1
|
Garibaldi N, Besio R, Pirota V, Albini B, Colombo G, Galinetto P, Doria F, Carriero A, Forlino A. A novel chemical chaperone ameliorates osteoblast homeostasis and extracellular matrix in osteogenesis imperfecta. Life Sci 2025; 361:123320. [PMID: 39706289 DOI: 10.1016/j.lfs.2024.123320] [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: 11/07/2024] [Revised: 12/13/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
AIMS Osteogenesis imperfecta (OI) is a collagen I-related heritable family of skeletal diseases associated to extreme bone fragility and deformity. Its classical forms are caused by dominant mutations in COL1A1 and COL1A2, which encode for the protein α chains, and are characterized by impairment in collagen I structure, folding, and secretion. Mutant collagen I assembles in an altered extracellular matrix affecting mineralization and bone properties and partially accumulating inside the cells, leading to impaired trafficking and cellular stress. Recently, the chemical chaperone 4-phenylbutyrate (4-PBA) has been proposed as an innovative drug for OI based on its ability to restore intracellular homeostasis, stimulate secretion, and ameliorate collagen-producing cell functions, positively affecting bone properties. However, the limited half-life of the molecule represents a serious hurdle for its use. MATERIALS AND METHODS To efficiently target cellular stress as OI treatment, two new compounds were designed by molecular modelling based on the 4-PBA structure to increase its stability and its ability to implement protein secretion. The short butyryl fatty acid chain of 4-PBA was substituted with a nitro functional group or with a glycine, respectively. The latter, N-benzyl glycine (N-BG), showed the best docking score, less toxicity, and higher stability than 4-PBA. KEY FINDINGS N-BG improved extracellular matrix quality and mineral content together with ameliorating OI cells' homeostasis by increasing ER-associated degradation pathway, reducing apoptosis, and stimulating protein secretion, thus facilitating intracellular clearance from accumulated misfolded proteins. SIGNIFICANCE In conclusion, N-BG represents a novel potential available compound to target altered homeostasis in OI with the aim to ameliorate the disease phenotype.
Collapse
Affiliation(s)
- Nadia Garibaldi
- Department of Biomedical Engineering, The City College of New York, New York, USA; Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy.
| | - Roberta Besio
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy.
| | | | | | | | | | - Filippo Doria
- Department of Chemistry, University of Pavia, Pavia, Italy.
| | - Alessandra Carriero
- Department of Biomedical Engineering, The City College of New York, New York, USA.
| | - Antonella Forlino
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, Pavia, Italy.
| |
Collapse
|
2
|
Ma L, Li K, Guo Y, Liu J, Dong J, Li J, Ren Y, Shi L. Selenium triggers AMPK-mTOR pathway to modulate autophagy related to oxidative stress of sheep Leydig cells. Reprod Biol 2024; 25:100973. [PMID: 39580868 DOI: 10.1016/j.repbio.2024.100973] [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: 03/20/2024] [Revised: 09/20/2024] [Accepted: 11/13/2024] [Indexed: 11/26/2024]
Abstract
The objective of this study was to investigate the effect of oxidative stress induced by excessive Se on autophagy of sheep Leydig cells and its underlying mechanism. Leydig cells isolated from the testis of 8-month-old sheep were purified using a discontinuous Percoll density gradient. Cells were divided into four treatment groups (0, 2.0, 4.0 and 8.0 μmol/L of Se). After treatment with Se for 48 h, cell proliferation was detected by CCK-8 assay kit. The biochemical methods were used to evaluate the antioxidant status of Leydig cells. The mRNA transcript and protein abundance related to the AMPK-mTOR pathway and autophagy were detected by real-time PCR and western blot analysis. The results showed that the Leydig cells treated with 8.0 μmol/L Se have the lowest cell viability. The greater ROS content and lower GSH-Px activity were also observed in the Se8.0 group. The inclusion of 2.0 μmol/L Se in the medium did not affect the autophagy of Leydig cells. However, the relative abundance of ATG5 protein and LC3II/I ratio were elevated in the Se8.0 group. Oxidative stress induced by excessive Se (8.0 μmol/L) dramatically improved the abundance of key proteins related to AMPK-mTOR pathway and led to an increase of phosphorylated AMPK, mTOR and ULK1. Compared with the Se8.0 group, compound C could significantly inhibit the key molecules of AMPK-mTOR signaling pathway and mitigate the autophagy of Leydig cells induced by excessive Se. These results indicate that appropriate Se (2.0 μmol/L) can enhance the viability of sheep Leydig cells. Oxidative stress caused by Se excess can induce cell autophagy via activating AMPK-mTOR signaling pathway. The existed crosstalk between autophagy and apoptosis could decide the fate of Leydig cells. This process could play a decisive role in the maintenance of normal male fertility and spermatogenesis by affecting the number of Leydig cells in testis.
Collapse
Affiliation(s)
- Liang Ma
- Laboratory of Animal Reproductive biotechnology, Shanxi Agricultural University, Taigu 030801, PR China
| | - Kexin Li
- Laboratory of Animal Reproductive biotechnology, Shanxi Agricultural University, Taigu 030801, PR China
| | - Yaru Guo
- Laboratory of Animal Reproductive biotechnology, Shanxi Agricultural University, Taigu 030801, PR China
| | - Jinyu Liu
- Laboratory of Animal Reproductive biotechnology, Shanxi Agricultural University, Taigu 030801, PR China
| | - Jianing Dong
- Laboratory of Animal Reproductive biotechnology, Shanxi Agricultural University, Taigu 030801, PR China
| | - Jun Li
- Laboratory of Animal Reproductive biotechnology, Shanxi Agricultural University, Taigu 030801, PR China
| | - Youshe Ren
- College of Animal Science, Shanxi Agricultural University, Taigu 030801, PR China; Laboratory of Animal Reproductive biotechnology, Shanxi Agricultural University, Taigu 030801, PR China.
| | - Lei Shi
- College of Animal Science, Shanxi Agricultural University, Taigu 030801, PR China; Laboratory of Animal Reproductive biotechnology, Shanxi Agricultural University, Taigu 030801, PR China.
| |
Collapse
|
3
|
Hamwi MN, Elsayed E, Dabash H, Abuawad A, Aweer NA, Al Zeir F, Pedersen S, Al-Mansoori L, Burgon PG. MLIP and Its Potential Influence on Key Oncogenic Pathways. Cells 2024; 13:1109. [PMID: 38994962 PMCID: PMC11240681 DOI: 10.3390/cells13131109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 05/27/2024] [Accepted: 06/19/2024] [Indexed: 07/13/2024] Open
Abstract
Muscle-enriched A-type lamin-interacting protein (MLIP) is an emerging protein involved in cellular homeostasis and stress adaptation. Eukaryotic cells regulate various cellular processes, including metabolism, DNA repair, and cell cycle progression, to maintain cellular homeostasis. Disruptions in this homeostasis can lead to diseases such as cancer, characterized by uncontrolled cell growth and division. This review aims to explore for the first time the unique role MLIP may play in cancer development and progression, given its interactions with the PI3K/Akt/mTOR pathway, p53, MAPK9, and FOXO transcription factors, all critical regulators of cellular homeostasis and tumor suppression. We discuss the current understanding of MLIP's involvement in pro-survival pathways and its potential implications in cancer cells' metabolic remodeling and dysregulated homeostasis. Additionally, we examine the potential of MLIP as a novel therapeutic target for cancer treatment. This review aims to shed light on MLIP's potential impact on cancer biology and contribute to developing innovative therapeutic strategies.
