1
|
Fu XT, Qie JB, Chen JF, Gao Z, Li XG, Feng SR, Dong EF, Shi YH, Tang Z, Liu WR, Zhang X, Huang A, Luo XM, Wu WX, Gao Q, Zhou J, Li T, Fan J, Ding ZB. Inhibition of SIRT1 relieves hepatocarcinogenesis via alleviating autophagy and inflammation. Int J Biol Macromol 2024; 278:134120. [PMID: 39074701 DOI: 10.1016/j.ijbiomac.2024.134120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/15/2024] [Accepted: 07/22/2024] [Indexed: 07/31/2024]
Abstract
Imbalanced Sirtuin 1 (SIRT1) levels may lead to liver diseases through abnormal regulation of autophagy, but the roles of SIRT1-regulated autophagy in hepatocellular carcinoma are still controversial. In this study, we found that SIRT1 mRNA and protein levels were upregulated in hepatocellular carcinoma, and high SIRT1 expression hinted an advanced stage and a poor prognosis. The differentially expressed proteins were significantly elevated in autophagy, cellular response to stress, and immune signaling pathways. In a thioacetamide-induced hepatocellular carcinoma mouse model, we found that SIRT1 expression was highly increased with increased autophagy and excessive macrophage inflammatory response. Next, we established a Hepa 1-6 cells and macrophage co-culture system in vitro to model the alteration of tumor microenvironment, and found that the medium from CCl4-treated or SIRT1-overexpressing Hepa 1-6 cells triggered the polarization of macrophage M1, and the culture medium derived from M1 macrophage promoted Hepa 1-6 cells growth and intracellular oxidative stress. The progression of liver fibrosis in the CCl4-induced liver fibrosis mouse model showed that inhibition of SIRT1 alleviated inflammatory response and ameliorated liver fibrosis. These findings suggest that SIRT1-regulated autophagy and inflammation are oncogenic in hepatocarcinogenesis.
Collapse
Affiliation(s)
- Xiu-Tao Fu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai, China
| | - Jing-Bo Qie
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, China; Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jia-Feng Chen
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai, China
| | - Zheng Gao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai, China
| | - Xiao-Gang Li
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai, China
| | - Shan-Ru Feng
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai, China
| | - En-Fu Dong
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai, China
| | - Ying-Hong Shi
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai, China
| | - Zheng Tang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai, China
| | - Wei-Ren Liu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai, China
| | - Xin Zhang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai, China
| | - Ao Huang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai, China
| | - Xuan-Ming Luo
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai, China
| | - Wei-Xun Wu
- Department of Liver Surgery, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, China
| | - Qiang Gao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai, China
| | - Jian Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai, China; Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China.
| | - Jia Fan
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai, China; Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Zhen-Bin Ding
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai, China; Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai, China; Department of Liver Surgery, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, China.
| |
Collapse
|
2
|
Yang Z, Verghese M, Yang S, Shah P, He YY. The m 6A reader YTHDC2 regulates UVB-induced DNA damage repair and histone modification. Photochem Photobiol 2024; 100:1031-1040. [PMID: 38190286 PMCID: PMC11228125 DOI: 10.1111/php.13904] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/10/2024]
Abstract
Ultraviolet B (UVB) radiation represents a major carcinogen for the development of all skin cancer types. Mechanistically, UVB induces damage to DNA in the form of lesions, including cyclobutane pyrimidine dimers (CPDs). Disruption of the functional repair processes, such as nucleotide excision repair (NER), allows persistence of DNA damage and contributes to skin carcinogenesis. Recent work has implicated m6A RNA methylation and its regulatory proteins as having critical roles in facilitating UVB-induced DNA damage repair. However, the biological functions of the m6A reader YTHDC2 are unknown in this context. Here, we show that YTHDC2 inhibition enhances the repair of UVB-induced DNA damage. We discovered that YTHDC2 inhibition increased the expression of PTEN while it decreased the expression of the PRC2 component SUZ12 and the levels of the histone modification H3K27me3. However, none of these functions were causally linked to the improvements in DNA repair, suggesting that the mechanism utilized by YTHDC2 may be unconventional. Moreover, inhibition of the m6A writer METTL14 reversed the effect of YTHDC2 inhibition on DNA repair while inhibition of the m6A eraser FTO mimicked the effect of YTHDC2 inhibition, indicating that YTHDC2 may regulate DNA repair through the m6A pathway. Finally, compared to normal human skin, YTHDC2 expression was upregulated in human cutaneous squamous cell carcinomas (cSCC), suggesting that it may function as a tumor-promoting factor in skin cancer. Taken together, our findings demonstrate that the m6A reader YTHDC2 plays a role in regulating UVB-induced DNA damage repair and may serve as a potential biomarker in cSCC.
