51
|
Morozevic GE, Kozlova NI, Gevorkian NM, Berman AE. [Integrin α3β1 signaling in regulation of the SK-Mel-147 melanoma cell senescence]. BIOMEDITSINSKAIA KHIMIIA 2022; 68:39-46. [PMID: 35221295 DOI: 10.18097/pbmc20226801039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Using a model of the human SK-Mel-147 melanoma cell line, it was shown that blocking the expression of integrin α3β1 by transduction of cells with α3-specific shRNA did not affect their proliferation, but sharply increased the proportion of SA-β-Gal-positive cells, a phenotypic feature of cell senescence. These findings were accompanied by a significant increase in the activity of the Akt and mTOR protein kinases and the expression of p53 and p21 oncosupressors. Pharmacological inhibition of mTORC1 reduced the number SA-β-Gal-positive cells in the SK-Mel-147 cell population depleted of α3β1. Based on our recent data on a non-canonical function of Akt isomers in the regulation of SK-Mel-147 cell senescence caused by deficiency of α2β1 receptor, we investigated the role of Akt isomers in senescence induced by the α3β1 knockdown. It appeared that in the cell population with downregulated α3β1, inhibition of Akt1 reduced the number SA-β-Gal positive cells to the level of control cell population, while inhibition of Akt2 had no visible effect. Our results demonstrate that the laminin-specific integrin α3β1, like the collagen-specific receptor α2β1, is involved in tumor cell protection from senescence, and senescence induced by α3β1 depletion, like that caused by α2β1 deficiency, is based on a signaling mechanism employing a non-canonical function of the Akt1 isoform.
Collapse
Affiliation(s)
| | - N I Kozlova
- Institute of Biomedical Chemistry, Moscow, Russia
| | | | - A E Berman
- Institute of Biomedical Chemistry, Moscow, Russia
| |
Collapse
|
52
|
Zhao Y, Liu X, Si F, Huang L, Gao A, Lin W, Hoft DF, Shao Q, Peng G. Citrate Promotes Excessive Lipid Biosynthesis and Senescence in Tumor Cells for Tumor Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2101553. [PMID: 34747157 PMCID: PMC8728847 DOI: 10.1002/advs.202101553] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 10/04/2021] [Indexed: 05/17/2023]
Abstract
Metabolic disorder is one of the hallmarks of cancers, and reprogramming of metabolism is becoming a novel strategy for cancer treatment. Citrate is a key metabolite and critical metabolic regulator linking glycolysis and lipid metabolism in cellular energy homeostasis. Here it is reported that citrate treatment (both sodium citrate and citric acid) significantly suppresses tumor cell proliferation and growth in various tumor types. Mechanistically, citrate promotes excessive lipid biosynthesis and induces disruption of lipid metabolism in tumor cells, resulting in tumor cell senescence and growth inhibition. Furthermore, ATM-associated DNA damage response cooperates with MAPK and mTOR signaling pathways to control citrate-induced tumor cell growth arrest and senescence. In vivo studies further demonstrate that citrate administration dramatically inhibits tumor growth and progression in a colon cancer xenograft model. Importantly, citrate administration combined with the conventional chemotherapy drugs exhibits synergistic antitumor effects in vivo in the colon cancer models. These results clearly indicate that citrate can reprogram lipid metabolism and cell fate in cancer cells, and targeting citrate can be a promising therapeutic strategy for tumor treatment.
Collapse
Affiliation(s)
- Yangjing Zhao
- Department of ImmunologyKey Laboratory of Medical Science and Laboratory Medicine of Jiangsu ProvinceSchool of MedicineJiangsu UniversityZhenjiang212013P. R. China
- Division of Infectious DiseasesAllergy & Immunology and Department of Internal MedicineSaint Louis University School of MedicineSaint LouisMO63104USA
| | - Xia Liu
- Division of Infectious DiseasesAllergy & Immunology and Department of Internal MedicineSaint Louis University School of MedicineSaint LouisMO63104USA
| | - Fusheng Si
- Division of Infectious DiseasesAllergy & Immunology and Department of Internal MedicineSaint Louis University School of MedicineSaint LouisMO63104USA
| | - Lan Huang
- Department of ImmunologyKey Laboratory of Medical Science and Laboratory Medicine of Jiangsu ProvinceSchool of MedicineJiangsu UniversityZhenjiang212013P. R. China
- Division of Infectious DiseasesAllergy & Immunology and Department of Internal MedicineSaint Louis University School of MedicineSaint LouisMO63104USA
| | - Aiqin Gao
- Division of Infectious DiseasesAllergy & Immunology and Department of Internal MedicineSaint Louis University School of MedicineSaint LouisMO63104USA
| | - Wenli Lin
- Division of Infectious DiseasesAllergy & Immunology and Department of Internal MedicineSaint Louis University School of MedicineSaint LouisMO63104USA
| | - Daniel F. Hoft
- Division of Infectious DiseasesAllergy & Immunology and Department of Internal MedicineSaint Louis University School of MedicineSaint LouisMO63104USA
- Department of Molecular Microbiology & ImmunologySaint Louis University School of MedicineSaint LouisMO63104USA
| | - Qixiang Shao
- Department of ImmunologyKey Laboratory of Medical Science and Laboratory Medicine of Jiangsu ProvinceSchool of MedicineJiangsu UniversityZhenjiang212013P. R. China
| | - Guangyong Peng
- Division of Infectious DiseasesAllergy & Immunology and Department of Internal MedicineSaint Louis University School of MedicineSaint LouisMO63104USA
- Department of Molecular Microbiology & ImmunologySaint Louis University School of MedicineSaint LouisMO63104USA
| |
Collapse
|
53
|
Jiang P, Li F, Liu Z, Hao S, Gao J, Li S. BTB and CNC homology 1 (Bach1) induces lung cancer stem cell phenotypes by stimulating CD44 expression. Respir Res 2021; 22:320. [PMID: 34949193 PMCID: PMC8697453 DOI: 10.1186/s12931-021-01918-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/17/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Growing evidence suggests that cancer stem cells (CSCs) are responsible for cancer initiation in tumors. Bach1 has been identified to contribute to several tumor progression, including lung cancer. The role of Bach1 in CSCs remains poorly known. Therefore, the function of Bach1 on lung CSCs was focused currently. METHODS The expression of Bach1, CD133, CD44, Sox2, Nanog and Oct4 mRNA was assessed using Real-Time Quantitative Reverse Transcription PCR (RT-qPCR). Protein expression of Bach1, CD133, CD44, Sox2, Nanog, Oct4, p53, BCL2, BAX, p-p38, p-AKT1, c-Fos and c-Jun protein was analyzed by western blotting. 5-ethynyl-29-deoxyuridine (EdU), colony formation, Flow cytometry analysis and transwell invasion assay were carried out to analyze lung cancer cell proliferation, apoptosis and invasion respectively. Tumor sphere formation assay was utilized to evaluate spheroid capacity. Flow cytometry analysis was carried out to isolate CD133 or CD44 positive lung cancer cells. The relationship between Bach1 and CD44 was verified using ChIP-qPCR and dual-luciferase reporter assay. Xenograft tumor tissues were collected for hematoxylin and eosin (HE) staining and IHC analysis to evaluate histology and Ki-67. RESULTS The ratio of CD44 + CSCs from A549 and SPC-A1 cells were significantly enriched. Tumor growth of CD44 + CSCs was obviously suppressed in vivo compared to CD44- CSCs. Bach1 expression was obviously increased in CD44 + CSCs. Then, via using the in vitro experiment, it was observed that CSCs proliferation and invasion were greatly reduced by the down-regulation of Bach1 while cell apoptosis was triggered by knockdown of Bach1. Loss of Bach1 was able to repress tumor-sphere formation and tumor-initiating CSC markers. A repression of CSCs growth and metastasis of shRNA-Bach1 was confirmed using xenograft models and caudal vein injection. The direct interaction between Bach1 and CD44 was confirmed by ChIP-qPCR and dual-luciferase reporter assay. Furthermore, mitogen-activated protein kinases (MAPK) signaling pathway was selected and we proved the effects of Bach1 on lung CSCs were associated with the activation of the MAPK pathway. As manifested, loss of Bach1 was able to repress p-p38, p-AKT1, c-Fos, c-Jun protein levels in lung CSCs. Inhibition of MAPK signaling remarkably restrained lung CSCs growth and CSCs properties induced by Bach1 overexpression. CONCLUSION In summary, we imply that Bach1 demonstrates great potential for the treatment of lung cancer metastasis and recurrence via activating CD44 and MPAK signaling.
Collapse
Affiliation(s)
- Pan Jiang
- Department of Nutrition, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China
| | - Fan Li
- Department of Nutrition, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China
| | - Zilong Liu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China
| | - Shengyu Hao
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China.
| | - Jian Gao
- Department of Nutrition, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China.
| | - Shanqun Li
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, China.
| |
Collapse
|
54
|
Min SN, Zhu MQ, Mao XD, Li W, Wei T, Mei M, Zhang Y, Wu LL, Yu GY, Cong X. The Contribution of the Interleukin-4-Induced Epithelial Cell Senescence to Glandular Fibrosis in IgG4-Related Sialadenitis. Arthritis Rheumatol 2021; 74:1070-1082. [PMID: 34927394 DOI: 10.1002/art.42052] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/17/2021] [Accepted: 12/14/2021] [Indexed: 01/10/2023]
Abstract
OBJECTIVE Immunoglobulin G4-related sialadenitis (IgG4-RS) is a chronic fibro-inflammatory disease characterized by glandular fibrosis and hyposalivation. Here we aimed to explore the role of cellular senescence in the pathogenesis of IgG4-RS-related fibrosis. METHODS The expression of senescence markers and proinflammatory cytokines in the submandibular glands (SMGs) of IgG4-RS patients (n = 18) and controls (n = 14) was determined by proteomics, real-time polymerase chain reaction (PCR), western blot, and immunohistochemistry. After interleukin-4 (IL-4) treatment, high-throughput RNA-sequencing was performed to identify the differentially expressed genes in SMG-C6 cells. A glandular fibrosis model was established by the intraglandular injection of IL-4 into mouse SMGs (n = 8 per group). RESULTS Salivary acinar and ductal epithelial cells underwent senescence in IgG4-RS patients, as indicated by the elevated activity of senescence-associated β-galactosidase, lipofuscin accumulation, enhanced expression of senescence markers (p53 and p16INK4A ), and upregulation of senescence-associated secretory phenotype factors. Moreover, there was a significant increase in IL-4 levels in SMGs of IgG4-RS patients, which positively correlated with p16INK4A expression and the fibrosis score. Incubation with IL-4 exacerbated salivary epithelial cell senescence by increasing the expression of p16INK4A through ROS-p38 mitogen-activated protein kinase (MAPK) pathway. The supernatant collected from IL-4-induced senescent SMG-C6 cells enhanced fibroblast activation and matrix protein production. Furthermore, injecting IL-4 promoted fibrosis and senescence phenotypes in the SMGs in vivo. CONCLUSION The cellular senescence induced by IL-4 through the ROS-p38 MAPK-p16INK4A pathway promoted fibrogenesis during IgG4-RS. Our data suggest that cellular senescence could serve as a novel therapeutic target for treating IgG4-RS.
Collapse
Affiliation(s)
- Sai-Nan Min
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, and Beijing Key Laboratory of Digital Stomatology, Beijing, P. R. China
| | - Meng-Qi Zhu
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, and Beijing Key Laboratory of Digital Stomatology, Beijing, P. R. China
| | - Xiang-Di Mao
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, P. R. China
| | - Wei Li
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, and Beijing Key Laboratory of Digital Stomatology, Beijing, P. R. China
| | - Tai Wei
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, and Beijing Key Laboratory of Digital Stomatology, Beijing, P. R. China
| | - Mei Mei
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, and Beijing Key Laboratory of Digital Stomatology, Beijing, P. R. China
| | - Yan Zhang
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, P. R. China
| | - Li-Ling Wu
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, P. R. China
| | - Guang-Yan Yu
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, and Beijing Key Laboratory of Digital Stomatology, Beijing, P. R. China
| | - Xin Cong
- Department of Physiology and Pathophysiology, Peking University School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, and Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, P. R. China
| |
Collapse
|
55
|
Jiang B, Kang X, Zhao G, Lu J, Wang Z. miR-138 Reduces the Dysfunction of T Follicular Helper Cells in Osteosarcoma via the PI3K/Akt/mTOR Pathway by Targeting PDK1. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:2895893. [PMID: 34950224 PMCID: PMC8691981 DOI: 10.1155/2021/2895893] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/05/2021] [Accepted: 11/12/2021] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To study the effect of miR-138 on the function of osteosarcoma (OS) T follicular helper cells (Tfh cells) and its mechanism. METHODS Peripheral blood mononuclear cells (PBMCs) were isolated from patients with osteosarcoma (OS group) and healthy volunteers (control group). CD4+CXCR5+ Tfh cells and CD9+ B cells were sorted by flow cytometry. qRT-PCR was used to detect the expression of miR-138 and PDK1 in the peripheral blood and CD4+CXCR5+ Tfh cells. Flow cytometry was employed to detect the proportion of CD4+CXCR5+ Tfh cells in CD4+ T cells, the level of CD40L in CD4+CXCR5+ Tfh cells, and the expression of CD27 and CD38 in B cells. Western blot was used to determine the protein expression of PDK1, PI3K, p-Akt, Akt, p-mTOR, and mTOR. In addition, dual-luciferase reporter assay was performed to verify the relationship between miR-138 and PDK1. ELISA method was used to determine the levels of IgM, IgG, IL-10, and IL-21. RESULTS Compared with that of the control group, the expression of miR-138 in PBMC and CD4+CXCR5+ Tfh cells of the OS group was lower; overexpression of miR-138 could promote the maturation of Tfh cells and immature B cells. The results of the dual-luciferase report experiment showed that miR-138 can target and negatively regulate PDK1, and PDK1 can reverse the effect of miR-138 on the function of Tfh cells and immature B cells. CONCLUSION miR-138 inhibits the PI3K/Akt/mTOR pathway by targeting and negatively regulating PDK1 to alleviate the dysfunction of T follicular helper cells in OS.