Collapse
Affiliation(s)
- Mahmoud N Hamwi
- College of Medicine, Qatar University, Doha P.O. Box 0974, Qatar
| | - Engy Elsayed
- College of Medicine, Qatar University, Doha P.O. Box 0974, Qatar
| | - Hanan Dabash
- Department of Chemistry and Earth Sciences, College of Arts and Sciences, Qatar University, Doha P.O. Box 2713, Qatar
| | - Amani Abuawad
- Department of Chemistry and Earth Sciences, College of Arts and Sciences, Qatar University, Doha P.O. Box 2713, Qatar
| | - Noor A Aweer
- College of Medicine, Qatar University, Doha P.O. Box 0974, Qatar
| | - Faissal Al Zeir
- College of Medicine, Qatar University, Doha P.O. Box 0974, Qatar
| | - Shona Pedersen
- College of Medicine, Qatar University, Doha P.O. Box 0974, Qatar
| | - Layla Al-Mansoori
- Biomedical Research Centre, Qatar University, Doha P.O. Box 2713, Qatar
| | - Patrick G Burgon
- Department of Chemistry and Earth Sciences, College of Arts and Sciences, Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
4
|
Jobst M, Hossain M, Kiss E, Bergen J, Marko D, Del Favero G. Autophagy modulation changes mechano-chemical sensitivity of T24 bladder cancer cells. Biomed Pharmacother 2024; 170:115942. [PMID: 38042111 DOI: 10.1016/j.biopha.2023.115942] [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: 10/27/2023] [Accepted: 11/21/2023] [Indexed: 12/04/2023] Open
Abstract
Bladder cancer cells possess unique adaptive capabilities: shaped by their environment, cells face a complex chemical mixture of metabolites and xenobiotics accompanied by physiological mechanical cues. These responses might translate into resistance to chemotherapeutical regimens and can largely rely on autophagy. Considering molecules capable of rewiring tumor plasticity, compounds of natural origin promise to offer valuable options. Fungal derived metabolites, such as bafilomycin and wortmannin are widely acknowledged as autophagy inhibitors. Here, their potential to tune bladder cancer cells´ adaptability to chemical and physical stimuli was assessed. Additionally, dietary occurring mycotoxins were also investigated, namely deoxynivalenol (DON, 0.1-10 µM) and fusaric acid (FA, 0.1-1 mM). Endowing a Janus' face behavior, DON and FA are on the one side described as toxins with detrimental health effects. Concomitantly, they are also explored experimentally for selective pharmacological applications including anticancer activities. In non-cytotoxic concentrations, bafilomycin (BAFI, 1-10 nM) and wortmannin (WORT, 1 µM) modified cell morphology and reduced cancer cell migration. Application of shear stress and inhibition of mechano-gated PIEZO channels reduced cellular sensitivity to BAFI treatment (1 nM). Similarly, for FA (0.5 mM) PIEZO1 expression and inhibition largely aligned with the modulatory potential on cancer cells motility. Additionally, this study highlighted that the activity profile of compounds with similar cytotoxic potential (e.g. co-incubation DON with BAFI or FA with WORT) can diverge substantially in the regulation of cell mechanotransduction. Considering the interdependence between tumor progression and response to mechanical cues, these data promise to provide a novel viewpoint for the study of chemoresistance and associated pathways.
Collapse
Affiliation(s)
- Maximilian Jobst
- Department of Food Chemistry and Toxicology, University of Vienna Faculty of Chemistry, Währinger Str. 38-40, 1090 Vienna, Austria; Core Facility Multimodal Imaging, University of Vienna Faculty of Chemistry, Währinger Str. 38-40, 1090 Vienna, Austria; University of Vienna, Vienna Doctoral School in Chemistry (DoSChem), Währinger Str. 42, 1090 Vienna, Austria
| | - Maliha Hossain
- Department of Food Chemistry and Toxicology, University of Vienna Faculty of Chemistry, Währinger Str. 38-40, 1090 Vienna, Austria
| | - Endre Kiss
- Core Facility Multimodal Imaging, University of Vienna Faculty of Chemistry, Währinger Str. 38-40, 1090 Vienna, Austria
| | - Janice Bergen
- Department of Food Chemistry and Toxicology, University of Vienna Faculty of Chemistry, Währinger Str. 38-40, 1090 Vienna, Austria; Core Facility Multimodal Imaging, University of Vienna Faculty of Chemistry, Währinger Str. 38-40, 1090 Vienna, Austria; University of Vienna, Vienna Doctoral School in Chemistry (DoSChem), Währinger Str. 42, 1090 Vienna, Austria
| | - Doris Marko
- Department of Food Chemistry and Toxicology, University of Vienna Faculty of Chemistry, Währinger Str. 38-40, 1090 Vienna, Austria
| | - Giorgia Del Favero
- Department of Food Chemistry and Toxicology, University of Vienna Faculty of Chemistry, Währinger Str. 38-40, 1090 Vienna, Austria; Core Facility Multimodal Imaging, University of Vienna Faculty of Chemistry, Währinger Str. 38-40, 1090 Vienna, Austria.
| |
Collapse
|
5
|
Inhibition of the MAPK/c-Jun-EGR1 Pathway Decreases Photoreceptor Cell Death in the rd1 Mouse Model for Inherited Retinal Degeneration. Int J Mol Sci 2022; 23:ijms232314600. [PMID: 36498926 PMCID: PMC9740268 DOI: 10.3390/ijms232314600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/17/2022] [Accepted: 11/20/2022] [Indexed: 11/24/2022] Open
Abstract
Retinitis pigmentosa (RP) is a group of inherited retinal dystrophies that typically results in photoreceptor cell death and vision loss. Here, we explored the effect of early growth response-1 (EGR1) expression on photoreceptor cell death in Pde6brd1 (rd1) mice and its mechanism of action. To this end, single-cell RNA-seq (scRNA-seq) was used to identify differentially expressed genes in rd1 and congenic wild-type (WT) mice. Chromatin immunoprecipitation (ChIP), the dual-luciferase reporter gene assay, and western blotting were used to verify the relationship between EGR1 and poly (ADP-ribose) polymerase-1 (PARP1). Immunofluorescence staining was used to assess PARP1 expression after silencing or overexpression of EGR1. Photoreceptor cell death was assessed using the TUNEL assay following silencing/overexpression of EGR1 or administration of MAPK/c-Jun pathway inhibitors tanzisertib and PD98059. Our results showed differential expression of ERG1 in rd1 and WT mice via scRNA-seq analysis. The ChIP assay demonstrated EGR1 binding to the PARP1 promoter region. The dual-luciferase reporter gene assay and western blotting results revealed that EGR1 upregulated PARP1 expression. Additionally, the TUNEL assay showed that silencing EGR1 effectively reduced photoreceptor cell death. Similarly, the addition of tanzisertib and PD98059 reduced the expression of c-Jun and EGR1 and decreased photoreceptor cell death. Our study revealed that inhibition of the MAPK/c-Jun pathway reduced the expression of EGR1 and PARP1 and prevented photoreceptor cell death. These results highlight the importance of EGR1 for photoreceptor cell death and identify a new avenue for therapeutic interventions in RP.
Collapse
|
6
|
Inflammation Causes Exacerbation of COVID-19: How about Skin Inflammation? Int J Mol Sci 2022; 23:ijms232012260. [PMID: 36293117 PMCID: PMC9603600 DOI: 10.3390/ijms232012260] [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: 08/05/2022] [Revised: 09/10/2022] [Accepted: 09/27/2022] [Indexed: 01/24/2023] Open
Abstract
COVID-19 is a recently emerged viral infection worldwide. SARS-CoV-2, the causative virus, is believed to have emerged from bat coronaviruses, probably through host conversion. The bat coronavirus which has the highest gene homology to SARS-CoV-2 specifically infects deep forest bats in China whose habitat extends through the Middle East to Southern Europe. Host conversion might have occurred due to the deforestation by humans exposing wild bats to the environment they had never encountered before. SARS-CoV-2 infects cells through two mechanisms: through its receptor ACE2 with the help of enzyme TMPRSS and through membrane fusion with the help of elastases in the inflammatory condition. Obesity, hypertension, diabetes mellitus, and pulmonary diseases cause poor prognosis of COVID-19. Aging is another factor promoting poor prognosis. These diseases and aging cause low-level and persistent inflammation in humans, which can promote poor prognosis of COVID-19. Psoriasis and atopic dermatitis are the major inflammatory skin diseases. These inflammatory skin conditions, however, do not seem to cause poor prognosis for COVID-19 based on the epidemiological data accumulated so far. These mechanisms need to be elucidated.
