse Cardiac Fibroblasts. Front Immunol 2019; 10:1285. [PMID: 31244838 PMCID: PMC6563870 DOI: 10.3389/fimmu.2019.01285] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 05/20/2019] [Indexed: 12/21/2022] Open
Abstract
Background: IL-1β is a highly potent pro-inflammatory cytokine and its secretion is tightly regulated. Inactive pro-IL-1β is transcribed in response to innate immune receptors activating NFκB. If tissue damage occurs, danger signals released from necrotic cells, such as ATP, can activate NLRP3-inflammasomes (multiprotein complexes consisting of NLRP3, ASC, and active caspase-1) which cleaves and activates pro-IL-1β. NLRP3 activation also depends on NEK7 and mitochondrial ROS-production. Thus, IL-1β secretion may be regulated at the level of each involved component. We have previously shown that NLRP3-dependent IL-1β release can be induced in cardiac fibroblasts by pro-inflammatory stimuli. However, anti-inflammatory mechanisms targeting IL-1β release in cardiac cells have not been investigated. mTOR is a key regulator of protein metabolism, including autophagy and proteasome activity. In this study we explored whether autophagy or proteasomal degradation are regulators of NLRP3 inflammasome activation and IL-1β release from cardiac fibroblasts. Methods and Results: Serum starvation selectively reduced LPS/ATP-induced IL-1β secretion from cardiac fibroblasts. However, no other inflammasome components, nor mitochondrial mass, were affected. The mTOR inhibitor rapamycin restored pro-IL-1β protein levels as well as LPS/ATP-induced IL-1β release from serum starved cells. However, neither serum starvation nor rapamycin induced autophagy in cardiac fibroblasts. Conversely, chloroquine and bafilomycin A (inhibitors of autophagy) and betulinic acid (a proteasome activator) effectively reduced LPS-induced pro-IL-1β protein levels. Key findings were reinvestigated in human monocyte-derived macrophages. Conclusion: In cardiac fibroblasts, mTOR inhibition selectively favors pro-IL-1β synthesis while proteasomal degradation and not autophagy is the major catabolic anti-inflammatory mechanism for degradation of this cytokine.
Collapse
Affiliation(s)
- May-Kristin Torp
- Division of Physiology, Department of Molecular Medicine, Faculty of Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Centre for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Kuan Yang
- Centre for Heart Failure Research, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Trine Ranheim
- Centre for Heart Failure Research, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Knut Husø Lauritzen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Katrine Alfsnes
- Centre for Heart Failure Research, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Leif E. Vinge
- Centre for Heart Failure Research, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Department of Internal Medicine, Diakonhjemmet Hospital, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Kåre-Olav Stensløkken
- Division of Physiology, Department of Molecular Medicine, Faculty of Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Centre for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Arne Yndestad
- Centre for Heart Failure Research, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Øystein Sandanger
- Centre for Heart Failure Research, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Section of Dermatology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| |
Collapse
|
48
|
Walczak J, Dębska-Vielhaber G, Vielhaber S, Szymański J, Charzyńska A, Duszyński J, Szczepanowska J. Distinction of sporadic and familial forms of ALS based on mitochondrial characteristics. FASEB J 2018; 33:4388-4403. [PMID: 30550341 DOI: 10.1096/fj.201801843r] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Bioenergetic failure, oxidative stress, and changes in mitochondrial morphology are common pathologic hallmarks of amyotrophic lateral sclerosis (ALS) in several cellular and animal models. Disturbed mitochondrial physiology has serious consequences for proper functioning of the cell, leading to the chronic mitochondrial stress. Mitochondria, being in the center of cellular metabolism, play a pivotal role in adaptation to stress conditions. We found that mitochondrial dysfunction and adaptation processes differ in primary fibroblasts derived from patients diagnosed with either sporadic or familial forms of ALS. The evaluation of mitochondrial parameters such as the mitochondrial membrane potential, the oxygen consumption rate, the activity and levels of respiratory chain complexes, and the levels of ATP, reactive oxygen species, and Ca2+ show that the bioenergetic properties of mitochondria are different in sporadic ALS, familial ALS, and control groups. Comparative statistical analysis of the data set (with use of principal component analysis and support vector machine) identifies and distinguishes 3 separate groups despite the small number of investigated cell lines and high variability in measured parameters. These findings could be a first step in development of a new tool for predicting sporadic and familial forms of ALS and could contribute to knowledge of its pathophysiology.-Walczak, J., Dębska-Vielhaber, G., Vielhaber, S., Szymański, J., Charzyńska, A., Duszyński, J., Szczepanowska, J. Distinction of sporadic and familial forms of ALS based on mitochondrial characteristics.