Collapse
Affiliation(s)
- Baoen Jiang
- Department of Traumatic Orthopaedics, Dongying People's Hospital, No. 317 Nanyi Road, Dongying District, Dongying, Shandong 257091, China
| | - Xiuqin Kang
- Department of Nursing, Dongying People's Hospital, No. 317 Nanyi Road, Dongying District, Dongying, Shandong 257091, China
| | - Gang Zhao
- Department of Traumatic Orthopaedics, Dongying People's Hospital, No. 317 Nanyi Road, Dongying District, Dongying, Shandong 257091, China
| | - Jianshu Lu
- Department of Traumatic Orthopaedics, Dongying People's Hospital, No. 317 Nanyi Road, Dongying District, Dongying, Shandong 257091, China
| | - Zhitao Wang
- Department of Traumatic Orthopaedics, Dongying People's Hospital, No. 317 Nanyi Road, Dongying District, Dongying, Shandong 257091, China
| |
Collapse
|
56
|
Bai Y, Sha J, Okui T, Moriyama I, Ngo HX, Tatsumi H, Kanno T. The Epithelial-Mesenchymal Transition Influences the Resistance of Oral Squamous Cell Carcinoma to Monoclonal Antibodies via Its Effect on Energy Homeostasis and the Tumor Microenvironment. Cancers (Basel) 2021; 13:5905. [PMID: 34885013 PMCID: PMC8657021 DOI: 10.3390/cancers13235905] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/18/2021] [Accepted: 11/20/2021] [Indexed: 12/12/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a major type of cancer that accounts for over 90% of all oral cancer cases. Recently developed evidence-based therapeutic regimens for OSCC based on monoclonal antibodies (mAbs), such as cetuximab, pembrolizumab, and nivolumab, have attracted considerable attention worldwide due to their high specificity, low toxicity, and low rates of intolerance. However, the efficacy of those three mAbs remains poor because of the low rate of responders and acquired resistance within a short period of time. The epithelial-mesenchymal transition (EMT) process is fundamental for OSCC growth and metastasis and is also responsible for the poor response to mAbs. During EMT, cancer cells consume abundant energy substrates and create an immunosuppressive tumor microenvironment to support their growth and evade T cells. In this review, we provide an overview of the complex roles of major substrates and signaling pathways involved in the development of therapeutic resistance in OSCC. In addition, we summarize potential therapeutic strategies that may help overcome this resistance. This review aims to help oral oncologists and researchers aiming to manage OSCC and establish new treatment modalities.
Collapse
Affiliation(s)
- Yunpeng Bai
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, Izumo, Shimane 693-8501, Japan; (Y.B.); (J.S.); (T.O.); (H.X.N.); (H.T.)
| | - Jingjing Sha
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, Izumo, Shimane 693-8501, Japan; (Y.B.); (J.S.); (T.O.); (H.X.N.); (H.T.)
| | - Tatsuo Okui
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, Izumo, Shimane 693-8501, Japan; (Y.B.); (J.S.); (T.O.); (H.X.N.); (H.T.)
| | - Ichiro Moriyama
- Department of Medical Oncology/Innovative Cancer Center, Shimane University Hospital, Izumo, Shimane 693-8501, Japan;
| | - Huy Xuan Ngo
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, Izumo, Shimane 693-8501, Japan; (Y.B.); (J.S.); (T.O.); (H.X.N.); (H.T.)
| | - Hiroto Tatsumi
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, Izumo, Shimane 693-8501, Japan; (Y.B.); (J.S.); (T.O.); (H.X.N.); (H.T.)
| | - Takahiro Kanno
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, Izumo, Shimane 693-8501, Japan; (Y.B.); (J.S.); (T.O.); (H.X.N.); (H.T.)
| |
Collapse
|
57
|
Genome-wide CRISPR-Cas9 screens identify mechanisms of BET bromodomain inhibitor sensitivity. iScience 2021; 24:103323. [PMID: 34805786 PMCID: PMC8581576 DOI: 10.1016/j.isci.2021.103323] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/23/2021] [Accepted: 10/19/2021] [Indexed: 01/06/2023] Open
Abstract
BET bromodomain inhibitors hold promise as therapeutic agents in diverse indications, but their clinical progression has been challenging and none have received regulatory approval. Early clinical trials in cancer have shown heterogeneous clinical responses, development of resistance, and adverse events. Increased understanding of their mechanism(s) of action and identification of biomarkers are needed to identify appropriate indication(s) and achieve efficacious dosing. Using genome-wide CRISPR-Cas9 screens at different concentrations, we report molecular mechanisms defining cellular responses to BET inhibitors, some of which appear specific to a single compound concentration. We identify multiple transcriptional regulators and mTOR pathway members as key determinants of JQ1 sensitivity and two Ca2+/Mn2+ transporters, ATP2C1 and TMEM165, as key determinants of JQ1 resistance. Our study reveals new molecular mediators of BET bromodomain inhibitor effects, suggests the involvement of manganese, and provides a rich resource for discovery of biomarkers and targets for combination therapies. CRISPR screens identify genes regulating sensitivity to BET bromodomain inhibitors Sensitivity and resistance hit lists are concentration-dependent mTOR pathway mediates sensitivity to BET bromodomain inhibitors Manganese regulates sensitivity to BET bromodomain inhibitors
Collapse
|
58
|
Effects of the Cytoplasm and Mitochondrial Specific Hydroxyl Radical Scavengers TA293 and mitoTA293 in Bleomycin-Induced Pulmonary Fibrosis Model Mice. Antioxidants (Basel) 2021; 10:antiox10091398. [PMID: 34573030 PMCID: PMC8469049 DOI: 10.3390/antiox10091398] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/02/2021] [Accepted: 08/25/2021] [Indexed: 12/14/2022] Open
Abstract
Lung fibrosis is the primary pathology in idiopathic pulmonary fibrosis and is considered to result from an increase in reactive oxygen species (ROS) levels in alveolar epithelial cells. However, the exact mechanism underlying lung fibrosis remains unclear and there is no effective therapy. The hydroxyl radical (•OH) has the strongest oxidizing potential among ROS. Recently, •OH localized to the cytoplasm (cyto •OH) was reported to induce cellular senescence, while mitochondria-localized •OH (mt •OH) was reported to induce apoptosis. We developed the cyto •OH- and mt •OH-scavenging antioxidants TA293 and mitoTA293 to evaluate the effects of cyto •OH and mt •OH in a bleomycin (BLM)-induced pulmonary fibrosis model. Treatment of BLM-induced pulmonary fibrosis mice with TA293 suppressed the induction of cellular senescence and fibrosis, as well as inflammation in the lung, but mitoTA293 exacerbated these. Furthermore, in BLM-stimulated primary alveolar epithelial cells, TA293 suppressed the activation of the p-ATMser1981/p-p53ser15/p21, p-HRI/p-eIF2ser51/ATF4/p16, NLRP3 inflammasome/caspase-1/IL-1β/IL1R/p-p38 MAPK/p16, and p21 pathways and the induction of cellular senescence. However, mitoTA293 suppressed the induction of mitophagy, enhanced the activation of the NLRP3 inflammasome/caspase-1/IL1β/IL1R/p-p38 MAPK/p16 and p21 pathways, and exacerbated cellular senescence, inflammation, and fibrosis. Our findings may help develop new strategies to treat idiopathic pulmonary fibrosis.
Collapse
|
59
|
Janelle V, Neault M, Lebel MÈ, De Sousa DM, Boulet S, Durrieu L, Carli C, Muzac C, Lemieux S, Labrecque N, Melichar HJ, Mallette FA, Delisle JS. p16 INK4a Regulates Cellular Senescence in PD-1-Expressing Human T Cells. Front Immunol 2021; 12:698565. [PMID: 34434190 PMCID: PMC8381277 DOI: 10.3389/fimmu.2021.698565] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 07/20/2021] [Indexed: 01/30/2023] Open
Abstract
T-cell dysfunction arising upon repeated antigen exposure prevents effective immunity and immunotherapy. Using various clinically and physiologically relevant systems, we show that a prominent feature of PD-1-expressing exhausted T cells is the development of cellular senescence features both in vivo and ex vivo. This is associated with p16INK4a expression and an impaired cell cycle G1 to S-phase transition in repeatedly stimulated T cells. We show that these T cells accumulate DNA damage and activate the p38MAPK signaling pathway, which preferentially leads to p16INK4a upregulation. However, in highly dysfunctional T cells, p38MAPK inhibition does not restore functionality despite attenuating senescence features. In contrast, p16INK4a targeting can improve T-cell functionality in exhausted CAR T cells. Collectively, this work provides insights into the development of T-cell dysfunction and identifies T-cell senescence as a potential target in immunotherapy.
Collapse
Affiliation(s)
- Valérie Janelle
- Research Centre, Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada
| | - Mathieu Neault
- Research Centre, Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada
| | - Marie-Ève Lebel
- Research Centre, Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada
| | - Dave Maurice De Sousa
- Research Centre, Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada.,Department of Microbiology, Infectious Diseases and Immunology, Université de Montréal, Montreal, QC, Canada
| | - Salix Boulet
- Research Centre, Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada
| | - Ludovic Durrieu
- Research Centre, Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada
| | - Cédric Carli
- Research Centre, Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada
| | - Chloé Muzac
- Research Centre, Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada
| | - Sébastien Lemieux
- Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, QC, Canada.,Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, QC, Canada
| | - Nathalie Labrecque
- Research Centre, Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada.,Department of Microbiology, Infectious Diseases and Immunology, Université de Montréal, Montreal, QC, Canada.,Department of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Heather J Melichar
- Research Centre, Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada.,Department of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Frédérick A Mallette
- Research Centre, Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada.,Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, QC, Canada.,Department of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Jean-Sébastien Delisle
- Research Centre, Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada.,Department of Medicine, Université de Montréal, Montreal, QC, Canada.,Division of Hematology-Oncology, Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada
| |
Collapse
|
60
|
Qian J, Shen Q, Yan CX, Yin H, Cao X, Lin ZH, Cai YF, Liu H. Atorvastatin improves bone marrow endothelial progenitor cell function from patients with immune-related hemocytopenia. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1142. [PMID: 34430583 PMCID: PMC8350688 DOI: 10.21037/atm-21-2459] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/13/2021] [Indexed: 11/06/2022]
Abstract
Background Immune-related hemocytopenia (IRH) is a type of autoimmune disease that targets bone marrow hematopoietic cells. This study investigated the influence of atorvastatin on the functional aspects of bone marrow endothelial progenitor cells (BM EPCs) in IRH patients. Methods BM EPCs were isolated from 15 patients with IRH and 20 normal controls. The isolated BM EPCs were characterized by flow cytometry. Cell counting kit-8, flow cytometry, and Transwell migration assays were used to determine the proliferation, apoptosis, and migration of BM EPCs, respectively. Protein levels were determined by western blot assay. Results The BM EPCs isolated from IRH patients showed reduced proliferation, increased apoptosis, and attenuated migratory ability compared to those from normal controls. Western blot analysis showed that the protein level of p-p38 was significantly increased, while that of Phosphorylated protein kinase B (p-AKT) was significantly decreased in the BM EPCs from IRH patients, compared to BM EPCs from healthy subjects. Cell proliferation and migration were significantly enhanced by atorvastatin, recombinant human thrombopoietin, and SB20358 compared to the untreated BM EPCs from IRH patients. Atorvastatin, Recombinant human thrombopoietin (TPO), and SB20358 treatment significantly suppressed the protein levels of p-p38 protein, but increased those of p-AKT in BM EPCS from IRH patients. Conclusions In summary, atorvastatin increases the number and function of BM EPCs in IRH patients by regulating the p38 and AKT signaling pathways.
Collapse
Affiliation(s)
- Juan Qian
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong, China
| | - Qian Shen
- Department of Oncology, Nantong Oncology Hospital, Nantong, China
| | | | - Hong Yin
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong, China
| | - Xin Cao
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong, China
| | - Zeng-Hua Lin
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong, China
| | - Yi-Feng Cai
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong, China
| | - Hong Liu
- Department of Hematology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
61
|
Liu J, Wu Z, Guo S, Zhang T, Ma X, Jiang K, Guo X, Deng G. IFN-τ Attenuates LPS-Induced Endometritis by Restraining HMGB1/NF-κB Activation in bEECs. Inflammation 2021; 44:1478-1489. [PMID: 33604776 DOI: 10.1007/s10753-021-01433-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 01/19/2021] [Accepted: 02/03/2021] [Indexed: 02/07/2023]
Abstract
Endometritis is a common inflammatory disease in uterine tissues that leads to animal infertility. Among the causes, Escherichia coli infection is one of the main reasons. Interferon-tau (IFN-τ) is the initial pregnancy signal for ruminant embryos and can induce immune tolerance in humans and other species. However, there are scarce reports on whether IFN-τ has a regulatory effect on endometrial inflammatory damage through HMGB1-NF-κB signalling. The purpose of this study was to investigate the regulatory mechanism of IFN-τ in HMGB1-NF-κB signalling in LPS-induced endometritis. ELISA and qPCR were used to detect the expression of LPS-induced pro-inflammatory cytokines in bovine endometrial epithelial cells (bEECs or BEND) under IFN-τ intervention, and the levels of HMGB1, p-IKK and p-p65 were detected by Western blotting. The nuclear translocation of NF-κB p65 was determined through immunofluorescence. In addition, bEECs were transfected with si-HMGB1 to elucidate the key role of HMGB1 and IFN-τ in the endometrial inflammatory cascade. The results indicated that IFN-τ inhibits the expression of related pro-inflammatory cytokines in an inflammatory injury model of bovine endometrial epithelial cells induced by LPS. Furthermore, experiments have proven that IFN-τ has protective effects on E. coli endotoxin-induced endometritis in mice in vivo. IFN-τ inhibited the HMGB1-NF-κB axis and significantly reduced the secretion of pro-inflammatory cytokines, the expression of HMGB1 protein and the levels of IKK and NF-κB p65 phosphorylation. In summary, our results showed that IFN-τ resists E. coli endotoxin-induced endometritis by attenuating HMGB1/NF-κB signalling.
Collapse
Affiliation(s)
- Junfeng Liu
- College of Animal Science, Tarim University, Alar, Xinjiang, 843300, People's Republic of China
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Zhimin Wu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Shuai Guo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Tao Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Xiaofei Ma
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - KangFeng Jiang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Xuefeng Guo
- College of Animal Science, Tarim University, Alar, Xinjiang, 843300, People's Republic of China.
| | - Ganzhen Deng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China.
| |
Collapse
|
62
|
Promising Anticancer Activities of Alismatis rhizome and Its Triterpenes via p38 and PI3K/Akt/mTOR Signaling Pathways. Nutrients 2021; 13:nu13072455. [PMID: 34371964 PMCID: PMC8308894 DOI: 10.3390/nu13072455] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 12/18/2022] Open
Abstract
The flowering plant genus Alisma, which belongs to the family Alismataceae, comprises 11 species, including Alisma orientale, Alisma canaliculatum, and Alisma plantago-aquatica. Alismatis rhizome (Ze xie in Chinese, Takusha in Japanese, and Taeksa in Korean, AR), the tubers of medicinal plants from Alisma species, have long been used to treat inflammatory diseases, hyperlipidemia, diabetes, bacterial infection, edema, oliguria, diarrhea, and dizziness. Recent evidence has demonstrated that its extract showed pharmacological activities to effectively reverse cancer-related molecular targets. In particular, triterpenes naturally isolated from AR have been found to exhibit antitumor activity. This study aimed to describe the biological activities and plausible signaling cascades of AR and its main compounds in experimental models representing cancer-related physiology and pathology. Available in vitro and in vivo studies revealed that AR extract possesses anticancer activity against various cancer cells, and the efficacy might be attributed to the cytotoxic and antimetastatic effects of its alisol compounds, such as alisol A, alisol B, and alisol B 23-acetate. Several beneficial functions of triterpenoids found in AR might be due to p38 activation and inhibition of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) signaling pathways. Moreover, AR and its triterpenes inhibit the proliferation of cancer cells that are resistant to chemotherapy. Thus, AR and its triterpenes may play potential roles in tumor attack, as well as a therapeutic remedy alone and in combination with other chemotherapeutic drugs.