Collapse
|
7
|
Hu D, Zhou Z, Wang J, Zhu K. Screening of ferroptosis-related genes with prognostic effect in colorectal cancer by bioinformatic analysis. Front Mol Biosci 2022; 9:979854. [PMID: 36203871 PMCID: PMC9531163 DOI: 10.3389/fmolb.2022.979854] [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: 06/28/2022] [Accepted: 07/25/2022] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) remains a common malignant tumor of digestive tract with high incidence rate and high mortality in the worldwide. The current clinical treatments of CRC often fail to achieve satisfactory results. Searching for more effective prediction or prognosis biomarkers, or developing more targeted therapeutic schedule may help to improve the outcomes of CRC patients. Here, we tried to study the effect of ferroptosis-related genes on CRC prognosis and make it clearer that ferroptosis has connection with immune environment. First, we obtained gene expression data of CRC and normal tissues, as well as corresponding clinical data from the Gene Expression Omnibus (GEO) database and the Cancer Genome Atlas (TCGA) database. The differentially expressed genes (DEGs) were intersected with ferroptosis-related gene set downloaded from FerrDb database, and 93 abnormally expressed ferroptosis-related genes were obtained. Then, these genes were analyzed for functional enrichment. Univariate Cox regression and multivariate Cox regression analyses were performed to establish prognostic model based on ferroptosis-related genes. In the process of exploring the correlation between prognostic genes and immune infiltration, we found that these genes were closely related to B cells, CD8+ T cells, CD4+ T cells, macrophages and other cells in CRC. In addition, we found a large proportion of plasma cells and macrophages in TCGA-COADREAD. Finally, a prognostic nomogram of ferroptosis-related genes was established, including age, sex, grade and other predicted values. To summary, we established a prognostic model of colorectal cancer (CRC) based on ferroptosis-related genes and further explored the relationship between these genes with immune microenvironment.
Collapse
Affiliation(s)
- Dongzhi Hu
- Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Zhengyang Zhou
- Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Junyi Wang
- Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Kegan Zhu
- Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| |
Collapse
|
8
|
Fu J, Yang Y, Zhu L, Chen Y, Liu B. Unraveling the Roles of Protein Kinases in Autophagy: An Update on Small-Molecule Compounds for Targeted Therapy. J Med Chem 2022; 65:5870-5885. [PMID: 35390258 DOI: 10.1021/acs.jmedchem.1c02053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Protein kinases, which catalyze the phosphorylation of proteins, are involved in several important cellular processes, such as autophagy. Of note, autophagy, originally described as a mechanism for intracellular waste disposal and recovery, has been becoming a crucial biological process closely related to many types of human diseases. More recently, the roles of protein kinases in autophagy have been gradually elucidated, and the design of small-molecule compounds to modulate targets to positively or negatively interfere with the cytoprotective autophagy or autophagy-associated cell death may provide a new clue on the current targeted therapy. Thus, in this Perspective, we focus on summarizing the different roles of protein kinases, including positive, negative, and bidirectional regulations of autophagy. Moreover, we discuss several small-molecule compounds targeting these protein kinases in human diseases, highlighting their pivotal roles in autophagy for targeted therapeutic purposes.
Collapse
Affiliation(s)
- Jiahui Fu
- State Key Laboratory of Biotherapy and Cancer Center, Department of Thoracic Surgery, and Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yushang Yang
- State Key Laboratory of Biotherapy and Cancer Center, Department of Thoracic Surgery, and Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lingjuan Zhu
- State Key Laboratory of Biotherapy and Cancer Center, Department of Thoracic Surgery, and Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yi Chen
- State Key Laboratory of Biotherapy and Cancer Center, Department of Thoracic Surgery, and Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bo Liu
- State Key Laboratory of Biotherapy and Cancer Center, Department of Thoracic Surgery, and Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
9
|
Mufangji Decoction and Its Active Ingredient Patchouli Alcohol Inhibit Tumor Growth through Regulating Akt/mTOR-Mediated Autophagy in Nonsmall-Cell Lung Cancer. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:2373865. [PMID: 34764997 PMCID: PMC8577897 DOI: 10.1155/2021/2373865] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/06/2021] [Accepted: 10/05/2021] [Indexed: 11/29/2022]
Abstract
Background Nonsmall-cell lung cancer (NSCLC) is the main type of lung cancer, whose morbidity and mortality rank first among malignant tumors. More than 70% of NSCLC patients are diagnosed at locally advanced or advanced stage, missing the best operation period. Chemotherapy and targeted therapy are important means for the treatment of advanced NSCLC, but various side effects seriously affect the curative effect and the life quality of NSCLC patients. Our previous clinical practice has shown that Mufangji Decoction, a classic traditional Chinese medicine, has a significant curative effect in the treatment of NSCLC, but the specific mechanism is not clear. This study intends to explore the potential mechanism of Mufangji Decoction and its active ingredient patchouli alcohol against NSCLC and to provide a scientific basis for the prevention and treatment of NSCLC by traditional Chinese medicine. Methods The in vivo and in vitro experiments were performed to evaluate the antitumor effects and investigate the underlying mechanism of Mufangji Decoction and its active ingredient patchouli alcohol. Network pharmacology was applied to analyze the effective ingredients and potential targets or signaling pathways of Mufangji Decoction. Results Our current study shows that Mufangji Decoction can effectively inhibit the growth of subcutaneous transplantation of NSCLC. The following network pharmacological analysis and in vivo experiment suggest that patchouli alcohol is one of the main active ingredients of Mufangji Decoction and exerts antitumor effects. Further mechanism investigation reveals that the antitumor effect of patchouli alcohol is related to the induction of Akt/mTOR signaling pathway-mediated autophagy in NSCLC cells. Conclusion Mufangji Decoction and its active ingredient patchouli alcohol might exert their antitumor effects in NSCLC partly through regulating Akt/mTOR-mediated autophagy, providing the evidence that traditional Chinese medicine might be a key approach for NSCLC treatment via targeting the Akt/mTOR signal axis.
Collapse
|
10
|
Lan CN, Cai WJ, Shi J, Yi ZJ. MAPK inhibitors protect against early‑stage osteoarthritis by activating autophagy. Mol Med Rep 2021; 24:829. [PMID: 34590154 PMCID: PMC8503737 DOI: 10.3892/mmr.2021.12469] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 09/01/2021] [Indexed: 12/26/2022] Open
Abstract
Osteoarthritis (OA) is a chronic, age-related osteoarthropathy that causes a considerable decline in quality of life, as well as economic losses due to its high incidence and poor prognosis. Mitogen-activated protein kinases (MAPKs) regulate multiple cellular processes, including proliferation, differentiation and apoptosis, in certain diseases, such as cancer, diabetes and Alzheimer's disease. The present study aimed to investigate the regulatory role of the MAPK signaling pathway in early-stage OA. A rabbit model of early-stage OA was induced by treatment with the enzyme papain. U0126 [an extracellular signal-regulated kinase (ERK) inhibitor], SP600125 [a Jun NH2-terminal kinase (JNK) inhibitor] and SB203580 (a p38 inhibitor) were administered to the rabbits via intra-articular injection. The severity of OA was assessed by histological examination using H&E, toluidine blue and safranin-O/fast green staining, as well by analyzing the glycosaminoglycan (GAG) content and determining the OA Research Society International (OARSI) score. Western blotting was used to detect the protein expression levels of matrix metalloproteinase-3 (MMP3), ERK, phosphorylated (p)-ERK, p38, p-p38, JNK, p-JNK, Beclin1, UNC-51-like kinase 1 (ULK1) and microtubule-associated protein 1 light chain 3 (LC3)II/I. U0126, SP600125 or SB203580 treatment significantly decreased the OARSI scores and significantly increased the GAG levels in the cartilaginous tissues of OA model rabbits. These results indicated that the MAPK inhibitors reduced the severity of OA-induced injury at the early stage. Western blotting results demonstrated that MAPK inhibition significantly decreased the protein expression levels of MMP3 in OA cartilage. The protective effect of MAPK inhibitors in OA was mediated via the activation of autophagy, as demonstrated by the increased protein expression levels of LC3II/I, ULK1 and Beclin1. Overall, the data indicated that MAPK inhibitors may exert a protective effect against OA by restoring compromised autophagy. Furthermore, the present study suggested that MAPK inhibitors may represent a potential pharmacological strategy for treating OA in the future.