Collapse
Affiliation(s)
- Jarosław Walczak
- Laboratory of Bioenergetics and Biomembranes, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | | | - Stefan Vielhaber
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany; and
| | - Jędrzej Szymański
- Laboratory of Bioenergetics and Biomembranes, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Agata Charzyńska
- Laboratory of Bioinformatics, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Jerzy Duszyński
- Laboratory of Bioenergetics and Biomembranes, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Joanna Szczepanowska
- Laboratory of Bioenergetics and Biomembranes, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
49
|
Kosztelnik M, Kurucz A, Papp D, Jones E, Sigmond T, Barna J, Traka MH, Lorincz T, Szarka A, Banhegyi G, Vellai T, Korcsmaros T, Kapuy O. Suppression of AMPK/aak-2 by NRF2/SKN-1 down-regulates autophagy during prolonged oxidative stress. FASEB J 2018; 33:2372-2387. [PMID: 30277819 PMCID: PMC6338645 DOI: 10.1096/fj.201800565rr] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
NF-E2–related factor 2 (NRF2) transcription factor has a fundamental role in cell homeostasis maintenance as one of the master regulators of oxidative and electrophilic stress responses. Previous studies have shown that a regulatory connection exists between NRF2 and autophagy during reactive oxygen species–generated oxidative stress. The aim of the present study was to investigate how autophagy is turned off during prolonged oxidative stress, to avoid overeating and destruction of essential cellular components. AMPK is a key cellular energy sensor highly conserved in eukaryotic organisms, and it has an essential role in autophagy activation at various stress events. Here the role of human AMPK and its Caenorhabditis elegans counterpart AAK-2 was explored upon oxidative stress. We investigated the regulatory connection between NRF2 and AMPK during oxidative stress induced by tert-butyl hydroperoxide (TBHP) in HEK293T cells and C. elegans. Putative conserved NRF2/protein skinhead-1 binding sites were found in AMPK/aak-2 genes by in silico analysis and were later confirmed experimentally by using EMSA. After addition of TBHP, NRF2 and AMPK showed a quick activation; AMPK was later down-regulated, however, while NRF2 level remained high. Autophagosome formation and Unc-51–like autophagy activating kinase 1 phosphorylation were initially stimulated, but they returned to basal values after 4 h of TBHP treatment. The silencing of NRF2 resulted in a constant activation of AMPK leading to hyperactivation of autophagy during oxidative stress. We observed the same effects in C. elegans demonstrating the conservation of this self-defense mechanism to save cells from hyperactivated autophagy upon prolonged oxidative stress. We conclude that NRF2 negatively regulates autophagy through delayed down-regulation of the expression of AMPK upon prolonged oxidative stress. This regulatory connection between NRF2 and AMPK may have an important role in understanding how autophagy is regulated in chronic human morbidities characterized by oxidative stress, such as neurodegenerative diseases, certain cancer types, and in metabolic diseases.—Kosztelnik, M., Kurucz, A., Papp, D., Jones, E., Sigmond, T., Barna, J., Traka, M. H., Lorincz, T., Szarka, A., Banhegyi, G., Vellai, T., Korcsmaros, T., Kapuy, O. Suppression of AMPK/aak-2 by NRF2/SKN-1 down-regulates autophagy during prolonged oxidative stress.