Collapse
|
63
|
Implication of integrin α2β1 in senescence of SK-Mel-147 human melanoma cells. Aging (Albany NY) 2021; 13:18006-18017. [PMID: 34257160 PMCID: PMC8351665 DOI: 10.18632/aging.203309] [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: 04/08/2021] [Accepted: 06/19/2021] [Indexed: 12/12/2022]
Abstract
This investigation addressed the impact of integrin-initiated signaling pathways on senescence of tumor cells. In a model of human SK-Mel-147 melanoma cells, the silencing of integrin α2β1 strongly reduced cell proliferation and enhanced the percentage of SA-β-Gal-positive cells, a phenotypic feature of cellular senescence. These changes were accompanied by a significant increase in the activity of Akt and mTOR protein kinases and also in the expression of p53 and p21 oncosuppressors. Pharmacological inhibition of Akt and mTORC1 and genetic inhibition of p53 and p21 reduced the senescence of α2β1-depleted SK-Mel-147 cells to the level of control cells. Based on our earlier data on the non-canonical functions of Akt isomers in the invasion and anoikis of SK-Mel-147 cells, we investigated the role of Akt isomers in senescence induced by α2β1 suppression. The inhibition of Akt1 strongly reduced the percentage of SA-β-Gal-positive cells in the α2β1-depleted cell population, while the inhibition of Akt2 did not have a noticeable effect. Our data demonstrated for the first time that α2β1 is involved in the protection of tumor cells against senescence and that senescence, which is induced by the downregulation of α2β, is based on a signaling mechanism in which Akt1 performs a non-canonical function.
Collapse
|
64
|
Cell-Free Culture Supernatant of Probiotic Lactobacillus fermentum Protects Against H 2O 2-Induced Premature Senescence by Suppressing ROS-Akt-mTOR Axis in Murine Preadipocytes. Probiotics Antimicrob Proteins 2021; 12:563-576. [PMID: 31332650 DOI: 10.1007/s12602-019-09576-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Information regarding cellular anti-senescence attributes of probiotic bacteria vis-à-vis modulation of senescence-associated secretory phenotype (SASP) and mTOR signaling is very limited. The present study assessed anti-senescence potential of secretory metabolites of probiotic Lactobacillus fermentum (Lact. fermentum) using H2O2-induced model of senescence in 3T3-L1 preadipocytes. Application of H2O2-induced cellular senescence characterized by increased cell size and SA-β-gal activity, activation of SASP and reactive oxygen species (ROS), DNA damage response and induction of cell cycle inhibitors (p53/p21WAF1/p16INK4a). Further, a robust stimulation of the PI3K/Akt/mTOR pathway and AMPK signaling was also observed in H2O2-treated cells. However, exposure of cells to cell-free supernatant of Lact. fermentum significantly attenuated phosphorylation of PI3K/Akt/mTOR pathway and alleviated senescence markers p53, p21WAF1, SA-β-gal, p38MAPK, iNOS, cox-2, ROS, NF-κB, and DNA damage response. These results provide evidence that secretory metabolites of Lact. fermentum can mitigate the development as well as severity of stress-induced senescence thereby indicating its utility for use as anti-aging or age-delaying agent.
Collapse
|
65
|
Dual targeting of MEK and PI3K effectively controls the proliferation of human EGFR-TKI resistant non-small cell lung carcinoma cell lines with different genetic backgrounds. BMC Pulm Med 2021; 21:208. [PMID: 34210314 PMCID: PMC8252311 DOI: 10.1186/s12890-021-01571-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 06/21/2021] [Indexed: 12/14/2022] Open
Abstract
Background Molecular targeted therapy for non-small cell lung carcinoma (NSCLC) is restricted due to resistance to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs). This study evaluated the effects of dual targeting of MEK and PI3K in human EGFR-TKI resistant NSCLC cell lines. Methods EGFR-TKI resistant NSCLC cell lines H1975, H460, and A549, with different mutation and amplification status in EGFR, K-RAS, PIK3CA, and MET genes, were treated with a MEK162 (MEK inhibitor) and BKM120 (PI3K inhibitor) combination or a BIBW2992 (EGFR inhibitor) and ARQ197 (MET inhibitor) combination and assayed for cell proliferation, apoptosis, and cell cycle distribution. Results Dual targeting of MEK and PI3K efficiently inhibited the cell proliferation, induced apoptosis and the G0/G1 cell cycle, and decreased the phosphorylation of ERK1/2, AKT, S6, and 4E-BP1. H460 cells with K-RAS and PIK3CA mutation were most sensitive to MEK162 and BKM120 combinations. H1975 cells with EGFR and PIK3CA mutation and MET amplification were sensitive to BIBW2992 and ARQ197 combinations. Conclusion Dual targeting regulated the proliferation of EGFR-TKI-resistant NSCLC cells, especially mutants in K-RAS and PIK3CA that are promising for EGFR-TKI-resistant NSCLC therapeutics.
Collapse
|
66
|
Danesh Pazhooh R, Rahnamay Farnood P, Asemi Z, Mirsafaei L, Yousefi B, Mirzaei H. mTOR pathway and DNA damage response: A therapeutic strategy in cancer therapy. DNA Repair (Amst) 2021; 104:103142. [PMID: 34102579 DOI: 10.1016/j.dnarep.2021.103142] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/26/2021] [Accepted: 05/31/2021] [Indexed: 10/21/2022]
Abstract
The mammalian target of rapamycin (mTOR) is a conserved serine/threonine-protein kinase, comprising two subunit protein complexes: mTORC1 and mTORC2. In response to insult and cancer, the mTOR pathway plays a crucial role in regulating growth, metabolism, cell survival, and protein synthesis. Key subunits of mTORC1/2 catalyze the phosphorylation of various molecules, including eukaryotic translation initiation factor 4E binding protein 1 (4E-BP1), ribosomal protein S6 kinase β-1 (S6K1). The DNA damage response (DDR) maintains genomic stability and provides an opportunity for treating tumors with defects caused by DNA damaging agents. Many mTOR inhibitors are utilized for the treatment of cancers. However, several clinical trials are still assessing the efficacy of mTOR inhibitors. This paper discusses the role of the mTOR signaling pathway and its regulators in developing cancer. In the following, we will review the interaction between DDR and mTOR signaling and the innovative therapies applied in preclinical and clinical trials for treating cancers.
Collapse
Affiliation(s)
| | | | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| | - Liaosadat Mirsafaei
- Department of Cardiology, Ramsar Campus, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
67
|
Haines CN, Klingensmith HD, Komara M, Burd CJ. GREB1 regulates PI3K/Akt signaling to control hormone-sensitive breast cancer proliferation. Carcinogenesis 2021; 41:1660-1670. [PMID: 32894276 DOI: 10.1093/carcin/bgaa096] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 08/17/2020] [Accepted: 09/02/2020] [Indexed: 01/07/2023] Open
Abstract
Over 70% of breast cancers express the estrogen receptor (ER) and depend on ER activity for survival and proliferation. While hormone therapies that target receptor activity are initially effective, patients invariably develop resistance which is often associated with activation of the phosphoinositide 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway. While the mechanism by which estrogen regulates proliferation is not fully understood, one gene target of ER, growth regulation by estrogen in breast cancer 1 (GREB1), is required for hormone-dependent proliferation. However, the molecular function by which GREB1 regulates proliferation is unknown. Herein, we validate that knockdown of GREB1 results in growth arrest and that exogenous GREB1 expression initiates senescence, suggesting that an optimal level of GREB1 expression is necessary for proliferation of breast cancer cells. Under both of these conditions, GREB1 is able to regulate signaling through the PI3K/Akt/mTOR pathway. GREB1 acts intrinsically through PI3K to regulate phosphatidylinositol (3,4,5)-triphosphate levels and Akt activity. Critically, growth suppression of estrogen-dependent breast cancer cells by GREB1 knockdown is rescued by expression of constitutively activated Akt. Together, these data identify a novel molecular function by which GREB1 regulates breast cancer proliferation through Akt activation and provides a mechanistic link between estrogen signaling and the PI3K pathway.
Collapse
Affiliation(s)
- Corinne N Haines
- Department of Molecular Genetics, The Ohio State University, Columbus, OH, USA.,The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Hope D Klingensmith
- Department of Molecular Genetics, The Ohio State University, Columbus, OH, USA
| | - Makanko Komara
- The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Craig J Burd
- Department of Molecular Genetics, The Ohio State University, Columbus, OH, USA.,The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
68
|
Abstract
Extracellular nucleosides and nucleotides have widespread functions in responding to physiological stress. The "purinome" encompasses 4 G-protein-coupled receptors (GPCRs) for adenosine, 8 GPCRs activated by nucleotides, 7 adenosine 5'-triphosphate-gated P2X ion channels, as well as the associated enzymes and transporters that regulate native agonist levels. Purinergic signaling modulators, such as receptor agonists and antagonists, have potential for treating chronic pain. Adenosine and its analogues potently suppress nociception in preclinical models by activating A1 and/or A3 adenosine receptors (ARs), but safely harnessing this pathway to clinically treat pain has not been achieved. Both A2AAR agonists and antagonists are efficacious in pain models. Highly selective A3AR agonists offer a novel approach to treat chronic pain. We have explored the structure activity relationship of nucleoside derivatives at this subtype using a computational structure-based approach. Novel A3AR agonists for pain control containing a bicyclic ring system (bicyclo [3.1.0] hexane) in place of ribose were designed and screened using an in vivo phenotypic model, which reflected both pharmacokinetic and pharmacodynamic parameters. High specificity (>10,000-fold selective for A3AR) was achieved with the aid of receptor homology models based on related GPCR structures. These A3AR agonists are well tolerated in vivo and highly efficacious in models of chronic neuropathic pain. Furthermore, signaling molecules acting at P2X3, P2X4, P2X7, and P2Y12Rs play critical roles in maladaptive pain neuroplasticity, and their antagonists reduce chronic or inflammatory pain, and, therefore, purine receptor modulation is a promising approach for future pain therapeutics. Structurally novel antagonists for these nucleotide receptors were discovered recently.
Collapse
|
69
|
Rhaponticin suppresses osteosarcoma through the inhibition of PI3K-Akt-mTOR pathway. Saudi J Biol Sci 2021; 28:3641-3649. [PMID: 34220214 PMCID: PMC8241634 DOI: 10.1016/j.sjbs.2021.05.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/01/2021] [Accepted: 05/02/2021] [Indexed: 02/04/2023] Open
Abstract
Osteosarcoma is the frequent pediatric bone cancer where pediatric osteosarcoma incidences are more than 10% within the population. Most of the patients with osteosarcoma fall within the age of 15-30 years. Therefore, in this research, we examined the anticancer effect of Rhaponticin against the human osteosarcoma (MG-63) cells. The cytotoxicity of Rhaponticin on the MC3T3-E1 and MG-63 cells was examined through the MTT assay. The intracellular ROS accumulation, cell nuclear morphological alterations, apoptotic cell death and nuclear damages, and MMP status of Rhaponticin administered MG-63 cells were inspected by fluorescent staining techniques. The cell migration was assessed through scratch assay. The mRNA expressions of PI3K-Akt-mTOR signaling proteins were studied by RT-PCR analysis. Rhaponticin showed potent cytotoxicity, substantially inhibited the MG-63 cell growth, and displayed morphological alterations. However, rhaponticin did not affect the MC3T3-E1 cell viability. Rhaponticin administered MG-63 cells demonstrated augmented intracellular ROS accretion, weakened MMP, increased nuclear damages, and increased apoptosis. Rhaponticin effectively down-regulated the PI3K-Akt-mTOR signaling cascade in the MG-63 cells. These outcomes proved that the Rhaponticin can be a hopeful chemotherapeutic agent in the future to treat human osteosarcoma.
Collapse
|
70
|
Zhu T, Liu X, Song J, Li D, Pang XJ, Wang SH, Li QR, Fu DJ, Zhang SY, Xie HZ. Ras/Raf/MEK/ERK pathway axis mediated neurotoxicity induced by high-risk pesticide residue-Avermectin. ENVIRONMENTAL TOXICOLOGY 2021; 36:984-993. [PMID: 33381906 DOI: 10.1002/tox.23086] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 06/12/2023]
Abstract
Pesticide residues have become a healthy threaten of human beings. Among the pesticides, many of them have neurotoxicity. Extracellular Regulated Protein Kinases (ERK) pathway is an important signaling pathway that regulates a variety of downstream progress. In this work, peach (PRUNUS persica) and cherry (PRUNUS cerasus) were sampled from over 300 plantations in China and assessed for the residue risk. In mechanism studies, high-risk pesticide Avermectin showed a high activity inhibiting three neurotoxicity models, SH-SY5Y, PC-12 and SK-N-SH cells. At protein levels, ERK pathway proteins and their downstream proteins were obviously down-regulated. Moreover, the effects of low-dose Avermectin can be accumulated at protein levels in the low-dose long-term chronic toxicology detection.
Collapse
Affiliation(s)
- Ting Zhu
- Zhengzhou Fruit Research Institute, Chinese Academy of Agricultural Sciences, Zhengzhou, China
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, China
- Xiangyang Central Hospital, Xiangyang, China
| | - Xu Liu
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, China
| | - Jian Song
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, China
| | - Dong Li
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, China
| | - Xiao-Jing Pang
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, China
| | - Sheng-Hui Wang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Qing-Rong Li
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Dong-Jun Fu
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, China
| | - Sai-Yang Zhang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, China
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, China
| | - Han-Zhong Xie
- Zhengzhou Fruit Research Institute, Chinese Academy of Agricultural Sciences, Zhengzhou, China
- Key Laboratory of Fruit Breeding Technology,Ministry of Agriculture and Rural Affairs, Zhengzhou, China
| |
Collapse
|
71
|
Cytoguardin: A Tryptophan Metabolite against Cancer Growth and Metastasis. Int J Mol Sci 2021; 22:ijms22094490. [PMID: 33925793 PMCID: PMC8123408 DOI: 10.3390/ijms22094490] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 01/10/2023] Open
Abstract
Cytoguardin was identified in the conditioned medium of fibroblasts as a tryptophan metabolite, 5-methoxytryptophan (5-MTP). It is synthesized via two enzymatic steps: tryptophan hydroxylase (TPH) and hydroxyindole O-methyltransferase (HIOMT). A truncated HIOMT isoform, HIOMT298, catalyzes 5-MTP synthesis. Cancer cells produce scarce 5-MTP due to defective HIOMT298 expression. 5-MTP inhibits cancer cell COX-2 expression and thereby reduces COX-2-mediated cell proliferation and migration. 5-MTP also inhibits MMP-9 expression and thereby reduces cancer cell invasion. 5-MTP exerts its anti-cancer effect by blocking p38 MAPK and p38-mediated NF-κB and p300 HAT activation. The stable transfection of A549 cells with HIOMT298 restores 5-MTP production which renders cancer cells less aggressive. The implantation of HIOMT-transfected A549 into subcutaneous tissues of a murine xenograft tumor model shows that HIOMT-transduced A549 cells form smaller tumors and generate fewer metastatic lung nodules than control A549 cells. HIOMT298 transfection suppresses aromatic amino acid decarboxylase (AADC) expression and serotonin production. Serotonin is a cancer-promoting factor. By restoring 5-MTP and suppressing serotonin production, HIOMT298 overexpression converts cancer cells into less malignant phenotypes. The analysis of HIOMT expression in a human cancer tissue array showed reduced HIOMT levels in a majority of colorectal, pancreatic, and breast cancer. HIOMT298 may be a biomarker of human cancer progression. Furthermore, 5-MTP has the potential to be a lead compound in the development of new therapy for the chemoprevention of certain cancers such as hepatocellular cancer.