Collapse
Affiliation(s)
- Chun-Na Lan
- Department of Rehabilitation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Wei-Jun Cai
- Department of Histology and Embryology, School of Basic Medicine, Central South University, Changsha, Hunan 410013, P.R. China
| | - Jie Shi
- Department of General Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Zhong-Jie Yi
- Department of Burn and Plastic Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
11
|
Zhou Z, Zhao J, Liu Y, Yan X, Sun H, Xia M, Su J. Autophagy promotes invadopodia formation in human ovarian cancer cells via the p62-extracellular signal-regulated kinase 1/2 pathway. Exp Ther Med 2021; 22:952. [PMID: 34335894 PMCID: PMC8290436 DOI: 10.3892/etm.2021.10384] [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: 12/26/2019] [Accepted: 05/21/2021] [Indexed: 11/18/2022] Open
Abstract
Invasiveness and metastatic potential are among the most essential characteristics of malignant tumors. Furthermore, it has been reported that autophagy and invasion are enhanced when tumor cells are grown in adverse conditions, such as nutritional deficiency and starvation. However, the association between autophagy and invasion remains largely unclear. In the present study, Earle's balanced salt solution (EBSS) was used to induce autophagy and an autophagy inhibitor was used to block autophagy. The results of Transwell assays revealed that autophagy inhibition limited the invasiveness of human ovarian cancer cells. Furthermore, the results of invadopodia formation assay indicated that autophagy stimulated invadopodia formation, and the selective autophagy receptor and signaling adaptor, sequestosome-1 (SQSTM1/p62 or simply p62), was closely associated with invadopodia formation in human ovarian cancer SKOV3 cells. The results of western blot analysis indicated that autophagy induced changes in p62 protein levels and p62 then functioned as a negative regulator of extracellular signal-regulated kinase 1/2 (ERK1/2) activity and invadopodia formation. The interaction between autophagy and invasion may thus be a self-protective mechanism for tumor cells in an unfavorable environment of nutritional deficiency, that maintains their survival and leads to increased invasiveness. An exploration of the intrinsic link between autophagy and invasion may provide a novel theoretical basis to reverse the resistance of tumor cells to a nutritional deficient environment.
Collapse
Affiliation(s)
- Zizhen Zhou
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, Basic College of Medicine, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jia Zhao
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, Basic College of Medicine, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yanan Liu
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, Basic College of Medicine, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xiaoyu Yan
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, Basic College of Medicine, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Hongyu Sun
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, Basic College of Medicine, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Meihui Xia
- Department of Obstetrics and Gynecology, The First Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jing Su
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, Basic College of Medicine, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
12
|
Zhao H, Huang C, Luo Y, Yao X, Hu Y, Wang M, Chen X, Zeng J, Hu W, Wang J, Li R, Yao X. A Correlation Study of Prognostic Risk Prediction for Colorectal Cancer Based on Autophagy Signature Genes. Front Oncol 2021; 11:595099. [PMID: 34168974 PMCID: PMC8218632 DOI: 10.3389/fonc.2021.595099] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 04/26/2021] [Indexed: 01/07/2023] Open
Abstract
Autophagy plays a complex role in tumors, sometimes promoting cancer cell survival and sometimes inducing apoptosis, and its role in the colorectal tumor microenvironment is controversial. The purpose of this study was to investigate the prognostic value of autophagy-related genes (ARGs) in colorectal cancer. We identified 37 differentially expressed autophagy-related genes by collecting TCGA colorectal tumor transcriptome data. A single-factor COX regression equation was used to identify 11 key prognostic genes, and a prognostic risk prediction model was constructed based on multifactor COX analysis. We classified patients into high and low risk groups according to prognostic risk parameters (p <0.001) and determined the prognostic value they possessed by survival analysis and the receiver operating characteristic (ROC) curve in the training and test sets of internal tests. In a multifactorial independent prognostic analysis, this risk value could be used as an independent prognostic indicator (HR=1.167, 95% CI=1.078-1.264, P<0.001) and was a robust predictor without any staging interference. To make it more applicable to clinical procedures, we constructed nomogram based on risk parameters and parameters of key clinical characteristics. The area under ROC curve for 3-year and 5-year survival rates were 0.735 and 0.718, respectively. These will better enable us to monitor patient prognosis, thus improve patient outcomes.
Collapse
Affiliation(s)
- Haibi Zhao
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China.,Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Ganzhou Hospital (Ganzhou Municipal Hospital), Guangdong Provincial People's Hospital, Ganzhou, China
| | - Chengzhi Huang
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Ganzhou Hospital (Ganzhou Municipal Hospital), Guangdong Provincial People's Hospital, Ganzhou, China.,School of Medicine, South China University of Technology, Guangzhou, China
| | - Yuwen Luo
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Ganzhou Hospital (Ganzhou Municipal Hospital), Guangdong Provincial People's Hospital, Ganzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Xiaoya Yao
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China.,Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Ganzhou Hospital (Ganzhou Municipal Hospital), Guangdong Provincial People's Hospital, Ganzhou, China
| | - Yong Hu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China.,Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Ganzhou Hospital (Ganzhou Municipal Hospital), Guangdong Provincial People's Hospital, Ganzhou, China
| | - Muqing Wang
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Ganzhou Hospital (Ganzhou Municipal Hospital), Guangdong Provincial People's Hospital, Ganzhou, China.,School of Medicine, South China University of Technology, Guangzhou, China
| | - Xin Chen
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Ganzhou Hospital (Ganzhou Municipal Hospital), Guangdong Provincial People's Hospital, Ganzhou, China.,Medical College, Shantou University, Shantou, China
| | - Jun Zeng
- Department of General Surgery, Baoan Central Hospital, The Fifth Affiliated Hospital of Shen Zhen University, Shen Zhen, China
| | - Weixian Hu
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Junjiang Wang
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,School of Medicine, South China University of Technology, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Rongjiang Li
- Department of General Surgery, Baoan Central Hospital, The Fifth Affiliated Hospital of Shen Zhen University, Shen Zhen, China
| | - Xueqing Yao
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China.,Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Ganzhou Hospital (Ganzhou Municipal Hospital), Guangdong Provincial People's Hospital, Ganzhou, China.,School of Medicine, South China University of Technology, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Medical College, Shantou University, Shantou, China
| |
Collapse
|
13
|
Chen J, Teng D, Wu Z, Li W, Feng Y, Tang Y, Liu G. Insights into the Molecular Mechanisms of Liuwei Dihuang Decoction via Network Pharmacology. Chem Res Toxicol 2020; 34:91-102. [PMID: 33332098 DOI: 10.1021/acs.chemrestox.0c00359] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The traditional Chinese medicines (TCMs) have been used to treat diseases over a long history, but it is still a great challenge to uncover the underlying mechanisms for their therapeutic effects due to the complexity of their ingredients. Based on a novel network pharmacology-based approach, we explored in this study the potential therapeutic targets of Liuwei Dihuang (LWDH) decoction in its neuroendocrine immunomodulation (NIM) function. We not only collected the known targets of the compounds in LWDH but also predicted the targets for these compounds using the balanced substructure-drug-target network-based inference (bSDTNBI), which is a target prediction method based on network inferring developed by our laboratory. A "target-(pathway)-target" (TPT) network, in which targets of LWDH were connected by relevant pathways, was constructed and divided into several separate modules with strong internal connections. Then the target module that contributes the most to NIM function was determined through a contribution scoring algorithm. Finally, the targets with the highest contribution score to NIM-related diseases in this target module were recommended as potential therapeutic targets of LWDH.