Collapse
Affiliation(s)
- Zizhao Yang
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, Illinois, USA
- These authors contributed equally to this work
| | - Michelle Verghese
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, Illinois, USA
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois, USA
- These authors contributed equally to this work
| | - Seungwon Yang
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, Illinois, USA
| | - Palak Shah
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, Illinois, USA
- Committee on Molecular Pathogenesis and Molecular Medicine, University of Chicago, Chicago, Illinois, USA
| | - Yu-Ying He
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, Illinois, USA
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois, USA
- Committee on Molecular Pathogenesis and Molecular Medicine, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
3
|
Qian L, Zhang Y, Wang G, Li B, Zhou H, Qiu J, Qin L. miR-29a-SIRT1-Wnt/β-Catenin Axis Regulates Tumor Progression and Survival in Hepatocellular Carcinoma. Biochem Genet 2024; 62:1895-1913. [PMID: 37776468 DOI: 10.1007/s10528-023-10521-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 09/05/2023] [Indexed: 10/02/2023]
Abstract
Sirtuin 1 (SIRT1) participates in the initiation and evolution of hepatocellular carcinoma (HCC). However, the specific mechanism of SIRT1 in HCC remains unclear. The mRNA expression of miR-29a in HCC were identified by qRT-PCR. miR-29a mimic and inhibitor were employed. The alteration of biological behavior was evaluated by Cell Counting Kit-8 (CCK8), clone formation, transwell and wound-healing assay. SIRT1 was verified to be a target gene which directly regulated by miR-29a. Luciferase reporter assay and co-IP were employed to evaluate the direct binding of miR-29a and SIRT1. Animal model was used to evaluate its function on tumor growth and metastasis in vivo. The relationship between miR-29a/SIRT1 and prognosis of HCC patients was analyzed. SIRT1 overexpression accompanied by low expression of miR-29a were detected in HCC which was negatively correlated, and associated with overall survival, vascular invasion and TNM stage. Up-regulation of miR-29a suppressed cell growth and motility. Deprivation of miR-29a expression led to opposite effect. The direct binding of miR-29a to SIRT1 was confirmed by luciferase reporter assay and co-IP. miR-29a repressed SIRT1, DKK2 and β-catenin, but their expression was obviously elevated by miR-29a inhibitor. Animal model suggested miR-29a could reduce the expression of SIRT1, thereby inhibiting HCC growth and metastasis by inactivating Wnt/β-catenin pathway. Low expression of miR-29a and high expression of SIRT1 predicted shorter survival time in HCC patients. miR-29a had the function of tumor suppressor which directly inhibited oncogenic SIRT1. The loss of miR-29a led to up-regulation of SIRT1, aggravate malignant transformation and poor prognosis of HCC.