Collapse
Affiliation(s)
| | - Anita Kurucz
- Department of Medical Chemistry, Molecular Biology, and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| | - Diana Papp
- Department of Genetics, Eötvös Loránd University, Budapest, Hungary
| | - Emily Jones
- Gut Health and Microbes, Quadram Institute, Norwich Research Park, Norwich, United Kingdom.,Earlham Institute, Norwich, United Kingdom
| | - Timea Sigmond
- Department of Genetics, Eötvös Loránd University, Budapest, Hungary
| | - Janos Barna
- Department of Genetics, Eötvös Loránd University, Budapest, Hungary
| | - Maria H Traka
- Food Innovation and Health, Quadram Institute, Norwich, United Kingdom
| | - Tamas Lorincz
- Laboratory of Biochemistry and Molecular Biology, Department of Applied Biotechnology and Food Science, Budapest University of Technology and Economics, Budapest, Hungary
| | - Andras Szarka
- Laboratory of Biochemistry and Molecular Biology, Department of Applied Biotechnology and Food Science, Budapest University of Technology and Economics, Budapest, Hungary
| | - Gabor Banhegyi
- Department of Medical Chemistry, Molecular Biology, and Pathobiochemistry, Semmelweis University, Budapest, Hungary.,Pathobiochemistry Research Group, Hungarian Academy of Sciences, Semmelweis University, Budapest, Hungary
| | - Tibor Vellai
- Department of Genetics, Eötvös Loránd University, Budapest, Hungary
| | - Tamas Korcsmaros
- Department of Genetics, Eötvös Loránd University, Budapest, Hungary.,Gut Health and Microbes, Quadram Institute, Norwich Research Park, Norwich, United Kingdom.,Earlham Institute, Norwich, United Kingdom
| | - Orsolya Kapuy
- Department of Medical Chemistry, Molecular Biology, and Pathobiochemistry, Semmelweis University, Budapest, Hungary
| |
Collapse
|
50
|
Liao L, Zhuang X, Li W, Su Q, Zhao J, Liu Y. Polysaccharide from Fuzi protects against Ox‑LDL‑induced calcification of human vascular smooth muscle cells by increasing autophagic activity. Mol Med Rep 2018; 17:5109-5115. [PMID: 29393437 PMCID: PMC5865975 DOI: 10.3892/mmr.2018.8488] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 09/01/2017] [Indexed: 02/01/2023] Open
Abstract
Polysaccharide from Fuzi (FPS) is a water‑soluble polysaccharide isolated from the traditional Chinese herbal medicine Fuzi. It has been demonstrated to protect hepatocytes against ischemia‑reperfusion injury through its potent antioxidant effects, and to attenuate starvation‑induced cytotoxicity in H9c2 cells by increasing autophagic activity. In the present study, Alizarin Red S staining was used to detect mineral deposition and reverse transcription‑quantitative polymerase chain reaction was used to detect the core binding factor α1 and smooth muscle 22α mRNA expression. To analyze autophagic activity, western blotting was used to detect microtubule‑associated protein 1A/1B light chain 3 and nucleoporin P62 expression. In addition, green fluorescent protein‑LC3 dots‑per‑cell was observed by fluorescence microscopy. It was demonstrated that oxidized low‑density lipoprotein (Ox‑LDL) could increase the calcification of human vascular smooth muscle cells (VSMCs) in a concentration‑dependent manner, and that FPS treatment had a significant protective effect against Ox‑LDL‑induced calcification of human VSMCs. Furthermore, FPS treatment alleviated the Ox‑LDL‑induced downregulation of autophagic activity, and the protective effect of FPS on Ox‑LDL‑induced calcification was attenuated by the autophagy inhibitor 3‑methyladenine. In conclusion, the present study demonstrated for the first time to the best of the authors' knowledge that FPS can protect against Ox‑LDL‑induced vascular calcification in human VSMCs, and that this likely occurs via the activation of autophagy. This supports the hypothesis that autophagy may be an endogenous protective mechanism counteracting vascular calcification, and that FPS may be used as a potential therapeutic for vascular calcification.
Collapse
Affiliation(s)
- Lizhen Liao
- Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, Guangdong 510006, P.R. China
| | - Xiaodong Zhuang
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Weidong Li
- Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, Guangdong 510006, P.R. China
| | - Qibiao Su
- Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, Guangdong 510006, P.R. China
| | - Jie Zhao
- Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, Guangdong 510006, P.R. China
| | - Ying Liu
- Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, Guangdong 510006, P.R. China
| |
Collapse
|