Collapse
Affiliation(s)
- Jianhui Chen
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Dan Teng
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Zengrui Wu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Weihua Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yuqian Feng
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yun Tang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Guixia Liu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
14
|
Shen F, Ge C, Yuan P. Aloe-emodin induces autophagy and apoptotic cell death in non-small cell lung cancer cells via Akt/mTOR and MAPK signaling. Eur J Pharmacol 2020; 886:173550. [DOI: 10.1016/j.ejphar.2020.173550] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 08/27/2020] [Accepted: 09/09/2020] [Indexed: 01/03/2023]
|
15
|
Han L, Ma Q, Yu J, Gong Z, Ma C, Xu Y, Deng G, Wu X. Autophagy plays a protective role during Pseudomonas aeruginosa-induced apoptosis via ROS-MAPK pathway. Innate Immun 2020; 26:580-591. [PMID: 32878509 PMCID: PMC7556189 DOI: 10.1177/1753425920952156] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 07/18/2020] [Accepted: 08/03/2020] [Indexed: 12/13/2022] Open
Abstract
Pseudomonas aeruginosa infection can induce alveolar macrophage apoptosis and autophagy, which play a vital role in eliminating pathogens. These two processes are usually not independent. Recently, autophagy has been found to interact with apoptosis during pathogen infections. Nevertheless, the role of autophagy in P. aeruginosa-infected cell apoptosis is unclear. In this study, we explored the impact of P. aeruginosa infection on autophagy and apoptosis in RAW264.7 cells. The autophagy activator rapamycin was used to stimulate autophagy and explore the role of autophagy on apoptosis in P. aeruginosa-infected RAW264.7 cells. The results indicated that P. aeruginosa infection induced autophagy and apoptosis in RAW264.7 cells, and that rapamycin could suppress P. aeruginosa-induced apoptosis by regulating the expression of apoptosis-related proteins. In addition, rapamycin scavenged the cellular reactive oxygen species (ROS) and diminished p-JNK, p-ERK1/2 and p-p38 expression of MAPK pathways in RAW264.7 cells infected with P. aeruginosa. In conclusion, the promotion of autophagy decreased P. aeruginosa-induced ROS accumulation and further attenuated the apoptosis of RAW264.7 cells through MAPK pathway. These results provide novel insights into host-pathogen interactions and highlight a potential role of autophagy in eliminating P. aeruginosa.
Collapse
Affiliation(s)
- Lu Han
- Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in western China, NingXia University, NingXia, Yinchuan, China
- School of Life Science, NingXia University, NingXia, Yinchuan, China
| | - Qinmei Ma
- Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in western China, NingXia University, NingXia, Yinchuan, China
- School of Life Science, NingXia University, NingXia, Yinchuan, China
| | - Jialin Yu
- Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in western China, NingXia University, NingXia, Yinchuan, China
- School of Life Science, NingXia University, NingXia, Yinchuan, China
| | - Zhaoqian Gong
- Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in western China, NingXia University, NingXia, Yinchuan, China
- School of Life Science, NingXia University, NingXia, Yinchuan, China
| | - Chenjie Ma
- Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in western China, NingXia University, NingXia, Yinchuan, China
- School of Life Science, NingXia University, NingXia, Yinchuan, China
| | - Yanan Xu
- Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in western China, NingXia University, NingXia, Yinchuan, China
- School of Life Science, NingXia University, NingXia, Yinchuan, China
| | - Guangcun Deng
- Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in western China, NingXia University, NingXia, Yinchuan, China
- School of Life Science, NingXia University, NingXia, Yinchuan, China
| | - Xiaoling Wu
- Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in western China, NingXia University, NingXia, Yinchuan, China
- School of Life Science, NingXia University, NingXia, Yinchuan, China
| |
Collapse
|
16
|
Liao Z, Zhang X, Song C, Lin W, Cheng Y, Xie Z, Chen S, Nie Y, Li A, Zhang H, Li H, Li H, Xie Q. ALV-J inhibits autophagy through the GADD45β/MEKK4/P38MAPK signaling pathway and mediates apoptosis following autophagy. Cell Death Dis 2020; 11:684. [PMID: 32826872 PMCID: PMC7442830 DOI: 10.1038/s41419-020-02841-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 07/29/2020] [Accepted: 07/29/2020] [Indexed: 02/06/2023]
Abstract
Autophagy and apoptosis, which are important processes for host immunity, are commonly exploited by viruses to facilitate their survival. However, to the best of our knowledge, very few studies have researched the mechanisms of action of the autophagic and apoptotic signaling pathways following viral infection. Thus, the present study aimed to investigate the mechanisms of action of growth arrest and DNA-damage-inducible β (GADD45β), an important resistance gene involved in the host resistance to ALV-J. Both ALV-J infection and the overexpression of GADD45β inhibited autophagy during the early stages, which prevented the autophagosomes from binding to the lysosomes and resulted in an incomplete autophagic flux. Notably, GADD45β was discovered to interact with MEKK4 in DF-1 cells. The genetic knockdown of GADD45β and MEKK4 using small interfering RNA-affected ALV-J infection, which suggested that ALV-J may promote the binding of GADD45β to MEKK4 to activate the p38MAPK signaling pathway, which subsequently inhibits autophagy. Furthermore, ALV-J was revealed to affect the autophagic pathway prior to affecting the apoptotic pathway. In conclusion, to the best of our knowledge, the present study was the first to investigate the combined effects of ALV-J infection on autophagy and apoptosis, and to suggest that ALV-J inhibits autophagy via the GADD45β/MEKK4/p38MAPK signaling pathway.
Collapse
Affiliation(s)
- Zhihong Liao
- Lingnan Guangdong Laboratory of Modern Agriculture & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 510642, Guangzhou, PR China
- Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangdong, 510642, Guangzhou, PR China
| | - Xinheng Zhang
- Lingnan Guangdong Laboratory of Modern Agriculture & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 510642, Guangzhou, PR China
- Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangdong, 510642, Guangzhou, PR China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, 510642, Guangzhou, PR China
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, 510642, Guangzhou, PR China
| | - Cailiang Song
- Lingnan Guangdong Laboratory of Modern Agriculture & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 510642, Guangzhou, PR China
- Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangdong, 510642, Guangzhou, PR China
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, 510642, Guangzhou, PR China
| | - Wencheng Lin
- Lingnan Guangdong Laboratory of Modern Agriculture & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 510642, Guangzhou, PR China
- Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangdong, 510642, Guangzhou, PR China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, 510642, Guangzhou, PR China
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, 510642, Guangzhou, PR China
| | - Yuzhen Cheng
- Lingnan Guangdong Laboratory of Modern Agriculture & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 510642, Guangzhou, PR China
| | - Zi Xie
- Lingnan Guangdong Laboratory of Modern Agriculture & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 510642, Guangzhou, PR China
- Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangdong, 510642, Guangzhou, PR China
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, 510642, Guangzhou, PR China
| | - Sheng Chen
- Lingnan Guangdong Laboratory of Modern Agriculture & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 510642, Guangzhou, PR China
- Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangdong, 510642, Guangzhou, PR China
| | - Yu Nie
- Lingnan Guangdong Laboratory of Modern Agriculture & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 510642, Guangzhou, PR China
- Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangdong, 510642, Guangzhou, PR China
| | - Aijun Li
- College of Science and Engineering, Jinan University, 510632, Guangzhou, PR China
| | - Huanmin Zhang
- USDA, Agriculture Research Service, Avian Disease and Oncology Laboratory, East Lansing, MI, 48823, USA
| | - Hongxin Li
- Lingnan Guangdong Laboratory of Modern Agriculture & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 510642, Guangzhou, PR China
- Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangdong, 510642, Guangzhou, PR China
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, 510642, Guangzhou, PR China
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, 510642, Guangzhou, PR China
| | - Haiyun Li
- Lingnan Guangdong Laboratory of Modern Agriculture & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 510642, Guangzhou, PR China
| | - Qingmei Xie
- Lingnan Guangdong Laboratory of Modern Agriculture & Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, 510642, Guangzhou, PR China.