Collapse
Affiliation(s)
- Liqiang Qian
- Department of General Surgery, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, China
- Department of General Surgery, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, China
| | - Yanjun Zhang
- School of Health and Social Care, Shanghai Urban Construction Vocational College, Shanghai, China
| | - Gang Wang
- Department of General Surgery, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, China
| | - Bin Li
- Department of General Surgery, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, China
| | - Hemei Zhou
- Department of General Surgery, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, China
| | - Jie Qiu
- Department of General Surgery, Suzhou Ninth Hospital Affiliated to Soochow University, Suzhou, China
| | - Lei Qin
- Department of General Surgery, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
4
|
Park G, Cui YH, Yang S, Sun M, Wilkinson E, Li H, Zhang YB, Chen J, Bissonnette M, Lin W, He YY. Moderate Low UVB Irradiation Modulates Tumor-associated Macrophages and Dendritic Cells and Promotes Antitumor Immunity in Tumor-bearing Mice. Photochem Photobiol 2023; 99:850-856. [PMID: 35962531 PMCID: PMC9884319 DOI: 10.1111/php.13684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 07/26/2022] [Indexed: 01/31/2023]
Abstract
Excessive, high doses of ultraviolet B (UVB) UVB irradiation are known to cause skin cancer, aging and immunosuppression. On the contrary, moderate low doses of UVB irradiation are shown to be essential and beneficial to human health, including a tumor-suppressive effect. However, the mechanism by which low levels of UVB suppress tumorigenesis remains unclear. Here, using tumor-bearing mouse models, we show that moderate low repetitive UVB irradiation increases the percentage of activated CD4+ and CD8+ T cells, and CD103+ conventional type 1 dendritic cells (cDC1s), while it decreases the number of immunosuppressive, M2-like macrophages in the tumors. Finally, in mice, deletion of Batf3, a transcription factor critical for the development of conventional dendritic cells, including the CD103+ cDC1s, showed increased tumor growth in both sham- and UVB-irradiated mice. Our findings demonstrate that moderate low UVB irradiation inhibits M2-like tumor-associated macrophages, increases CD103+ cDC1s and promotes antitumor immunity in mice with an established tumor.
Collapse
Affiliation(s)
- Gayoung Park
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, USA
| | - Yan-Hong Cui
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, USA
| | - Seungwon Yang
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, USA
| | - Ming Sun
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, USA
| | - Emma Wilkinson
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, USA
| | - Haixia Li
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, USA
| | - Yuhan Blair Zhang
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, USA
| | - Jing Chen
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL, USA
| | - Marc Bissonnette
- Department of Medicine, Section of Gastroenterology, Hepatology & Nutrition, University of Chicago, Chicago, IL, USA
| | - Wenbin Lin
- Department of Chemistry, University of Chicago, Chicago, IL, USA
| | - Yu-Ying He
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, USA
| |
Collapse
|
5
|
Sirtuin1 (SIRT1) is involved in the anticancer effect of black raspberry anthocyanins in colorectal cancer. Eur J Nutr 2023; 62:395-406. [PMID: 36056948 DOI: 10.1007/s00394-022-02989-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 08/11/2022] [Indexed: 02/07/2023]
Abstract
PURPOSE Abnormal acetylation modification is a common epigenetic change in tumorigenesis and is closely related to the progression of colorectal cancer (CRC). Our previous studies have suggested that black raspberry (BRB) anthocyanins have a significant chemopreventive effect against CRC. This study investigated whether protein acetylation plays an important role in BRB anthocyanins-mediated regulation of CRC progression. METHODS We used the AOM-induced CRC mouse model and the CRC cell lines SW480 and Caco-2 to explore the potential role of acetylation of histone H4 and NF-κB signaling pathway-related proteins (non-histone proteins) in the antitumor process mediated by BRB anthocyanins. The expression of related proteins was detected by western blot. ROS level was detected by immunofluorescence. RESULTS BRB anthocyanins affected the acetylation level by down-regulating the expression of Sirtuin1 (SIRT1) and up-regulating the expression of MOF and EP300. The acetylation level of lysine sites on histone H4 (H4K5, H4K12 and H4K16) was increased. Furthermore, following BRB anthocyanins treatment, the expression of ac-p65 was significantly up-regulated and the NF-κB signal pathway was activated, which in turn up-regulated Bax expression and inhibited Bcl-2, cyclin-D1, c-myc and NLRP3 expression to promote CRC cell cycle arrest, apoptosis and relieve inflammation. CONCLUSION The findings suggested that protein acetylation could play a critical role in BRB anthocyanins-regulated CRC development.