Collapse
|
72
|
Jiang Q, Yan M, Zhao Y, Zhou X, Yin L, Feng L, Liu Y, Jiang W, Wu P, Wang Y, Chen D, Yang S, Huang X, Jiang J. Dietary isoleucine improved flesh quality, muscle antioxidant capacity, and muscle growth associated with AKT/TOR/S6K1 and AKT/FOXO3a signaling in hybrid bagrid catfish (Pelteobagrus vachelli♀ × Leiocassis longirostris♂). J Anim Sci Biotechnol 2021; 12:53. [PMID: 33866964 PMCID: PMC8054373 DOI: 10.1186/s40104-021-00572-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 02/21/2021] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Muscle is the complex and heterogeneous tissue, which comprises the primary edible part of the trunk of fish and mammals. Previous studies have shown that dietary isoleucine (Ile) exerts beneficial effects on growth in aquatic animals. However, there were limited studies regarding the benefits of Ile on fish muscle and their effects on flesh quality and muscle growth. Thus, this study was conducted to explore whether dietary Ile had affected flesh quality and muscle growth in hybrid bagrid catfish (Pelteobagrus vachelli♀ × Leiocassis longirostris♂). METHODS A total of 630 hybrid fish, with an initial average body weight of 33.11 ± 0.09 g, were randomly allotted into seven experimental groups with three replicates each, and respectively fed seven diets with 5.0, 7.5, 10.0, 12.5, 15.0, 17.5, and 20.0 g Ile/kg diets for 8 weeks. RESULTS In the present study, we demonstrated that Ile significantly: (1) increased muscle protein and lipid contents and the frequency distribution of myofibers with ≤ 20 μm and ≥ 50 μm of diameter; (2) improved pH value, shear force, cathepsin B and L activities, hydroxyproline content, resilience, cohesiveness, and decreased cooking loss, lactate content, hardness, springiness, gumminess, and chewiness; (3) decreased reactive oxygen species (ROS), malondialdehyde (MDA), and protein carbonyl (PC) contents, GCLC and Keap1 mRNA levels, and up-regulated CuZnSOD, CAT, GPX1a, GST, and Nrf2 mRNA levels; (4) up-regulated the insulin-like growth factor 1, 2 (IGF-1, IGF-2), insulin-like growth factor 1 receptor (IGF-1R), proliferating cell nuclear antigen (PCNA), Myf5, Myod, Myog, Mrf4, and MyHC mRNA levels, and decreased MSTN mRNA level; (5) increased muscle protein deposition by activating AKT-TOR-S6K1 and AKT-FOXO3a signaling pathways. CONCLUSION These results revealed that dietary Ile improved flesh quality, which might be due to increasing nutritional content, physicochemical, texture parameters, and antioxidant ability; promoting muscle growth by affecting myocytes hyperplasia and hypertrophy, and muscle protein deposition associated with protein synthesis and degradation signaling pathways. Finally, the quadratic regression analysis of chewiness, ROS, and protein contents against dietary Ile levels suggested that the optimal dietary Ile levels for hybrid bagrid catfish was estimated to be 14.19, 12.36, and 12.78 g/kg diet, corresponding to 36.59, 31.87, and 32.96 g/kg dietary protein, respectively.
Collapse
Affiliation(s)
- Qin Jiang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Mingyao Yan
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Ye Zhao
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Xiaoqiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China.
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an, 625014, China.
| | - Long Yin
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Lin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an, 625014, China
| | - Yang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an, 625014, China
| | - Weidan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an, 625014, China
| | - Pei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an, 625014, China
| | - Yan Wang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Defang Chen
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Shiyong Yang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Xiaoli Huang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Jun Jiang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China.
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China.
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Ya'an, 625014, China.
| |
Collapse
|
73
|
Wang Y, Liu N, Li MY, Du MF. Long non-coding RNA ZEB2-AS1 regulates osteosarcoma progression by acting as a molecular sponge of miR-107 to modulate SALL4 expression. Am J Transl Res 2021; 13:1140-1154. [PMID: 33841645 PMCID: PMC8014379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 11/30/2020] [Indexed: 06/12/2023]
Abstract
Increasing evidence has confirmed long non-coding RNAs (lncRNAs) as important regulators involved in several pathophysiological processes in many diseases. The aim of this study was to investigate the roles of lncRNA ZEB2-AS1 (ZEB2-AS1) in osteosarcoma (OS). The levels of ZEB2-AS1 in OS tissues and cells were detected using RT-PCR. The clinical significance of ZEB2-AS1 expressions in OS patients was statistically analyzed. The functional effects of ZEB2-AS1 on the proliferation, apoptosis, invasion, and metastasis of OS cells was determined by a series of cellular experiments. Bioinformatic analysis, dual-luciferase reporter assays and pull-down assays were carried out for the confirmation of the molecular binding. We found that ZEB2-AS1 expression was distinctly upregulated in OS specimens and cell lines. Higher levels of ZEB2-AS1 in OS patients were associated with clinical stage, distant metastasis and unfavorable survivals. A multivariate Cox model revealed that ZEB2-AS1 expression was an independent prognostic factor for OS patients. Cellular experiments revealed that knockdown of ZEB2-AS1 inhibited proliferation and metastasis, and induced apoptosis in vitro. Mechanistic investigation revealed that ZEB2-AS1 acted as a sponge for miR-107 and blocked the inhibition of spalt like transcription factor 4 (SALL4) via miR-107 in OS cells. Rescue experiments suggested that up-regulation of ZEB2-AS1 could partly attenuate the miR-107 mediated inhibition of SALL4 expression in OS cells. To sum up, our data revealed that ZEB2-AS1 played an oncogenic role in OS progression, and could serve as a novel molecular target for treating this tumor.
Collapse
Affiliation(s)
- Yu Wang
- Department of Medical Laboratory, Zhumadian Central HospitalZhumadian 463000, Henan, China
| | - Ning Liu
- Department of Head and Neck Surgery, Linyi Cancer HospitalLinyi 276000, Shandong, China
| | - Ming-Yue Li
- Department of Head and Neck Surgery, Linyi Cancer HospitalLinyi 276000, Shandong, China
| | - Mao-Fang Du
- Department of Operating Room, Linyi Cancer HospitalLinyi 276000, Shandong, China
| |
Collapse
|
74
|
Hu D, Yuan S, Zhong J, Liu Z, Wang Y, Liu L, Li J, Wen F, Liu J, Zhang J. Cellular senescence and hematological malignancies: From pathogenesis to therapeutics. Pharmacol Ther 2021; 223:107817. [PMID: 33587950 DOI: 10.1016/j.pharmthera.2021.107817] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 02/07/2021] [Accepted: 02/09/2021] [Indexed: 12/24/2022]
Abstract
Cellular senescence constitutes a permanent state of cell cycle arrest in proliferative cells induced by different stresses. The exploration of tumor pathogenesis and therapies has been a research hotspot in recent years. Cellular senescence is a significant mechanism to prevent the proliferation of potential tumor cells, but it can also promote tumor growth. Increasing evidence suggests that cellular senescence is involved in the pathogenesis and development of hematological malignancies, including leukemia, myelodysplastic syndrome (MDS) and multiple myeloma (MM). Cellular senescence is associated with functional decline of hematopoietic stem cells (HSCs) and increased risk of hematological malignancies. Moreover, the bone marrow (BM) microenvironment has a crucial regulatory effect in the process of these diseases. The senescence-associated secretory phenotype (SASP) in the BM microenvironment establishes a protumor environment that supports the proliferation and survival of tumor cells. Therefore, a series of therapeutic strategies targeting cellular senescence have been gradually developed, including the induction of cellular senescence and elimination of senescent cells. This review systematically summarizes the emerging information describing the roles of cellular senescence in tumorigenesis and potential clinical applications, which may be beneficial for designing rational therapeutic strategies for various hematopoietic malignancies.
Collapse
Affiliation(s)
- Dingyu Hu
- Department of Clinical Laboratory, The First Affiliated Hospital, University of South China, Hengyang 421001, Hunan, China
| | - Shunling Yuan
- Department of Clinical Laboratory, The First Affiliated Hospital, University of South China, Hengyang 421001, Hunan, China
| | - Jing Zhong
- Institute of Clinical Medicine, First Affiliated Hospital of University of South China, Hengyang 421001, Hunan, China
| | - Zhaoping Liu
- Department of Clinical Laboratory, The First Affiliated Hospital, University of South China, Hengyang 421001, Hunan, China
| | - Yanyan Wang
- Department of Clinical Laboratory, The First Affiliated Hospital, University of South China, Hengyang 421001, Hunan, China
| | - Li Liu
- Department of Clinical Laboratory, The First Affiliated Hospital, University of South China, Hengyang 421001, Hunan, China
| | - Junjun Li
- Department of Hematology, The First Affiliated Hospital, University of South China, Hengyang 421001, Hunan, China
| | - Feng Wen
- Department of Hematology, The First Affiliated Hospital, University of South China, Hengyang 421001, Hunan, China
| | - Jing Liu
- Hunan Province Key Laboratory of Basic and Applied Hematology, Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, Hunan, China.
| | - Ji Zhang
- Department of Clinical Laboratory, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, Guangdong, China; Department of Clinical Laboratory, The First Affiliated Hospital, University of South China, Hengyang 421001, Hunan, China.
| |
Collapse
|
75
|
Cancer Stem Cells-Key Players in Tumor Relapse. Cancers (Basel) 2021; 13:cancers13030376. [PMID: 33498502 PMCID: PMC7864187 DOI: 10.3390/cancers13030376] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/10/2021] [Accepted: 01/18/2021] [Indexed: 02/06/2023] Open
Abstract
Tumor relapse and treatment failure are unfortunately common events for cancer patients, thus often rendering cancer an uncurable disease. Cancer stem cells (CSCs) are a subset of cancer cells endowed with tumor-initiating and self-renewal capacity, as well as with high adaptive abilities. Altogether, these features contribute to CSC survival after one or multiple therapeutic approaches, thus leading to treatment failure and tumor progression/relapse. Thus, elucidating the molecular mechanisms associated with stemness-driven resistance is crucial for the development of more effective drugs and durable responses. This review will highlight the mechanisms exploited by CSCs to overcome different therapeutic strategies, from chemo- and radiotherapies to targeted therapies and immunotherapies, shedding light on their plasticity as an insidious trait responsible for their adaptation/escape. Finally, novel CSC-specific approaches will be described, providing evidence of their preclinical and clinical applications.
Collapse
|
76
|
Control of Mesenchymal Stromal Cell Senescence by Tryptophan Metabolites. Int J Mol Sci 2021; 22:ijms22020697. [PMID: 33445766 PMCID: PMC7828284 DOI: 10.3390/ijms22020697] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/03/2021] [Accepted: 01/05/2021] [Indexed: 12/12/2022] Open
Abstract
Cellular senescence contributes to aging and age-related disorders. High glucose (HG) induces mesenchymal stromal/stem cell (MSC) senescence, which hampers cell expansion and impairs MSC function. Intracellular HG triggers metabolic shift from aerobic glycolysis to oxidative phosphorylation, resulting in reactive oxygen species (ROS) overproduction. It causes mitochondrial dysfunction and morphological changes. Tryptophan metabolites such as 5-methoxytryptophan (5-MTP) and melatonin attenuate HG-induced MSC senescence by protecting mitochondrial integrity and function and reducing ROS generation. They upregulate the expression of antioxidant enzymes. Both metabolites inhibit stress-induced MSC senescence by blocking p38 MAPK signaling pathway, NF-κB, and p300 histone acetyltransferase activity. Furthermore, melatonin upregulates SIRT-1, which reduces NF-κB activity by de-acetylation of NF-κB subunits. Melatonin and 5-MTP are a new class of metabolites protecting MSCs against replicative and stress-induced cellular senescence. They provide new strategies to improve the efficiency of MSC-based therapy for diverse human diseases.
Collapse
|
77
|
Harman JC, Otohinoyi DA, Reitnauer JW, Stowe AM, Gidday JM. Differential regulation of cerebral microvascular transcription by single and repetitive hypoxic conditioning. CONDITIONING MEDICINE 2021; 4:58-68. [PMID: 34414361 PMCID: PMC8372757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Systemic conditioning therapeutics afford brain protection at all levels of organization, occurring autonomously for neurons, glia, vascular smooth muscle, and endothelium, which are mediated systemically for the adaptive and innate immune system. The present study was undertaken to examine acute (3 h) and delayed (2 days) gene expression changes in mouse cerebral microvessels following single hypoxic conditioning (HX1) and repetitive hypoxic conditioning (HX9), the latter for which we showed previously to extend focal stroke tolerance from days to months. Microarray (Illumina) analyses were performed on microvessel-enriched fractions of adult mouse brain obtained from the following five groups (naïve; HX1-3h; HX1-2days; HX9-3h; HX9-2days). Differentially expressed genes were analyzed bioinformatically using Ingenuity Pathway Analysis software, with qPCR validating selected up- and down-regulated genes. As expected, some differentially expressed genes were common to more than one treatment or time point, whereas others were unique to treatment or time point. Bioinformatic analyses provided insights into acute (3h) inflammatory and immune signaling pathways that may be differentially activated by HX1 and HX9, with anti-inflammatory and trophic pathways coincident with the ischemia-tolerant phenotype two days after HX1. Interestingly, two days after HX9, microvessels were transcriptionally silent, with only five genes remaining differentially expressed relative to naïve mice. Our microarray findings and bioinformatic analyses suggest that cerebral microvessels from HX1-treated mice exhibit early activation of immune system signaling that is largely suppressed in microvessels from HX9-treated mice. These and other differences between these responses require further study, including at the proteomic level, and with pharmacologic and genetic experiments designed to reveal causality, to reveal further insights into the mechanisms underlying long-lasting stroke tolerance.