- Key Laboratory of Animal Health Aquaculture and Environmental Control, Guangdong, 510642, Guangzhou, PR China.
- South China Collaborative Innovation Center for Poultry Disease Control and Product Safety, 510642, Guangzhou, PR China.
- Guangdong Engineering Research Center for Vector Vaccine of Animal Virus, 510642, Guangzhou, PR China.
| |
Collapse
|
17
|
Insulin-Like Growth Factor I Prevents Cellular Aging via Activation of Mitophagy. J Aging Res 2020; 2020:4939310. [PMID: 32802505 PMCID: PMC7416301 DOI: 10.1155/2020/4939310] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 05/22/2020] [Accepted: 07/06/2020] [Indexed: 12/18/2022] Open
Abstract
Mitochondrial dysfunction is a hallmark of cellular aging. Mitophagy is a critical mitochondrial quality control mechanism that removes dysfunctional mitochondria and contributes to cell survival. Insulin-like growth factor 1 (IGF-1) promotes survival of smooth muscle cells (SMCs), but its potential effect on cellular aging is unknown yet. We found that IGF-1 decreased cell senescence, prevented DNA telomere shortening, increased mitochondrial membrane potential, activated cytochrome C oxidase, and reduced mitochondrial DNA damage in long-term cultured (aged) aortic SMC, suggesting an antiaging effect. IGF-1 increased mitophagy in aged cells, and this was associated with decreased expression of cyclin-dependent kinase inhibitors p16 and p21 and elevated levels of Nrf2 and Sirt3, regulators of mitophagy and mitochondrial biogenesis. SiRNA-induced inhibition of either Nrf2 or Sirt3 blocked IGF-1-induced upregulation of mitophagy, suggesting that the Nrf2/Sirt3 pathway was required for IGF-1's effect on mitophagy. PINK1 is a master regulator of mitophagy. PINK1 silencing suppressed mitophagy and inhibited IGF-1-induced antiaging effects in aged SMC, consistent with an essential role of mitophagy in IGF-1's effect on cellular aging. Thus, IGF-1 inhibited cellular aging via Nrf2/Sirt3-dependent activation of mitophagy. Our data suggest that activation of IGF-1 signaling is a novel potential strategy to activate mitophagy and slow cellular aging.
Collapse
|
18
|
Xiang H, Zhang J, Lin C, Zhang L, Liu B, Ouyang L. Targeting autophagy-related protein kinases for potential therapeutic purpose. Acta Pharm Sin B 2020; 10:569-581. [PMID: 32322463 PMCID: PMC7161711 DOI: 10.1016/j.apsb.2019.10.003] [Citation(s) in RCA: 157] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 08/06/2019] [Accepted: 09/09/2019] [Indexed: 02/08/2023] Open
Abstract
Autophagy, defined as a scavenging process of protein aggregates and damaged organelles mediated by lysosomes, plays a significant role in the quality control of macromolecules and organelles. Since protein kinases are integral to the autophagy process, it is critically important to understand the role of kinases in autophagic regulation. At present, intervention of autophagic processes by small-molecule modulators targeting specific kinases has becoming a reasonable and prevalent strategy for treating several varieties of human disease, especially cancer. In this review, we describe the role of some autophagy-related kinase targets and kinase-mediated phosphorylation mechanisms in autophagy regulation. We also summarize the small-molecule kinase inhibitors/activators of these targets, highlighting the opportunities of these new therapeutic agents.
Collapse
Key Words
- 4E-BP1, eukaryotic translation initiation factor 4E-binding protein
- AKT1, AKT serine/threonine kinase 1
- AMBRA1, autophagy/beclin-1 regulator 1
- AMPK, AMP-activated protein kinase
- ARF, auxin response factor gene
- ATG, autophagy-related protein
- Autophagy
- Autophagy-related kinase
- CaMKK2, calcium/calmodulin-dependent protein kinase kinase 2
- DAPK, death associated protein kinase
- FIP200, FAK family kinase-interacting protein of 200 kDa
- GAP, GTPase-activating protein
- GO, gene ontology
- GSK3α, glycogen synthase kinase 3 alpha
- HMGB1, high mobility group protein B1
- Human disease therapy
- JNK1, C-Jun N-terminal kinase
- LC3, microtubule-associated protein 1 light chain 3
- LKB1, serine/threonine-protein kinase stk11
- LPS, lipopolysaccharide
- LRRK2, leucine rich repeat kinase 2
- PD, Parkinson's disease
- PI, phosphatidylinositol
- PI3 kinase, phosphoinositide 3-kinase
- PI3P, phosphatidylinositol triphosphate
- PIM2, proviral insertion in murine lymphomas 2
- PINK1, PTEN-induced putative kinase 1
- PIP2, phosphatidylinositol-4,5-bisphosphate
- PKACα, a protein kinase cAMP-activated catalytic subunit alpha
- PKCα, protein kinase C alpha type
- PKD1, polycystin-1
- PPIs, protein–protein interactions
- PROTAC, proteolysis targeting chimeras
- PTMs, post-translational modifications
- Phosphorylation
- Protein kinases
- Rheb, the RAS homolog enriched in brain
- Small-molecule kinase inhibitors/activators
- TAK1, transforming growth factor activated kinase-1
- TFEB, transcription factor EB
- TNBC, triple-negative breast cancer
- TSC1/2, tuberous sclerosis complex proteins 1/2
- ULK complex, ULK1–mATG13–FIP200–ATG101 complex
- ULK1, unc-51-like kinase 1
- UVRAG, ultraviolet resistance-associated gene
- mTOR, mammalian target of rapamycin
- mTORC1, mammalian target of rapamycin complex 1
Collapse
Affiliation(s)
- Honggang Xiang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Jifa Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Congcong Lin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Lan Zhang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Bo Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Liang Ouyang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, China
| |
Collapse
|
19
|
Lotsberg ML, Wnuk-Lipinska K, Terry S, Tan TZ, Lu N, Trachsel-Moncho L, Røsland GV, Siraji MI, Hellesøy M, Rayford A, Jacobsen K, Ditzel HJ, Vintermyr OK, Bivona TG, Minna J, Brekken RA, Baguley B, Micklem D, Akslen LA, Gausdal G, Simonsen A, Thiery JP, Chouaib S, Lorens JB, Engelsen AST. AXL Targeting Abrogates Autophagic Flux and Induces Immunogenic Cell Death in Drug-Resistant Cancer Cells. J Thorac Oncol 2020; 15:973-999. [PMID: 32018052 PMCID: PMC7397559 DOI: 10.1016/j.jtho.2020.01.015] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 12/29/2019] [Accepted: 01/19/2020] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Acquired cancer therapy resistance evolves under selection pressure of immune surveillance and favors mechanisms that promote drug resistance through cell survival and immune evasion. AXL receptor tyrosine kinase is a mediator of cancer cell phenotypic plasticity and suppression of tumor immunity, and AXL expression is associated with drug resistance and diminished long-term survival in a wide range of malignancies, including NSCLC. METHODS We aimed to investigate the mechanisms underlying AXL-mediated acquired resistance to first- and third-generation small molecule EGFR tyrosine kinase inhibitors (EGFRi) in NSCLC. RESULTS We found that EGFRi resistance was mediated by up-regulation of AXL, and targeting AXL reduced reactivation of the MAPK pathway and blocked onset of acquired resistance to long-term EGFRi treatment in vivo. AXL-expressing EGFRi-resistant cells revealed phenotypic and cell signaling heterogeneity incompatible with a simple bypass signaling mechanism, and were characterized by an increased autophagic flux. AXL kinase inhibition by the small molecule inhibitor bemcentinib or siRNA mediated AXL gene silencing was reported to inhibit the autophagic flux in vitro, bemcentinib treatment blocked clonogenicity and induced immunogenic cell death in drug-resistant NSCLC in vitro, and abrogated the transcription of autophagy-associated genes in vivo. Furthermore, we found a positive correlation between AXL expression and autophagy-associated gene signatures in a large cohort of human NSCLC (n = 1018). CONCLUSION Our results indicate that AXL signaling supports a drug-resistant persister cell phenotype through a novel autophagy-dependent mechanism and reveals a unique immunogenic effect of AXL inhibition on drug-resistant NSCLC cells.