Collapse
|
6
|
Krasikova YS, Lavrik OI, Rechkunova NI. The XPA Protein-Life under Precise Control. Cells 2022; 11:cells11233723. [PMID: 36496984 PMCID: PMC9739396 DOI: 10.3390/cells11233723] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 11/24/2022] Open
Abstract
Nucleotide excision repair (NER) is a central DNA repair pathway responsible for removing a wide variety of DNA-distorting lesions from the genome. The highly choreographed cascade of core NER reactions requires more than 30 polypeptides. The xeroderma pigmentosum group A (XPA) protein plays an essential role in the NER process. XPA interacts with almost all NER participants and organizes the correct NER repair complex. In the absence of XPA's scaffolding function, no repair process occurs. In this review, we briefly summarize our current knowledge about the XPA protein structure and analyze the formation of contact with its protein partners during NER complex assembling. We focus on different ways of regulation of the XPA protein's activity and expression and pay special attention to the network of post-translational modifications. We also discuss the data that is not in line with the currently accepted hypothesis about the functioning of the XPA protein.
Collapse
Affiliation(s)
- Yuliya S. Krasikova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Olga I. Lavrik
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, Novosibirsk 630090, Russia
- Department of Natural Sciences, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Nadejda I. Rechkunova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, Novosibirsk 630090, Russia
- Correspondence:
| |
Collapse
|
7
|
Volatier T, Schumacher B, Cursiefen C, Notara M. UV Protection in the Cornea: Failure and Rescue. BIOLOGY 2022; 11:biology11020278. [PMID: 35205145 PMCID: PMC8868636 DOI: 10.3390/biology11020278] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/28/2022] [Accepted: 01/28/2022] [Indexed: 01/07/2023]
Abstract
Simple Summary The sun is a deadly laser, and its damaging rays harm exposed tissues such as our skin and eyes. The skin’s protection and repair mechanisms are well understood and utilized in therapeutic approaches while the eye lacks such complete understanding of its defenses and therefore often lacks therapeutic support in most cases. The aim here was to document the similarities and differences between the two tissues as well as understand where current research stands on ocular, particularly corneal, ultraviolet protection. The objective is to identify what mechanisms may be best suited for future investigation and valuable therapeutic approaches. Abstract Ultraviolet (UV) irradiation induces DNA lesions in all directly exposed tissues. In the human body, two tissues are chronically exposed to UV: the skin and the cornea. The most frequent UV-induced DNA lesions are cyclobutane pyrimidine dimers (CPDs) that can lead to apoptosis or induce tumorigenesis. Lacking the protective pigmentation of the skin, the transparent cornea is particularly dependent on nucleotide excision repair (NER) to remove UV-induced DNA lesions. The DNA damage response also triggers intracellular autophagy mechanisms to remove damaged material in the cornea; these mechanisms are poorly understood despite their noted involvement in UV-related diseases. Therapeutic solutions involving xenogenic DNA-repair enzymes such as T4 endonuclease V or photolyases exist and are widely distributed for dermatological use. The corneal field lacks a similar set of tools to address DNA-lesions in photovulnerable patients, such as those with genetic disorders or recently transplanted tissue.
Collapse
Affiliation(s)
- Thomas Volatier
- Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 62, 50937 Cologne, Germany; (C.C.); (M.N.)
- Correspondence:
| | - Björn Schumacher
- Cologne Excellence Cluster for Cellular Stress Responses, Aging-Associated Diseases (CECAD) and Center for Molecular Medicine (CMMC), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany;
| | - Claus Cursiefen
- Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 62, 50937 Cologne, Germany; (C.C.); (M.N.)
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, 21, 50931 Cologne, Germany
| | - Maria Notara
- Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 62, 50937 Cologne, Germany; (C.C.); (M.N.)