Collapse
Affiliation(s)
- Jarrod C Harman
- Departments of Ophthalmology, Louisiana State University School of Medicine, LSUHSC, New Orleans, LA
- Biochemistry & Molecular Biology, Louisiana State University School of Medicine, LSUHSC, New Orleans, LA
- Neuroscience Center of Excellence Louisiana State University School of Medicine, LSUHSC, New Orleans, LA
| | - David A Otohinoyi
- Biochemistry & Molecular Biology, Louisiana State University School of Medicine, LSUHSC, New Orleans, LA
| | - John W Reitnauer
- Department of Neurology, University of Kentucky College of Medicine, Lexington, KY
| | - Ann M Stowe
- Department of Neurology, University of Kentucky College of Medicine, Lexington, KY
| | - Jeff M Gidday
- Departments of Ophthalmology, Louisiana State University School of Medicine, LSUHSC, New Orleans, LA
- Biochemistry & Molecular Biology, Louisiana State University School of Medicine, LSUHSC, New Orleans, LA
- Neuroscience Center of Excellence Louisiana State University School of Medicine, LSUHSC, New Orleans, LA
- Physiology, Louisiana State University School of Medicine, LSUHSC, New Orleans, LA
| |
Collapse
|
78
|
Paredes-Céspedes DM, Rojas-García AE, Medina-Díaz IM, Ramos KS, Herrera-Moreno JF, Barrón-Vivanco BS, González-Arias CA, Bernal-Hernández YY. Environmental and socio-cultural impacts on global DNA methylation in the indigenous Huichol population of Nayarit, Mexico. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:4472-4487. [PMID: 32940839 DOI: 10.1007/s11356-020-10804-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/09/2020] [Indexed: 06/11/2023]
Abstract
Alterations of global DNA methylation have been evaluated in several studies worldwide; however, Long Interspersed Nuclear Elements-1 (LINE-1) methylation in genetically conserved populations such as indigenous communities have not, to our knowledge, been reported. The aim of this study was to evaluate the relationship between LINE-1 methylation patterns and factors such as pesticide exposure and socio-cultural characteristics in the Indigenous Huichol Population of Nayarit, Mexico. A cross-sectional study was conducted in 140 Huichol indigenous individuals. A structured questionnaire was used to determine general and anthropometric characteristics, diet, harmful habits, and pesticide exposure. DNA methylation was determined by pyrosequencing of bisulfite-treated DNA. A lower level of LINE-1 methylation was found in the indigenous population when compared to a Mestizo population previously studied by our group. This difference might be due to the influence of the genetic admixture and differing dietary and lifestyle habits. The males in the indigenous population exhibited increased LINE-1 methylation in comparison to the females. Sex and alcohol consumption showed positive associations with LINE-1 methylation, while weight, current work in the field, current pesticide usage, and folate intake exhibited negative associations with LINE-1 methylation. The results suggest that ethnicity, as well as other internal and environmental factors, might influence LINE-1 methylation.
Collapse
Affiliation(s)
- Diana Marcela Paredes-Céspedes
- Posgrado en Ciencias Biológico Agropecuarias, Unidad Académica de Agricultura, Km. 9 Carretera Tepic-Compostela, Xalisco, Nayarit, México
- Laboratorio de Contaminación y Toxicología Ambiental, Secretaría de Investigación y Posgrado, Universidad Autónoma de Nayarit, Ciudad de la Cultura s/n. C.P, 6300, Tepic, Nayarit, México
| | - Aurora Elizabeth Rojas-García
- Laboratorio de Contaminación y Toxicología Ambiental, Secretaría de Investigación y Posgrado, Universidad Autónoma de Nayarit, Ciudad de la Cultura s/n. C.P, 6300, Tepic, Nayarit, México
| | - Irma Martha Medina-Díaz
- Laboratorio de Contaminación y Toxicología Ambiental, Secretaría de Investigación y Posgrado, Universidad Autónoma de Nayarit, Ciudad de la Cultura s/n. C.P, 6300, Tepic, Nayarit, México
| | - Kenneth S Ramos
- Institute of Biosciences and Technology, Texas A&M University Health Science Center, 121 W. Holcombe Blvd, Houston, TX, 77030 m EE,UU, USA
| | - José Francisco Herrera-Moreno
- Posgrado en Ciencias Biológico Agropecuarias, Unidad Académica de Agricultura, Km. 9 Carretera Tepic-Compostela, Xalisco, Nayarit, México
- Laboratorio de Contaminación y Toxicología Ambiental, Secretaría de Investigación y Posgrado, Universidad Autónoma de Nayarit, Ciudad de la Cultura s/n. C.P, 6300, Tepic, Nayarit, México
| | - Briscia Socorro Barrón-Vivanco
- Laboratorio de Contaminación y Toxicología Ambiental, Secretaría de Investigación y Posgrado, Universidad Autónoma de Nayarit, Ciudad de la Cultura s/n. C.P, 6300, Tepic, Nayarit, México
| | - Cyndia Azucena González-Arias
- Laboratorio de Contaminación y Toxicología Ambiental, Secretaría de Investigación y Posgrado, Universidad Autónoma de Nayarit, Ciudad de la Cultura s/n. C.P, 6300, Tepic, Nayarit, México
| | - Yael Yvette Bernal-Hernández
- Laboratorio de Contaminación y Toxicología Ambiental, Secretaría de Investigación y Posgrado, Universidad Autónoma de Nayarit, Ciudad de la Cultura s/n. C.P, 6300, Tepic, Nayarit, México.
| |
Collapse
|
79
|
IER2-induced senescence drives melanoma invasion through osteopontin. Oncogene 2021; 40:6494-6512. [PMID: 34611309 PMCID: PMC8616759 DOI: 10.1038/s41388-021-02027-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 09/01/2021] [Accepted: 09/17/2021] [Indexed: 01/07/2023]
Abstract
Expression of the immediate-early response gene IER2 has been associated with the progression of several types of cancer, but its functional role is poorly understood. We found that increased IER2 expression in human melanoma is associated with shorter overall survival, and subsequently investigated the mechanisms through which IER2 exerts this effect. In experimental melanoma models, sustained expression of IER2 induced senescence in a subset of melanoma cells in a p53/MAPK/AKT-dependent manner. The senescent cells produced a characteristic secretome that included high levels of the extracellular phosphoglycoprotein osteopontin. Nuclear localization of the IER2 protein was critical for both the induction of senescence and osteopontin secretion. Osteopontin secreted by IER2-expressing senescent cells strongly stimulated the migration and invasion of non-senescent melanoma cells. Consistently, we observed coordinate expression of IER2, p53/p21, and osteopontin in primary human melanomas and metastases, highlighting the pathophysiological relevance of IER2-mediated senescence in melanoma progression. Together, our study reveals that sustained IER2 expression drives melanoma invasion and progression through stimulating osteopontin secretion via the stochastic induction of senescence.
Collapse
|
80
|
Cuollo L, Antonangeli F, Santoni A, Soriani A. The Senescence-Associated Secretory Phenotype (SASP) in the Challenging Future of Cancer Therapy and Age-Related Diseases. BIOLOGY 2020; 9:biology9120485. [PMID: 33371508 PMCID: PMC7767554 DOI: 10.3390/biology9120485] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/18/2020] [Accepted: 12/19/2020] [Indexed: 12/13/2022]
Abstract
Simple Summary A fundamental feature of cellular senescence is the emergence of the Senescence-Associated Secretory Phenotype (SASP), which represents a considerable source of inflammatory and tissue-remodeling cues. The pathophysiological relevance of senescence and SASP has generated a fertile area of research aimed at manipulating the SASP to fight cancer and age-related conditions. This review enlightens the most important mechanisms that regulate the SASP and summarizes the current evidence on the feasibility of intervening on its composition, providing a reading frame of the general potentialities of SASP modulation. Abstract Cellular senescence represents a robust tumor-protecting mechanism that halts the proliferation of stressed or premalignant cells. However, this state of stable proliferative arrest is accompanied by the Senescence-Associated Secretory Phenotype (SASP), which entails the copious secretion of proinflammatory signals in the tissue microenvironment and contributes to age-related conditions, including, paradoxically, cancer. Novel therapeutic strategies aim at eliminating senescent cells with the use of senolytics or abolishing the SASP without killing the senescent cell with the use of the so-called “senomorphics”. In addition, recent works demonstrate the possibility of modifying the composition of the secretome by genetic or pharmacological intervention. The purpose is not to renounce the potent immunostimulatory nature of SASP, but rather learning to modulate it for combating cancer and other age-related diseases. This review describes the main molecular mechanisms regulating the SASP and reports the evidence of the feasibility of abrogating or modulating the SASP, discussing the possible implications of both strategies.
Collapse
Affiliation(s)
- Lorenzo Cuollo
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00161 Rome, Italy; (L.C.); (A.S.)
- Center for Life Nano Science, Sapienza, Istituto Italiano di Tecnologia, 00161 Rome, Italy
| | - Fabrizio Antonangeli
- Institute of Molecular Biology and Pathology, National Research Council (CNR), 00185 Rome, Italy;
| | - Angela Santoni
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00161 Rome, Italy; (L.C.); (A.S.)
- IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) Neuromed, 86077 Pozzilli, Italy
| | - Alessandra Soriani
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00161 Rome, Italy; (L.C.); (A.S.)
- Correspondence:
| |
Collapse
|
81
|
An S, Cho SY, Kang J, Lee S, Kim HS, Min DJ, Son E, Cho KH. Inhibition of 3-phosphoinositide-dependent protein kinase 1 (PDK1) can revert cellular senescence in human dermal fibroblasts. Proc Natl Acad Sci U S A 2020; 117:31535-31546. [PMID: 33229519 PMCID: PMC7733858 DOI: 10.1073/pnas.1920338117] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Cellular senescence is defined as a stable, persistent arrest of cell proliferation. Here, we examine whether senescent cells can lose senescence hallmarks and reenter a reversible state of cell-cycle arrest (quiescence). We constructed a molecular regulatory network of cellular senescence based on previous experimental evidence. To infer the regulatory logic of the network, we performed phosphoprotein array experiments with normal human dermal fibroblasts and used the data to optimize the regulatory relationships between molecules with an evolutionary algorithm. From ensemble analysis of network models, we identified 3-phosphoinositide-dependent protein kinase 1 (PDK1) as a promising target for inhibitors to convert the senescent state to the quiescent state. We showed that inhibition of PDK1 in senescent human dermal fibroblasts eradicates senescence hallmarks and restores entry into the cell cycle by suppressing both nuclear factor κB and mTOR signaling, resulting in restored skin regeneration capacity. Our findings provide insight into a potential therapeutic strategy to treat age-related diseases associated with the accumulation of senescent cells.
Collapse
Affiliation(s)
- Sugyun An
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Si-Young Cho
- R&D Unit, Amorepacific Corporation, 17074 Gyeonggi-do, Republic of Korea
| | - Junsoo Kang
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Soobeom Lee
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Hyung-Su Kim
- R&D Unit, Amorepacific Corporation, 17074 Gyeonggi-do, Republic of Korea
| | - Dae-Jin Min
- R&D Unit, Amorepacific Corporation, 17074 Gyeonggi-do, Republic of Korea
| | - EuiDong Son
- R&D Unit, Amorepacific Corporation, 17074 Gyeonggi-do, Republic of Korea
| | - Kwang-Hyun Cho
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea;
| |
Collapse
|
82
|
Lee HS, Lee S, Cho KH. Cotargeting BET proteins overcomes resistance arising from PI3K/mTOR blockade-induced protumorigenic senescence in colorectal cancer. Int J Cancer 2020; 147:2824-2837. [PMID: 32599680 DOI: 10.1002/ijc.33047] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 03/19/2020] [Accepted: 04/14/2020] [Indexed: 12/13/2022]
Abstract
Therapeutics targeting the phosphatidylinositol 3-kinase/mammalian target of rapamycin (PI3K/mTOR) pathway initially produce potent antitumor effects, but resistance frequently occurs. Using a phosphoproteome analysis, we found that colorectal cancer (CRC) cells exhibit resistance against PI3K/mTOR inhibition through feedback activation of multiple receptor tyrosine kinases, and their downstream focal adhesion kinase, Src and extracellular signal-regulated kinases signaling. Unexpectedly, PI3K/mTOR blockade causes senescence, mediated by the activation of the stress kinase p38. The senescent cancer cells induce the secretion of various cytokines and this senescence-associated secretome increases migration and invasion capabilities of CRC cells. We found that cotargeting PI3K/mTOR and bromodomain and extra-terminal domain can suppress activation of many oncogenic kinases involved in resistance to the PI3K/mTOR inhibition, induce cell death in vitro and tumor regression in vivo, and further prolong the survival of xenograft models. Our findings provide a rationale for a novel therapeutic strategy to overcome resistance to the PI3K/mTOR inhibitors in CRC.
Collapse
Affiliation(s)
- Ho-Sung Lee
- Laboratory for Systems Biology and Bio-Inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea.,Graduate school of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Soobeom Lee
- Laboratory for Systems Biology and Bio-Inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Kwang-Hyun Cho
- Laboratory for Systems Biology and Bio-Inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea.,Graduate school of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| |
Collapse
|
83
|
Zhang X, Peng Y, Yuan Y, Gao Y, Hu F, Wang J, Zhu X, Feng X, Cheng Y, Wei Y, Fan X, Xie Y, Lv Y, Ashktorab H, Smoot D, Li S, Meltzer SJ, Hou G, Jin Z. Histone methyltransferase SET8 is regulated by miR-192/215 and induces oncogene-induced senescence via p53-dependent DNA damage in human gastric carcinoma cells. Cell Death Dis 2020; 11:937. [PMID: 33127874 PMCID: PMC7599338 DOI: 10.1038/s41419-020-03130-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 10/09/2020] [Accepted: 10/12/2020] [Indexed: 12/21/2022]
Abstract
Gastric cancer (GC) is the most common cancer throughout the world. Despite advances of the treatments, detailed oncogenic mechanisms are largely unknown. In our previous study, we investigated microRNA (miR) expression profiles in human GC using miR microarrays. We found miR-192/215 were upregulated in GC tissues. Then gene microarray was implemented to discover the targets of miR-192/215. We compared the expression profile of BGC823 cells transfected with miR-192/215 inhibitors, and HFE145 cells transfected with miR-192/-215 mimics, respectively. SET8 was identified as a proposed target based on the expression change of more than twofold. SET8 belongs to the SET domain-containing methyltransferase family and specifically catalyzes monomethylation of H4K20me. It is involved in diverse functions in tumorigenesis and metastasis. Therefore, we focused on the contributions of miR-192/215/SET8 axis to the development of GC. In this study, we observe that functionally, SET8 regulated by miR-192/215 is involved in GC-related biological activities. SET8 is also found to trigger oncogene-induced senescence (OIS) in GC in vivo and in vitro, which is dependent on the DDR (DNA damage response) and p53. Our findings reveal that SET8 functions as a negative regulator of metastasis via the OIS-signaling pathway. Taken together, we investigated the functional significance, molecular mechanisms, and clinical impact of miR-192/215/SET8/p53 in GC.