Collapse
Affiliation(s)
- Maria L Lotsberg
- Centre for Cancer Biomarkers CCBIO, University of Bergen, Bergen, Norway; Department of Biomedicine, University of Bergen, Bergen, Norway; Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Katarzyna Wnuk-Lipinska
- Department of Biomedicine, University of Bergen, Bergen, Norway; BerGenBio ASA, Bergen, Norway
| | - Stéphane Terry
- INSERM UMR 1186, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Tuan Zea Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Ning Lu
- Centre for Cancer Biomarkers CCBIO, University of Bergen, Bergen, Norway; Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Laura Trachsel-Moncho
- Department of Molecular Medicine, Institute of Basic Medical Sciences and Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Gro V Røsland
- Department of Biomedicine, University of Bergen, Bergen, Norway; Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway
| | | | | | - Austin Rayford
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Kirstine Jacobsen
- Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Henrik J Ditzel
- Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark; Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Olav K Vintermyr
- Department of Pathology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Trever G Bivona
- Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - John Minna
- Hamon Center for Therapeutic Oncology Research, Simmons Comprehensive Cancer Center, Departments of Surgery, Pharmacology and Internal Medicine, UT Southwestern Medical Center, Dallas, Texas
| | - Rolf A Brekken
- Centre for Cancer Biomarkers CCBIO, University of Bergen, Bergen, Norway; Hamon Center for Therapeutic Oncology Research, Simmons Comprehensive Cancer Center, Departments of Surgery, Pharmacology and Internal Medicine, UT Southwestern Medical Center, Dallas, Texas
| | - Bruce Baguley
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | | | - Lars A Akslen
- Centre for Cancer Biomarkers CCBIO, University of Bergen, Bergen, Norway; Department of Pathology, Haukeland University Hospital, Bergen, Norway; Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | | | - Anne Simonsen
- Department of Molecular Medicine, Institute of Basic Medical Sciences and Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Jean Paul Thiery
- Centre for Cancer Biomarkers CCBIO, University of Bergen, Bergen, Norway; INSERM UMR 1186, Gustave Roussy, Université Paris-Saclay, Villejuif, France; Cancer Science Institute of Singapore, National University of Singapore, Singapore; Biomedical Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, A-STAR, Singapore; Guangzhou Institutes of Biomedicine and Health, Guangzhou, People's Republic of China; Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, Hong Kong University, Hong Kong
| | - Salem Chouaib
- Department of Pathology, Haukeland University Hospital, Bergen, Norway; Thumbay Research Institute for Precision Medicine, GMU Ajman, United Arab Emirates
| | - James B Lorens
- Centre for Cancer Biomarkers CCBIO, University of Bergen, Bergen, Norway; Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Agnete Svendsen Tenfjord Engelsen
- Centre for Cancer Biomarkers CCBIO, University of Bergen, Bergen, Norway; Department of Biomedicine, University of Bergen, Bergen, Norway; INSERM UMR 1186, Gustave Roussy, Université Paris-Saclay, Villejuif, France.
| |
Collapse
|
20
|
Jin S. The Cross-Regulation Between Autophagy and Type I Interferon Signaling in Host Defense. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1209:125-144. [PMID: 31728868 DOI: 10.1007/978-981-15-0606-2_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The production of type I interferons (IFNs) is one of the hallmarks of intracellular antimicrobial program. Typical type I IFN response activates the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway, which results in the transcription of plentiful IFN-stimulated genes (ISGs) to establish the comprehensive antiviral states. Type I IFN signaling should initiate timely to provoke innate and adaptive immune responses for effective elimination of the invading pathogens. Meanwhile, a precise control must come on the stage to restrain the persistent activation of type I IFN responses to avoid attendant toxicity. Autophagy, a conserved eukaryotic degradation system, mediated by a number of autophagy-related (ATG) proteins, plays an essential role in the clearance of invading microorganism and manipulation of type I responses. Autophagy modulates type I IFN responses through regulatory integration with innate immune signaling pathways, and by removing endogenous ligands of innate immune sensors. Moreover, selective autophagy governs the choice of innate immune factors as specific cargoes for degradation, thus tightly monitoring the type I IFN responses. This review will focus on the cross-regulation between autophagy and type I IFN signaling in host defense.
Collapse
Affiliation(s)
- Shouheng Jin
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510006, Guangdong, China.
| |
Collapse
|
21
|
Zhang Y, Li J, Wang S, Yang F, Zhou Y, Liu Y, Zhu W, Shi X. HBx‑associated long non‑coding RNA activated by TGF‑β promotes cell invasion and migration by inducing autophagy in primary liver cancer. Int J Oncol 2019; 56:337-347. [PMID: 31746419 DOI: 10.3892/ijo.2019.4908] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 03/07/2019] [Indexed: 12/25/2022] Open
Abstract
Hepatitis B virus (HBV) x protein (HBx) has been reported as the primary pathogenic factor involved in HBV‑related liver cancer; however, the mechanisms underlying how HBx promotes tumor‑associated invasion and metastasis remain unclear. Long noncoding RNA activated by transforming growth factor (TGF)‑β (lncRNA‑ATB) is a novel oncogenic lncRNA stimulated by TGF‑β, which is closely associated with the invasion and metastasis of liver cancer. In the present study, whether lncRNA‑ATB was involved in HBx‑mediated hepatocarcinogenesis was investigated. The expression of lncRNA‑ATB in 26 primary liver cancer tissues and lentivirus transfected HBx‑HepG2 cell lines was detected, and it was revealed that more advanced tumor‑node‑metastasis staging and increased expression of lncRNA‑ATB in liver cancer tissues were significantly associated with HBV infection. It was further demonstrated that the expression levels of lncRNA‑ATB and TGF‑β were elevated in HepG2 cells following HBx‑vector transfection, which was accompanied with increased autophagy. Conversely, knockdown of lncRNA‑ATB or TGF‑β could suppress this effect. Furthermore, such suppression on autophagy in HepG2 cells could be alleviated by the induction of starvation. In addition, the invasive and migration abilities of HBx‑HepG2 cells were increased compared with HepG2 cells, while knockdown of lncRNA‑ATB or TGF‑β could reduce these abilities. In conclusion, the results of the present study revealed that HBx was closely associated with oncogenic lncRNA‑ATB. HBx‑induced autophagy could upregulate the expression of TGF‑β and lncRNA‑ATB. This may be considered to be a potential mechanism underlying HBV‑induced hepatocarcinogenesis.