Collapse
Affiliation(s)
- Xiaojing Zhang
- grid.263488.30000 0001 0472 9649Guangdong Key Laboratory for Genome Stability & Disease Prevention and Regional Immunity and Diseases, Department of Pathology, Shenzhen University School of Medicine, Shenzhen, Guangdong 518060 People’s Republic of China
| | - Yin Peng
- grid.263488.30000 0001 0472 9649Guangdong Key Laboratory for Genome Stability & Disease Prevention and Regional Immunity and Diseases, Department of Pathology, Shenzhen University School of Medicine, Shenzhen, Guangdong 518060 People’s Republic of China
| | - Yuan Yuan
- grid.263488.30000 0001 0472 9649Guangdong Key Laboratory for Genome Stability & Disease Prevention and Regional Immunity and Diseases, Department of Pathology, Shenzhen University School of Medicine, Shenzhen, Guangdong 518060 People’s Republic of China
| | - Yuli Gao
- grid.263488.30000 0001 0472 9649Guangdong Key Laboratory for Genome Stability & Disease Prevention and Regional Immunity and Diseases, Department of Pathology, Shenzhen University School of Medicine, Shenzhen, Guangdong 518060 People’s Republic of China
| | - Fan Hu
- grid.263488.30000 0001 0472 9649Guangdong Key Laboratory for Genome Stability & Disease Prevention and Regional Immunity and Diseases, Department of Pathology, Shenzhen University School of Medicine, Shenzhen, Guangdong 518060 People’s Republic of China
| | - Jian Wang
- grid.263488.30000 0001 0472 9649Guangdong Key Laboratory for Genome Stability & Disease Prevention and Regional Immunity and Diseases, Department of Pathology, Shenzhen University School of Medicine, Shenzhen, Guangdong 518060 People’s Republic of China
| | - Xiaohui Zhu
- grid.263488.30000 0001 0472 9649Guangdong Key Laboratory for Genome Stability & Disease Prevention and Regional Immunity and Diseases, Department of Pathology, Shenzhen University School of Medicine, Shenzhen, Guangdong 518060 People’s Republic of China
| | - Xianling Feng
- grid.263488.30000 0001 0472 9649Guangdong Key Laboratory for Genome Stability & Disease Prevention and Regional Immunity and Diseases, Department of Pathology, Shenzhen University School of Medicine, Shenzhen, Guangdong 518060 People’s Republic of China
| | - Yulan Cheng
- grid.21107.350000 0001 2171 9311Department of Medicine/GI Division, Johns Hopkins University School of Medicine and Sidney Ki-mmel Comprehensive Cancer Center, Baltimore, MD 21287 USA
| | - Yanjie Wei
- grid.458489.c0000 0001 0483 7922Center for High Performance Computing, Shenzhen Institutes of Advanced Technology, Shenzhen, Guangdong 518000 People’s Republic of China
| | - Xinmin Fan
- grid.263488.30000 0001 0472 9649Guangdong Key Laboratory for Genome Stability & Disease Prevention and Regional Immunity and Diseases, Department of Pathology, Shenzhen University School of Medicine, Shenzhen, Guangdong 518060 People’s Republic of China
| | - Yaohong Xie
- grid.263488.30000 0001 0472 9649Guangdong Key Laboratory for Genome Stability & Disease Prevention and Regional Immunity and Diseases, Department of Pathology, Shenzhen University School of Medicine, Shenzhen, Guangdong 518060 People’s Republic of China
| | - Yansi Lv
- grid.263488.30000 0001 0472 9649Guangdong Key Laboratory for Genome Stability & Disease Prevention and Regional Immunity and Diseases, Department of Pathology, Shenzhen University School of Medicine, Shenzhen, Guangdong 518060 People’s Republic of China
| | - Hassan Ashktorab
- grid.257127.40000 0001 0547 4545Department of Medicine and Cancer Center, Howard University College of Medicine, Washington, DC 20060 USA
| | - Duane Smoot
- Department of Medicine, Meharry Medical Center, Nashville, TN 37208 USA
| | - Song Li
- grid.454883.6Shenzhen Science & Technology Development Exchange Center, Shenzhen Science and Technology Building, Shenzhen, Guangdong 518055 People’s Republic of China
| | - Stephen J. Meltzer
- grid.21107.350000 0001 2171 9311Department of Medicine/GI Division, Johns Hopkins University School of Medicine and Sidney Ki-mmel Comprehensive Cancer Center, Baltimore, MD 21287 USA
| | - Gangqiang Hou
- Department of Medical Image Center, Kangning Hospital, Shenzhen, Guangdong Province, 518000, People's Republic of China.
| | - Zhe Jin
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and Regional Immunity and Diseases, Department of Pathology, Shenzhen University School of Medicine, Shenzhen, Guangdong, 518060, People's Republic of China.
| |
Collapse
|
84
|
Lim JS, Lee DY, Kim HS, Park SC, Park JT, Kim HS, Oh WK, Cho KA. Identification of a novel senomorphic agent, avenanthramide C, via the suppression of the senescence-associated secretory phenotype. Mech Ageing Dev 2020; 192:111355. [PMID: 32941937 DOI: 10.1016/j.mad.2020.111355] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/14/2020] [Accepted: 09/07/2020] [Indexed: 01/07/2023]
Abstract
Senescent cells are deeply involved in the induction of tissue damage and aging-related diseases. The identification of factors that eliminate senescent cells or inhibit the senescence-associated secretory phenotype (SASP) in these cells is necessary. Here, we report an avenanthramice C (Avn C) extracted from oat as a new SASP modulator. Treatment with Avn C led to a significant reduction in the levels of markers of senescent cells, with no toxicity observed. The SASP was also inhibited by Avn C treatment, similar to non-senescent cells, and the suppression of cell division by autocrine signals associated with SASP was restored. To investigate the mechanism underlying SASP inhibition by Avn C, we analyzed the effect of Avn C in lipopolysaccharide (LPS)-induced inflammation in non-senescent cells. Avn C inhibited nuclear factor κB (NF-κB) activity and the secretion of inflammatory cytokines before or after LPS treatment. Although the activity of MAP kinases, which are NF-κB upstream signals, was inhibited by Avn C in LPS-induced inflammation, only p38 activity was specifically inhibited in senescent cells. Interestingly, the inhibition of p38 in senescent cells was observed through Avn C-induced 5'-adenosine monophosphate-activated protein kinase (AMPK) activity. Avn C-induced inhibition of the SASP is triggered by senescence-related stress.
Collapse
Affiliation(s)
- Jae Sung Lim
- Department of Biochemistry, Chonanm National University Medical School, 264 Seoyang-ro, Hwasun-gun, Jeonnam-do, 58128, Republic of Korea; Combinatorial Tumor Immunotherapy Medical Research Center, Chonnam National University Medical School, 264 Seoyang-ro, Hwasun-gun, Jeonnam-do, 58128, Republic of Korea
| | - Da Young Lee
- Department of Biochemistry, Chonanm National University Medical School, 264 Seoyang-ro, Hwasun-gun, Jeonnam-do, 58128, Republic of Korea; Center for Creative Biomedical Scientists, Chonnam National University Medical School, 264 Seoyang-ro, Hwasun-gun, Jeonnam-do, 58128, Republic of Korea
| | - Hyung Seok Kim
- Center for Creative Biomedical Scientists, Chonnam National University Medical School, 264 Seoyang-ro, Hwasun-gun, Jeonnam-do, 58128, Republic of Korea; Department of Forensic Science, Chonnam National University Medical School, 264 Seoyang-ro, Hwasun-gun, Jeonnam-do, 58128, Republic of Korea
| | - Sang Chul Park
- Future Life and Society Research Center, Chonnam National University Medical School, 264 Seoyang-ro, Hwasun-gun, Jeonnam-do, 58128, Republic of Korea
| | - Joon Tae Park
- Department of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Hyeon Sik Kim
- Medical Photonic Research Center, Korea Photonics Technology Institute, Gwangju, 61007, Republic of Korea
| | - Won Keun Oh
- Korea Bioactive Natural Material Bank, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Kyung A Cho
- Department of Biochemistry, Chonanm National University Medical School, 264 Seoyang-ro, Hwasun-gun, Jeonnam-do, 58128, Republic of Korea; Center for Creative Biomedical Scientists, Chonnam National University Medical School, 264 Seoyang-ro, Hwasun-gun, Jeonnam-do, 58128, Republic of Korea; Future Life and Society Research Center, Chonnam National University Medical School, 264 Seoyang-ro, Hwasun-gun, Jeonnam-do, 58128, Republic of Korea.
| |
Collapse
|
85
|
Jiang SB, Lu YS, Liu T, Li LM, Wang HX, Wu Y, Gao XH, Chen HD. UVA influenced the SIRT1-miR-27a-5p-SMAD2-MMP1/COL1/BCL2 axis in human skin primary fibroblasts. J Cell Mol Med 2020; 24:10027-10041. [PMID: 32790210 PMCID: PMC7520305 DOI: 10.1111/jcmm.15610] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 06/10/2020] [Accepted: 06/17/2020] [Indexed: 12/21/2022] Open
Abstract
Both SIRT1 and UVA radiation are involved in cellular damage processes such as apoptosis, senescence and ageing. MicroRNAs (miRNAs) have been reported to be closely related to UV radiation, as well as to SIRT1. In this study, we investigated the connections among SIRT1, UVA and miRNA in human skin primary fibroblasts. Our results showed that UVA altered the protein level of SIRT1 in a time point–dependent manner. Using miRNA microarray, bioinformatics analysis, we found that knocking down SIRT1 could cause up‐regulation of miR‐27a‐5p and the latter could down‐regulate SMAD2, and these results were verified by qRT‐PCR or Western blot. Furthermore, UVA radiation (5 J/cm2), knocking down SIRT1 or overexpression of miR‐27a‐5p led to increased expression of MMP1, and decreased expressions of COL1 and BCL2. We also found additive impacts on MMP1, COL1 and BCL2 under the combination of UVA radiation + Sirtinol (SIRT1 inhibitor), or UVA radiation + miR‐27a‐5p mimic. SIRT1 activator resveratrol could reverse damage changes caused by UVA radiation. Besides, absent of SIRT1 or overexpression of miR‐27a‐5p increased cell apoptosis and induced cell arrest in G2/M phase. Taken together, these results demonstrated that UVA could influence a novel SIRT1‐miR‐27a‐5p‐SMAD2‐MMP1/COL1/BCL2 axis in skin primary fibroblasts, and may provide potential therapeutic targets for UVA‐induced skin damage.
Collapse
Affiliation(s)
- Shi-Bin Jiang
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China
| | - Yan-Song Lu
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China
| | - Tao Liu
- Department of Urinary Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Liang-Man Li
- Department of Orthopedics, The First Hospital of China Medical University, Shenyang, China
| | - He-Xiao Wang
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China
| | - Yan Wu
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China
| | - Xing-Hua Gao
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China
| | - Hong-Duo Chen
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
86
|
Wissler Gerdes EO, Zhu Y, Weigand BM, Tripathi U, Burns TC, Tchkonia T, Kirkland JL. Cellular senescence in aging and age-related diseases: Implications for neurodegenerative diseases. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 155:203-234. [PMID: 32854855 PMCID: PMC7656525 DOI: 10.1016/bs.irn.2020.03.019] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Aging is the major predictor for developing multiple neurodegenerative diseases, including Alzheimer's disease (AD) other dementias, and Parkinson's disease (PD). Senescent cells, which can drive aging phenotypes, accumulate at etiological sites of many age-related chronic diseases. These cells are resistant to apoptosis and can cause local and systemic dysfunction. Decreasing senescent cell abundance using senolytic drugs, agents that selectively target these cells, alleviates neurodegenerative diseases in preclinical models. In this review, we consider roles of senescent cells in neurodegenerative diseases and potential implications of senolytic agents as an innovative treatment.
Collapse
Affiliation(s)
| | - Yi Zhu
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States
| | - B Melanie Weigand
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States
| | - Utkarsh Tripathi
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States
| | - Terence C Burns
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States
| | - Tamar Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States.
| |
Collapse
|
87
|
IGF‑IR promotes clonal cell proliferation in myelodysplastic syndromes via inhibition of the MAPK pathway. Oncol Rep 2020; 44:1094-1104. [PMID: 32583001 PMCID: PMC7388562 DOI: 10.3892/or.2020.7652] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 05/19/2020] [Indexed: 12/12/2022] Open
Abstract
Type 1 insulin-like growth factor receptor (IGF-IR) signaling is considered to serve a key role in the development of cancer. However, the effects of IGF-IR on the malignant characteristics of myelodysplastic syndrome (MDS) clonal cells remains to be determined. In the present study it was demonstrated that knockdown of IGF-IR reduced the proliferation and increased the apoptosis of MDS/leukemia cells. Integrated analysis of gene expression profiles using bioinformatics identified the MAPK signaling pathway as a critical downstream factor of IGF-IR, and this was confirmed in vitro using western blotting which revealed that IGF-IR knockdown significantly increased the expression of activated MAPK. Furthermore, IGF-IR signaling was inhibited to investigate the potential of IGF-IR as a therapeutic target of MDS. The results revealed that the IGF-IR inhibitor picropodophyllin (PPP) inhibited cell proliferation, promoted cell apoptosis and arrested the cell cycle at the G2/M phase in MDS/leukemia cells. Similar to the effects of IGF-IR knockdown, PPP treatment also increased MAPK signaling in vitro. In conclusion, IGF-IR may serve as a potential therapeutic target of MDS.
Collapse
|
88
|
WIP1 promotes cancer stem cell properties by inhibiting p38 MAPK in NSCLC. Signal Transduct Target Ther 2020; 5:36. [PMID: 32296033 PMCID: PMC7156655 DOI: 10.1038/s41392-020-0126-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 02/02/2020] [Accepted: 02/04/2020] [Indexed: 11/09/2022] Open
Abstract
Cancer stem cells (CSCs) are a small population of stem cell-like cancer cells that can initiate tumors in vivo, and are the major source of cancer initiation, relapse, and drug resistance. We previously reported that the p38 MAPK, through its downstream effectors MK2 and HSP27, suppressed CSC properties by downregulating the expression of transcription factors that mediate stemness in non-small-cell lung cancer (NSCLC) cells, and that despite unaltered total expression of total p38 proteins, the levels of activated p38 were reduced in NSCLC tissues. However, the mechanism underlying the reduced levels of activated p38 in NSCLC is unknown. In this study, we identified WIP1, a p38 phosphatase frequently overexpressed in cancer, as a suppressor of p38 in a pathway that regulates CSC properties in NSCLC. Increased WIP1 expression correlated with reduced levels of activated p38, and with increased levels of a CSC marker in NSCLC tissues. Further investigation revealed that WIP1 promoted stemness-related protein expression and CSC properties by inhibiting p38 activity in NSCLC cells. WIP1 inhibitors are currently under development as anticancer drugs based on their ability to reactivate p53. We found that a WIP1 inhibitor suppressed stemness-related protein expression and CSC properties by activating p38 in NSCLC cells in vitro and in vivo. These studies have identified the WIP1–p38–MK2–HSP27 cascade as a novel signaling pathway that, when altered, promotes CSC properties in NSCLC development, and have defined novel mechanisms underlying the oncogenic activity of WIP1 and the anticancer efficacy of WIP1 inhibitors.
Collapse
|
89
|
Anerillas C, Abdelmohsen K, Gorospe M. Regulation of senescence traits by MAPKs. GeroScience 2020; 42:397-408. [PMID: 32300964 PMCID: PMC7205942 DOI: 10.1007/s11357-020-00183-3] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 03/10/2020] [Indexed: 01/10/2023] Open
Abstract
A phenotype of indefinite growth arrest acquired in response to sublethal damage, cellular senescence affects normal aging and age-related disease. Mitogen-activated protein kinases (MAPKs) are capable of sensing changes in cellular conditions, and in turn elicit adaptive responses including cell senescence. MAPKs modulate the levels and function of many proteins, including proinflammatory factors and factors in the p21/p53 and p16/RB pathways, the main senescence-regulatory axes. Through these actions, MAPKs implement key traits of senescence-growth arrest, cell survival, and the senescence-associated secretory phenotype (SASP). In this review, we summarize and discuss our current knowledge of the impact of MAPKs in senescence. In addition, given that eliminating or suppressing senescent cells can improve health span, we discuss the function and possible exploitation of MAPKs in the elimination (senolysis) or suppression (senostasis) of senescent cells.
Collapse
Affiliation(s)
- Carlos Anerillas
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Kotb Abdelmohsen
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, 21224, USA.
| |
Collapse
|
90
|
Tian W, Wang ZW, Yuan BM, Bao YG. Calycosin induces apoptosis in osteosarcoma cell line via ERβ‑mediated PI3K/Akt signaling pathways. Mol Med Rep 2020; 21:2349-2356. [PMID: 32236598 PMCID: PMC7185272 DOI: 10.3892/mmr.2020.11039] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 02/24/2020] [Indexed: 02/06/2023] Open
Abstract
Previous studies have shown that calycosin, a natural phytoestrogen which is structurally similar to estrogen, inhibits proliferation and induces apoptosis in estrogen-dependent cancer types via the estrogen receptor (ER)β-induced inhibition of PI3K/Akt. Therefore, the aims of the present study were to investigate the effects of calycosin on human osteosarcoma (OS), and to examine the molecular mechanisms associated with ERβ. Human OS MG-63 cells were treated with various concentrations of calycosin, and MTT and flow cytometry assays were used to assess the effects of calycosin on cellular proliferation and apoptosis. In addition, protein expression levels of ERβ, phosphorylated (p)-PI3K, p-Akt, cleaved poly (ADP-ribose) polymerase 1 (PARP) and cleaved caspase-3 were evaluated by western blot analysis. The present results suggested that calycosin inhibited proliferation and induced apoptosis in MG-63 cells. Furthermore, increased ERβ expression was detected in OS MG-63 cells treated with calycosin, and an ERβ inhibitor (PHTPP) reversed calycosin-induced cytotoxicity and apoptosis. Moreover, phosphorylation levels of PI3K and Akt were significantly downregulated after calycosin treatment, whereas PHTPP reversed their phosphorylation. ERβ-mediated PI3K/Akt downstream signaling pathways were found to influence the activity of poly (ADP-ribose) polymerase 1 and caspase-3. Thus, the present results indicated that calycosin inhibited proliferation and induced apoptosis in OS MG-63 cells, and that these effects were mediated by ERβ-dependent inhibition of the PI3K/Akt pathways.
Collapse
Affiliation(s)
- Wei Tian
- Department of Orthopedics, Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia Autonomous Region 028007, P.R. China
| | - Zhi-Wei Wang
- Department of Orthopedics, Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia Autonomous Region 028007, P.R. China
| | - Bao-Ming Yuan
- Department of Orthopedics, Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia Autonomous Region 028007, P.R. China
| | - Yong-Ge Bao
- Department of Orthopedics, Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia Autonomous Region 028007, P.R. China
| |
Collapse
|
91
|
Zhu H, Blake S, Kusuma FK, Pearson RB, Kang J, Chan KT. Oncogene-induced senescence: From biology to therapy. Mech Ageing Dev 2020; 187:111229. [PMID: 32171687 DOI: 10.1016/j.mad.2020.111229] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 03/08/2020] [Accepted: 03/09/2020] [Indexed: 12/15/2022]
Abstract
Oncogene-induced senescence (OIS) is a powerful intrinsic tumor-suppressive mechanism, arresting cell cycle progression upon oncogene-activating genomic alterations. The discovery and characterization of the senescence-associated secretome unveiled a rich additional complexity to the senescence phenotype, including extrinsic impacts on the microenvironment and engagement of the immune response. Emerging evidence suggests that senescence phenotypes vary depending on the oncogenic stimulus. Therefore, understanding the mechanisms underlying OIS and how they are subverted in cancer will provide invaluable opportunities to identify alternative strategies for treating oncogene-driven cancers. In this review, we primarily discuss the key mechanisms governing OIS driven by the RAS/MAPK and PI3K/AKT pathways and how understanding the biology of senescent cells has uncovered new therapeutic possibilities to target cancer.
Collapse
Affiliation(s)
- Haoran Zhu
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia
| | - Shaun Blake
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia
| | - Frances K Kusuma
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia
| | - Richard B Pearson
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, 3052, Australia; Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, 3052, Australia; Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, 3168, Australia.
| | - Jian Kang
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Keefe T Chan
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, 3052, Australia.
| |
Collapse
|
92
|
Mijit M, Caracciolo V, Melillo A, Amicarelli F, Giordano A. Role of p53 in the Regulation of Cellular Senescence. Biomolecules 2020; 10:biom10030420. [PMID: 32182711 PMCID: PMC7175209 DOI: 10.3390/biom10030420] [Citation(s) in RCA: 350] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/04/2020] [Accepted: 03/05/2020] [Indexed: 12/12/2022] Open
Abstract
The p53 transcription factor plays a critical role in cellular responses to stress. Its activation in response to DNA damage leads to cell growth arrest, allowing for DNA repair, or directs cellular senescence or apoptosis, thereby maintaining genome integrity. Senescence is a permanent cell-cycle arrest that has a crucial role in aging, and it also represents a robust physiological antitumor response, which counteracts oncogenic insults. In addition, senescent cells can also negatively impact the surrounding tissue microenvironment and the neighboring cells by secreting pro-inflammatory cytokines, ultimately triggering tissue dysfunction and/or unfavorable outcomes. This review focuses on the characteristics of senescence and on the recent advances in the contribution of p53 to cellular senescence. Moreover, we also discuss the p53-mediated regulation of several pathophysiological microenvironments that could be associated with senescence and its development.
Collapse
Affiliation(s)
- Mahmut Mijit
- Sbarro Institute for Cancer Research and Molecular Medicine, Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
- Department of Medical Biotechnologies, University of Siena, 67100 Siena, Italy
| | - Valentina Caracciolo
- Sbarro Institute for Cancer Research and Molecular Medicine, Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Antonio Melillo
- Sbarro Institute for Cancer Research and Molecular Medicine, Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Fernanda Amicarelli
- Department of Medical Biotechnologies, University of Siena, 67100 Siena, Italy
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 53100 L’Aquila, Italy
- Correspondence:
| |
Collapse
|
93
|
Yu S, Cheng Y, Li B, Xue J, Yin Y, Gao J, Gong Z, Wang J, Mu Y. M1 macrophages accelerate renal glomerular endothelial cell senescence through reactive oxygen species accumulation in streptozotocin-induced diabetic mice. Int Immunopharmacol 2020; 81:106294. [PMID: 32062081 DOI: 10.1016/j.intimp.2020.106294] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/30/2020] [Accepted: 02/05/2020] [Indexed: 01/07/2023]
Abstract
Cellular senescence is a fundamental aging mechanism leading to tissue dysfunction. Accumulation of senescent cells is observed in the context of diabetes, which plays an important role in the pathogenesis of diabetes and its complications. Macrophages, the most prevalent leucocytes found in diabetic kidney, have been implicated in the modulation of cellular senescence; however, their role and mechanism in cellular senescence of diabetic kidney have not been determined. In this study, we found trends of cellular senescence in the glomeruli of streptozotocin-induced diabetic mice. The onset of glomerular senescence was confirmed by increased SA-β-gal staining, the upregulation of p16INK4a, p21, and p53 protein levels and the increased expression of SASP RNA. The senescent cells in the glomeruli were mainly endothelial cells. We next confirmed that M1 macrophages accumulated in the glomeruli, occurred just shortly before glomerular senescence. Therefore, we examined whether M1 macrophage accumulation is associated with glomerular endothelial cell senescence. Thus, an in vitro co-culture model was established using human renal glomerular endothelial cells (HRGECs) and M1-polarized THP-1 macrophages. Indeed, M1 macrophages induced senescence in HRGECs. Furthermore, intracellular ROS levels and p38 MAPK signalling activation were significantly increased in HRGECs and reducing ROS generation significantly abolished M1 macrophage-mediated endothelial senescence and p38 MAPK activation, suggesting that M1 macrophage-mediated endothelial senescence is largely dependent on ROS. Thus, our results demonstrate that kidney M1 macrophage accumulation is in connection with endothelial cell senescence and strategy to modulate M1 macrophages accumulation is promising to be a new target for immunotherapy for diabetic kidney disease and other age-related diseases.
Collapse
Affiliation(s)
- Songyan Yu
- School of Medicine, Nankai University, Tianjin, China; Department of Endocrinology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Yu Cheng
- Department of Endocrinology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Bing Li
- Department of Endocrinology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Jing Xue
- Department of Endocrinology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Yaqi Yin
- Department of Endocrinology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Jieqing Gao
- Department of Endocrinology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Zhengyuan Gong
- Department of Endocrinology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Jie Wang
- School of Medicine, Nankai University, Tianjin, China; Department of Endocrinology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Yiming Mu
- School of Medicine, Nankai University, Tianjin, China; Department of Endocrinology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China.
| |
Collapse
|
94
|
Hamsanathan S, Alder JK, Sellares J, Rojas M, Gurkar AU, Mora AL. Cellular Senescence: The Trojan Horse in Chronic Lung Diseases. Am J Respir Cell Mol Biol 2020; 61:21-30. [PMID: 30965013 DOI: 10.1165/rcmb.2018-0410tr] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Senescence is a cell fate decision characterized by irreversible arrest of proliferation accompanied by a senescence-associated secretory phenotype. Traditionally, cellular senescence has been recognized as a beneficial physiological mechanism during development and wound healing and in tumor suppression. However, in recent years, evidence of negative consequences of cellular senescence has emerged, illuminating its role in several chronic pathologies. In this context, senescent cells persist or accumulate and have detrimental consequences. In this review, we discuss the possibility that in chronic obstructive pulmonary disease, persistent senescence impairs wound healing in the lung caused by secretion of proinflammatory senescence-associated secretory phenotype factors and exhaustion of progenitor cells. In contrast, in idiopathic pulmonary fibrosis, chronic senescence in alveolar epithelial cells exacerbates the accumulation of senescent fibroblasts together with production of extracellular matrix. We review how cellular senescence may contribute to lung disease pathology.
Collapse
Affiliation(s)
| | - Jonathan K Alder
- 2 Division of Pulmonary Allergy and Critical Care Medicine, and.,3 Dorothy P. and Richard P. Simmons Center for Interstitial Lung Diseases
| | - Jacobo Sellares
- 4 Interstitial Lung Disease Program, Servei de Pneumologia, Hospital Clínic Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain.,5 Centro de Investigaciones Biomedicas en Red-Enfermedades Respiratorias (CibeRes CB06/06/0028), Instituto de Salud Carlos III, Barcelona, Spain; and
| | - Mauricio Rojas
- 2 Division of Pulmonary Allergy and Critical Care Medicine, and.,3 Dorothy P. and Richard P. Simmons Center for Interstitial Lung Diseases.,6 McGowan Institute of Regenerative Medicine, and
| | - Aditi U Gurkar
- 1 Aging Institute.,7 Division of Geriatric Medicine, Department of Medicine.,8 Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
| | - Ana L Mora
- 1 Aging Institute.,2 Division of Pulmonary Allergy and Critical Care Medicine, and.,9 Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
95
|
Cabrera-Andrade A, López-Cortés A, Jaramillo-Koupermann G, Paz-y-Miño C, Pérez-Castillo Y, Munteanu CR, González-Díaz H, Pazos A, Tejera E. Gene Prioritization through Consensus Strategy, Enrichment Methodologies Analysis, and Networking for Osteosarcoma Pathogenesis. Int J Mol Sci 2020; 21:E1053. [PMID: 32033398 PMCID: PMC7038221 DOI: 10.3390/ijms21031053] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/30/2020] [Accepted: 01/30/2020] [Indexed: 12/12/2022] Open
Abstract
Osteosarcoma is the most common subtype of primary bone cancer, affecting mostly adolescents. In recent years, several studies have focused on elucidating the molecular mechanisms of this sarcoma; however, its molecular etiology has still not been determined with precision. Therefore, we applied a consensus strategy with the use of several bioinformatics tools to prioritize genes involved in its pathogenesis. Subsequently, we assessed the physical interactions of the previously selected genes and applied a communality analysis to this protein-protein interaction network. The consensus strategy prioritized a total list of 553 genes. Our enrichment analysis validates several studies that describe the signaling pathways PI3K/AKT and MAPK/ERK as pathogenic. The gene ontology described TP53 as a principal signal transducer that chiefly mediates processes associated with cell cycle and DNA damage response It is interesting to note that the communality analysis clusters several members involved in metastasis events, such as MMP2 and MMP9, and genes associated with DNA repair complexes, like ATM, ATR, CHEK1, and RAD51. In this study, we have identified well-known pathogenic genes for osteosarcoma and prioritized genes that need to be further explored.
Collapse
Affiliation(s)
- Alejandro Cabrera-Andrade
- Grupo de Bio-Quimioinformática, Universidad de Las Américas, Quito 170125, Ecuador;
- Carrera de Enfermería, Facultad de Ciencias de la Salud, Universidad de Las Américas, Quito 170125, Ecuador
- RNASA-IMEDIR, Computer Sciences Faculty, University of A Coruna, 15071 A Coruña, Spain; (A.L.-C.); (C.R.M.); (A.P.)
| | - Andrés López-Cortés
- RNASA-IMEDIR, Computer Sciences Faculty, University of A Coruna, 15071 A Coruña, Spain; (A.L.-C.); (C.R.M.); (A.P.)
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170129, Ecuador;
| | - Gabriela Jaramillo-Koupermann
- Laboratorio de Biología Molecular, Subproceso de Anatomía Patológica, Hospital de Especialidades Eugenio Espejo, Quito 170403, Ecuador;
| | - César Paz-y-Miño
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170129, Ecuador;
| | - Yunierkis Pérez-Castillo
- Grupo de Bio-Quimioinformática, Universidad de Las Américas, Quito 170125, Ecuador;
- Escuela de Ciencias Físicas y Matemáticas, Universidad de Las Américas, Quito 170125, Ecuador
| | - Cristian R. Munteanu
- RNASA-IMEDIR, Computer Sciences Faculty, University of A Coruna, 15071 A Coruña, Spain; (A.L.-C.); (C.R.M.); (A.P.)