Collapse
Affiliation(s)
- Yuheng Zhang
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Jun Li
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Shuai Wang
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Faji Yang
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Yuan Zhou
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Yang Liu
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Wei Zhu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| | - Xiaolei Shi
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, P.R. China
| |
Collapse
|
22
|
Lu J, Shan J, Liu N, Ding Y, Wang P. Tanshinone IIA Can Inhibit Angiotensin II-Induced Proliferation and Autophagy of Vascular Smooth Muscle Cells via Regulating the MAPK Signaling Pathway. Biol Pharm Bull 2019; 42:1783-1788. [PMID: 31391347 DOI: 10.1248/bpb.b19-00053] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To examine the effect of tanshinone IIA on Angiotensin II (Ang II)-induced proliferation and autophagy in vascular smooth muscle cells (VSMCs) and the related mechanism. VSMCs were treated with Ang II with or without tanshinone IIA (1, 5 and 10 µg/mL), and the proliferation, apoptosis in cells with different treatment were examined by methylthiazolyl tetrazolium (MTT) and flow cytometry methods. Moreover, the expression of autophagy related proteins and mitogen-activated protein kinase (MAPK) signaling molecules were examined by RT-quantitative (q)PCR and Western blot methods. Ang II induced significantly increase in the proliferation and autophagy of VSMCs, and the MAPK signaling was activated. Tanshinone IIA can attenuate Ang II-induced effects via down-regulating the MAPK signaling pathway. Tanshinone IIA can inhibit Ang II-induced proliferation and autophagy of VSMCs via regulating the MAPK signaling pathway.
Collapse
Affiliation(s)
- Jingping Lu
- Department of Cardiology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine
| | - Jinjun Shan
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatics, Affiliated Hospital of Nanjing University of Chinese Medicine
| | - Ning Liu
- Department of Cardiology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine
| | - Yao Ding
- Department of Cardiology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine
| | - Pei Wang
- Department of Cardiology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine
| |
Collapse
|
23
|
Wen J, Xu B, Sun Y, Lian M, Li Y, Lin Y, Chen D, Diao Y, Almoiliqy M, Wang L. Paeoniflorin protects against intestinal ischemia/reperfusion by activating LKB1/AMPK and promoting autophagy. Pharmacol Res 2019; 146:104308. [PMID: 31181335 DOI: 10.1016/j.phrs.2019.104308] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 05/09/2019] [Accepted: 06/06/2019] [Indexed: 12/18/2022]
Abstract
Intestinal ischemia-reperfusion (I/R) injury is a common pathological process with high clinical morbidity and mortality. Paeoniflorin, a monoterpene glucoside, is found to have diverse health beneficial effects including autophagy modulation, anti-inflammatory, anti-apoptotic, and anti-oxidative effects. Based on our pre-experiments, we proposed that paeoniflorin could ameliorate intestinal I/R injury and restore autophagy through activating LKB1/AMPK signal pathway. Our proposal was verified using rat intestinal I/R model in vivo and intestinal epithelial cell line (IEC-6 cells) hypoxia/reoxygenation (H/R) model in vitro. Our results showed that paeoniflorin pretreatment exerted protective effects in rat intestinal I/R injury by reducing intestinal morphological damage, inflammation, oxidative stress, and apoptosis. Paeoniflorin restored H/R-impaired autophagy flux by up-regulating autophagy-related protein p62/SQSTM1 degradation, LC3II and beclin-1 expression, and autophagosomes synthesis without significantly affecting control IEC-6 cells. Paeoniflorin pretreatment significantly activated LKB1/AMPK signaling pathway by reversing the decreased LKB1 and AMPK phosphorylation without affecting total LKB1 both in vivo and in vitro. LKB1 knockdown reduced AMPK phosphorylation, suppressed LC3II and Beclin-1 level, and decreased the degradation of SQSTM/p62, and the knockdown weakened the effects of paeoniflorin in restoring the impaired autophagy flux in H/R injured IEC-6 cells, suggesting that paeoniflorin mitigated the intestinal I/R-impaired autophagy flux by activating LKB1/AMPK signaling pathway. Our study may provide valuable information for further studies.
Collapse
Affiliation(s)
- Jin Wen
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Bin Xu
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Yuchao Sun
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Mengqiao Lian
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Yanli Li
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Yuan Lin
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China.
| | - Dapeng Chen
- Laboratory Animal Center, Dalian Medical University, Dalian, 116044, China
| | - Yunpeng Diao
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China.
| | - Marwan Almoiliqy
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Li Wang
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
| |
Collapse
|
24
|
Tian Y, Wang ML, Zhao J. Crosstalk between Autophagy and Type I Interferon Responses in Innate Antiviral Immunity. Viruses 2019; 11:v11020132. [PMID: 30717138 PMCID: PMC6409909 DOI: 10.3390/v11020132] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/25/2019] [Accepted: 01/28/2019] [Indexed: 12/25/2022] Open
Abstract
Autophagy exhibits dual effects during viral infections, promoting the clearance of viral components and activating the immune system to produce antiviral cytokines. However, some viruses impair immune defenses by collaborating with autophagy. Mounting evidence suggests that the interaction between autophagy and innate immunity is critical to understanding the contradictory roles of autophagy. Type I interferon (IFN-I) is a crucial antiviral factor, and studies have indicated that autophagy affects IFN-I responses by regulating IFN-I and its receptors expression. Similarly, IFN-I and interferon-stimulated gene (ISG) products can harness autophagy to regulate antiviral immunity. Crosstalk between autophagy and IFN-I responses could be a vital aspect of the molecular mechanisms involving autophagy in innate antiviral immunity. This review briefly summarizes the approaches by which autophagy regulates antiviral IFN-I responses and highlights the recent advances on the mechanisms by which IFN-I and ISG products employ autophagy against viruses.
Collapse
Affiliation(s)
- Yu Tian
- Department of Microbiology, Anhui Medical University, Hefei 230032, China.
| | - Ming-Li Wang
- Department of Microbiology, Anhui Medical University, Hefei 230032, China.
- Wuhu Interferon Bio-Products Industry Research Institute Co., Ltd., Wuhu 241000, China.
| | - Jun Zhao
- Department of Microbiology, Anhui Medical University, Hefei 230032, China.
- Wuhu Interferon Bio-Products Industry Research Institute Co., Ltd., Wuhu 241000, China.
| |
Collapse
|
25
|
Wang R, Wang G. Protein Modification and Autophagy Activation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1206:237-259. [PMID: 31776989 DOI: 10.1007/978-981-15-0602-4_12] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Protein modification refers to the chemical modification of proteins after their biosynthesis, which is also called posttranslational modification (PTM). PTM causes changes in protein properties and functions. PTM includes an attachment of addition of functional groups, such as methylation, acetylation, glycosylation and phosphorylation; a covalent coupling of small peptides or proteins, such as ubiquitination and SUMOylation; or chemical changes in amino acids, such as citrullination (conversion of arginine to citrulline). Protein modification plays an important role in cellular processes. Since a protein can be modified in different ways, such as acetylation, methylation and phosphorylation, the functions of proteins are different under different modification states. Moreover, the same modification at different sites may have completely different effects on protein function. For example, phosphorylation at some sites in a protein may lead to a functional activation, while phosphorylation at other sites may cause an inhibition of the functions. Thus, different modifications, combinations and sites changes lead to different functional regulations of a protein, resulting in different effects in the cells. In autophagy, PTMs are widely involved in the regulation of autophagy, including ubiquitination, phosphorylation and acetylation. Ubiquitination is the covalent conjugation of ubiquitin to the substrates through a series of enzymes. Phosphorylation refers to an attachment of a phosphoryl group into a protein, primarily on serine, threonine and tyrosine, which is catalyzed by the kinases. Phosphorylation, a common modification, regulates protein function and localization. Phosphorylation in autophagy regulates the activity of autophagy-associated proteins and the initiation and progression of autophagy by regulating signaling pathways. Acetylation means the addition of acetyl groups onto lysine or N-terminal segment of target proteins through acetyltransferases. Acetylation and deacetylation are both involved in the regulation of autophagy initiation and selective autophagy by controlling the acetylation level of important proteins in the autophagy process. In this chapter, we will focus on the regulation of ubiquitination and phosphorylation in autophagy.
Collapse
Affiliation(s)
- Rui Wang
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Neuropsychiatric Diseases & Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Guanghui Wang
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Neuropsychiatric Diseases & Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China.
| |
Collapse
|