- Biomedical Research Institute of A Coruña (INIBIC), University Hospital Complex of A Coruña (CHUAC), 15006 A Coruña, Spain
- Centro de Investigación en Tecnologías de la Información y las Comunicaciones (CITIC), Campus de Elviña s/n, 15071 A Coruña, Spain
| | - Humbert González-Díaz
- Department of Organic Chemistry II, University of the Basque Country UPV/EHU, 48940 Leioa, Spain
- IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain;
| | - Alejandro Pazos
- RNASA-IMEDIR, Computer Sciences Faculty, University of A Coruna, 15071 A Coruña, Spain; (A.L.-C.); (C.R.M.); (A.P.)
- Biomedical Research Institute of A Coruña (INIBIC), University Hospital Complex of A Coruña (CHUAC), 15006 A Coruña, Spain
- Centro de Investigación en Tecnologías de la Información y las Comunicaciones (CITIC), Campus de Elviña s/n, 15071 A Coruña, Spain
| | - Eduardo Tejera
- Grupo de Bio-Quimioinformática, Universidad de Las Américas, Quito 170125, Ecuador;
- Facultad de Ingeniería y Ciencias Agropecuarias, Universidad de Las Américas, Quito 170125, Ecuador
| |
Collapse
|
96
|
Jin XF, Spöttl G, Maurer J, Nölting S, Auernhammer CJ. Inhibition of Wnt/β-Catenin Signaling in Neuroendocrine Tumors in vitro: Antitumoral Effects. Cancers (Basel) 2020; 12:cancers12020345. [PMID: 32033025 PMCID: PMC7072467 DOI: 10.3390/cancers12020345] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 01/18/2020] [Accepted: 01/30/2020] [Indexed: 12/17/2022] Open
Abstract
Background and aims: Inhibition of Wnt/β-catenin signaling by specific inhibitors is currently being investigated as an antitumoral strategy for various cancers. The role of Wnt/β-catenin signaling in neuroendocrine tumors still needs to be further investigated. Methods: This study investigated the antitumor activity of the porcupine (PORCN) inhibitor WNT974 and the β-catenin inhibitor PRI-724 in human neuroendocrine tumor (NET) cell lines BON1, QGP-1, and NCI-H727 in vitro. NET cells were treated with WNT974, PRI-724, or small interfering ribonucleic acids against β-catenin, and subsequent analyses included cell viability assays, flow cytometric cell cycle analysis, caspase3/7 assays and Western blot analysis. Results: Treatment of NET cells with WNT974 significantly reduced NET cell viability in a dose- and time-dependent manner by inducing NET cell cycle arrest at the G1 and G2/M phases without inducing apoptosis. WNT974 primarily blocked Wnt/β-catenin signaling by the dose- and time-dependent downregulation of low-density lipoprotein receptor-related protein 6 (LRP6) phosphorylation and non-phosphorylated β-catenin and total β-catenin, as well as the genes targeting the latter (c-Myc and cyclinD1). Furthermore, the WNT974-induced reduction of NET cell viability occurred through the inhibition of GSK-3-dependent or independent signaling (including pAKT/mTOR, pEGFR and pIGFR signaling). Similarly, treatment of NET cells with the β-catenin inhibitor PRI-724 caused significant growth inhibition, while the knockdown of β-catenin expression by siRNA reduced NET tumor cell viability of BON1 cells but not of NCI-H727 cells. Conclusions: The PORCN inhibitor WNT974 possesses antitumor properties in NET cell lines by inhibiting Wnt and related signaling. In addition, the β-catenin inhibitor PRI-724 possesses antitumor properties in NET cell lines. Future studies are needed to determine the role of Wnt/β-catenin signaling in NET as a potential therapeutic target.
Collapse
Affiliation(s)
- Xi-Feng Jin
- Department of Internal Medicine 4, University-Hospital, Klinikum der Universitaet Muenchen, Ludwig-Maximilians-University of Munich, 81377 Munich, Germany; (X.-F.J.); (G.S.); (J.M.); (S.N.)
| | - Gerald Spöttl
- Department of Internal Medicine 4, University-Hospital, Klinikum der Universitaet Muenchen, Ludwig-Maximilians-University of Munich, 81377 Munich, Germany; (X.-F.J.); (G.S.); (J.M.); (S.N.)
| | - Julian Maurer
- Department of Internal Medicine 4, University-Hospital, Klinikum der Universitaet Muenchen, Ludwig-Maximilians-University of Munich, 81377 Munich, Germany; (X.-F.J.); (G.S.); (J.M.); (S.N.)
| | - Svenja Nölting
- Department of Internal Medicine 4, University-Hospital, Klinikum der Universitaet Muenchen, Ludwig-Maximilians-University of Munich, 81377 Munich, Germany; (X.-F.J.); (G.S.); (J.M.); (S.N.)
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM), Klinikum der Universitaet Muenchen, Ludwig-Maximilians-University of Munich, Campus Grosshadern, Marchioninistr. 15, 81377 Munich, Germany
| | - Christoph Josef Auernhammer
- Department of Internal Medicine 4, University-Hospital, Klinikum der Universitaet Muenchen, Ludwig-Maximilians-University of Munich, 81377 Munich, Germany; (X.-F.J.); (G.S.); (J.M.); (S.N.)
- Interdisciplinary Center of Neuroendocrine Tumors of the GastroEnteroPancreatic System (GEPNET-KUM), Klinikum der Universitaet Muenchen, Ludwig-Maximilians-University of Munich, Campus Grosshadern, Marchioninistr. 15, 81377 Munich, Germany
- Correspondence:
| |
Collapse
|
97
|
Triana-Martínez F, Loza MI, Domínguez E. Beyond Tumor Suppression: Senescence in Cancer Stemness and Tumor Dormancy. Cells 2020; 9:cells9020346. [PMID: 32028565 PMCID: PMC7072600 DOI: 10.3390/cells9020346] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/23/2020] [Accepted: 01/29/2020] [Indexed: 12/12/2022] Open
Abstract
Here, we provide an overview of the importance of cellular fate in cancer as a group of diseases of abnormal cell growth. Tumor development and progression is a highly dynamic process, with several phases of evolution. The existing evidence about the origin and consequences of cancer cell fate specification (e.g., proliferation, senescence, stemness, dormancy, quiescence, and cell cycle re-entry) in the context of tumor formation and metastasis is discussed. The interplay between these dynamic tumor cell phenotypes, the microenvironment, and the immune system is also reviewed in relation to cancer. We focus on the role of senescence during cancer progression, with a special emphasis on its relationship with stemness and dormancy. Selective interventions on senescence and dormancy cell fates, including the specific targeting of cancer cell populations to prevent detrimental effects in aging and disease, are also reviewed. A new conceptual framework about the impact of synthetic lethal strategies by using senogenics and then senolytics is given, with the promise of future directions on innovative anticancer therapies.
Collapse
|
98
|
Teng Z, Xu S, Lei Q. Tanshinone IIA enhances the inhibitory effect of imatinib on proliferation and motility of acute leukemia cell line TIB‑152 in vivo and in vitro by inhibiting the PI3K/AKT/mTOR signaling pathway. Oncol Rep 2020; 43:503-515. [PMID: 31894340 PMCID: PMC6967082 DOI: 10.3892/or.2019.7453] [Citation(s) in RCA: 6] [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: 04/18/2019] [Accepted: 09/06/2019] [Indexed: 02/07/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) is a malignant hematological disease. Tanshinone IIA (Tan IIA) has antitumor activity in vitro and in vivo. The aim of the present study was to investigate the effects of Tan IIA in combination with imatinib (IM) on the proliferation, apoptosis, migration and invasion of acute T lymphocytic leukemia TIB‑152 cells in vivo and in vitro, and analyze the potential underlying mechanism. Tan IIA and IM, alone and in combination, significantly inhibited proliferation, migration and invasion of TIB‑152 cells, and promoted apoptosis; the effect of co‑treatment with Tan IIA plus IM was enhanced. IGF‑1 promoted the proliferation, migration and invasion of TIB‑152 cells and inhibited apoptosis, while Tan IIA treatment significantly reversed these effects. In vivo experiments demonstrated that treatment with Tan IIA and IM, alone or in combination, significantly inhibited tumor growth in TIB‑152 xenograft mice; the growth inhibition of Tan IIA plus IM was the strongest observed. Western blot analysis revealed that the combination of Tan IIA and IM resulted in significantly lower levels of p‑PI3K, p‑AKT and p‑mTOR in cells and tissues compared with the IM and Tan alone treatment groups. In addition, the combination of Tan IIA and IM significantly decreased the levels of Ki67, cleaved caspase‑3, VEGF and MMP‑9 in cells and tissues, and the level of caspase‑3 was significantly increased. Taken together, the results revealed that Tan IIA enhanced the inhibitory effect of imatinib on TIB‑152 cell proliferation, migration and invasion, and induced apoptosis, which may be associated with inhibition of the PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Zhi Teng
- Department of Hematology, 215 Hospital of Shanxi Nuclear Industry, Xianyang, Shanxi 712000, P.R. China
| | - Shijuan Xu
- Department of Hematology, 215 Hospital of Shanxi Nuclear Industry, Xianyang, Shanxi 712000, P.R. China
| | - Qin Lei
- Department of Hematology, 215 Hospital of Shanxi Nuclear Industry, Xianyang, Shanxi 712000, P.R. China
| |
Collapse
|
99
|
Zhao Y, Li JY, Jiang Q, Zhou XQ, Feng L, Liu Y, Jiang WD, Wu P, Zhou J, Zhao J, Jiang J. Leucine Improved Growth Performance, Muscle Growth, and Muscle Protein Deposition Through AKT/TOR and AKT/FOXO3a Signaling Pathways in Hybrid Catfish Pelteobagrus v achelli × Leiocassis longirostris. Cells 2020; 9:cells9020327. [PMID: 32019276 PMCID: PMC7072317 DOI: 10.3390/cells9020327] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/21/2020] [Accepted: 01/29/2020] [Indexed: 02/06/2023] Open
Abstract
(1) Background: l-leucine (Leu) plays a positive role in regulating protein turnover in skeletal muscle in mammal. However, the molecular mechanism for the effects of Leu on muscle growth and protein deposition is not clearly demonstrated in fish. This study investigated the effects of dietary Leu on growth performance and muscle growth, protein synthesis, and degradation-related signaling pathways of hybrid catfish (Pelteobagrus vachelli♀ × Leiocassis longirostris♂). (2) Methods: A total of 630 hybrid catfish (23.19 ± 0.20 g) were fed 6 different experimental diets containing graded levels of Leu at 10.0 (control), 15.0, 20.0, 25.0, 30.0, 35.0, and 40.0 g Leu kg-1 for 8 weeks. (3) Results: Results showed that dietary Leu increased percent weight gain (PWG), specific growth rate (SGR), FI (feed intake), feed efficiency (FE), protein efficiency ratio (PER), muscle fibers diameter, and muscle fibers density; up-regulated insulin-like growth factor I (IGF-I), insulin-like growth factor I receptor (IGF-IR), proliferating cell nuclear antigen (PCNA), myogenic regulation factors (MyoD, Myf5, MyoG, and Mrf4), and MyHC mRNA levels; increased muscle protein synthesis via regulating the AKT/TOR signaling pathway; and attenuated protein degradation via regulating the AKT/FOXO3a signaling pathway. (4) Conclusions: These results suggest that Leu has potential role to improve muscle growth and protein deposition in fish, which might be due to the regulation of IGF mRNA expression, muscle growth related gene, and protein synthesis and degradation-related signaling pathways. Based on the broken-line model, the Leu requirement of hybrid catfish (23.19-54.55 g) for PWG was estimated to be 28.10 g kg-1 of the diet (73.04 g kg-1 of dietary protein). These results will improve our understanding of the mechanisms responsible for muscle growth and protein deposition effects of Leu in fish.
Collapse
Affiliation(s)
- Ye Zhao
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Jin-Yang Li
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Qin Jiang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiao-Qiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Lin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Wei-Dan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Pei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Jian Zhou
- Fisheries Institute of Sichuan Academy of Agricultural Science, Chengdu 611731, China
| | - Juan Zhao
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Jun Jiang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
- Correspondence: ; Tel.: +86-28-8629-1133
| |
Collapse
|
100
|
Lu ZW, Wen D, Wei WJ, Han LT, Xiang J, Wang YL, Wang Y, Liao T, Ji QH. Silencing of PPM1D inhibits cell proliferation and invasion through the p38 MAPK and p53 signaling pathway in papillary thyroid carcinoma. Oncol Rep 2020; 43:783-794. [PMID: 31922231 PMCID: PMC7040886 DOI: 10.3892/or.2020.7458] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 12/04/2019] [Indexed: 02/06/2023] Open
Abstract
Endeavors towards identifying key molecular markers for early diagnosis and treatment are driving the clinical study of papillary thyroid carcinoma (PTC). Recent studies have indicated that protein phosphatase, Mg2+/Mn2+ dependent, 1D (PPM1D) exerts an oncogenic function by increasing cell proliferation, migration and invasion in various cancer types. In addition, PPM1D has a high frequency of genetic alterations and has been proposed as a tumor driver in thyroid cancer, making PPM1D an attractive potential oncotarget for cancer treatment. The aims of the present study were to investigate the downstream targets of PPM1D and the potential molecular mechanisms of its oncogenic activities, as well as its clinical significance in PTC. As anticipated, PPM1D overexpression was confirmed in PTC clinical specimens. Furthermore, knockdown of PPM1D in thyroid cancer cell lines significantly suppressed the proliferation, migration and invasion but facilitated cell apoptosis. The protein levels of phosphorylated p38 mitogen-activated protein kinase (MAPK), p53 and Bax were increased in PPM1D-knockdown cells, while inhibition of p38 phosphorylation restored cell migration, proliferation and cell apoptosis. In addition, silencing of PPM1D expression induced nuclear translocation of p53 in K-1 and TPC-1 cells. The present results demonstrated that PPM1D regulated p38 MAPK and p53 signaling pathways to promote thyroid cancer progression. Collectively with the clinical results, these data qualified PPM1D as a potential diagnostic biomarker and therapeutic target in human thyroid cancer.
Collapse
Affiliation(s)
- Zhong-Wu Lu
- Department of Head and Neck Surgery, Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Duo Wen
- Department of Head and Neck Surgery, Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Wen-Jun Wei
- Department of Head and Neck Surgery, Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Li-Tao Han
- Department of Head and Neck Surgery, Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Jun Xiang
- Department of Head and Neck Surgery, Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Yu-Long Wang
- Department of Head and Neck Surgery, Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Yu Wang
- Department of Head and Neck Surgery, Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Tian Liao
- Department of Head and Neck Surgery, Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| | - Qing-Hai Ji
- Department of Head and Neck Surgery, Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|