- Cologne Excellence Cluster for Cellular Stress Responses, Aging-Associated Diseases (CECAD) and Center for Molecular Medicine (CMMC), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany;
| |
Collapse
|
8
|
Adams AC, Macy AM, Saboda K, Dickinson SE, Glembocki DJ, Roe DJ, Hastings KT. Solar Simulated Light Induces Cutaneous Squamous Cell Carcinoma in Inbred Mice: A Clinically Relevant Model to Investigate T-Cell Responses. J Invest Dermatol 2021; 141:2990-2993.e6. [PMID: 34252399 PMCID: PMC9261959 DOI: 10.1016/j.jid.2021.06.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/26/2021] [Accepted: 06/09/2021] [Indexed: 10/20/2022]
Affiliation(s)
- Anngela C Adams
- Department of Basic Medical Sciences, College of Medicine - Phoenix, The University of Arizona, Phoenix, Arizona, USA
| | - Anne M Macy
- Department of Basic Medical Sciences, College of Medicine - Phoenix, The University of Arizona, Phoenix, Arizona, USA
| | | | - Sally E Dickinson
- Cancer Center, The University of Arizona, Tucson, Arizona, USA; Department of Pharmacology, College of Medicine - Tucson, The University of Arizona, Tucson, Arizona, USA
| | - David J Glembocki
- US Dermatology Partners Pathology Laboratory, Scottsdale, Arizona, USA
| | - Denise J Roe
- Cancer Center, The University of Arizona, Tucson, Arizona, USA; Department of Epidemiology and Biostatistics, Mel & Enid Zuckerman College of Public Health, The University of Arizona, Tucson, Arizona, USA
| | - Karen Taraszka Hastings
- Department of Basic Medical Sciences, College of Medicine - Phoenix, The University of Arizona, Phoenix, Arizona, USA; Cancer Center, The University of Arizona, Tucson, Arizona, USA.
| |
Collapse
|
9
|
Yousafzai NA, Jin H, Ullah M, Wang X. Recent advances of SIRT1 and implications in chemotherapeutics resistance in cancer. Am J Cancer Res 2021; 11:5233-5248. [PMID: 34873458 PMCID: PMC8640807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 08/20/2021] [Indexed: 06/13/2023] Open
Abstract
Cancer is a big group of diseases and one of the leading causes of mortality worldwide. Despite enormous studies and efforts are being carried out in understanding the cancer and developing drugs against tumorigenesis, drug resistance is the main obstacle in cancer treatments. Chemotherapeutic treatment is an important part of cancer treatment and drug resistance is getting gradually multidimensional with the advancement of studies in cancer. The underlying mechanisms of drug resistance are largely unknown. Sirtuin1 (SIRT1) is a type of the Class III histone deacetylase family that is distinctively dependent on nicotinamide adenine dinucleotide (NAD+) for catalysis reaction. SIRT1 is a molecule which upon upregulation directly influences tumor progression, metastasis, tumor cell apoptosis, autophagy, DNA repair, as well as other interlinked tumorigenesis mechanism. It is involved in drug metabolism, apoptosis, DNA damage, DNA repair, and autophagy, which are key hallmarks of drug resistance and may contribute to multidrug resistance. Thus, understanding the role of SIRT1 in drug resistance could be important. This study focuses on the SIRT1 based mechanisms that might be a potential underlying approach in the development of cancer drug resistance and could be a potential target for drug development.
Collapse
Affiliation(s)
- Neelum Aziz Yousafzai
- Department of Medical Oncology, Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang UniversityHangzhou 310020, Zhejiang, China
- Department of Medical and Health Sciences, University of Poonch RawalakotAJK 12350, Pakistan
| | - Hongchuan Jin
- Department of Medical Oncology, Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang UniversityHangzhou 310020, Zhejiang, China
| | - Mujib Ullah
- Institute for Immunity and Transplantation, Stem Cell Biology and Regenerative Medicine, School of Medicine, Stanford UniversityPalo Alto, CA 94304, United States
| | - Xian Wang
- Department of Medical Oncology, Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang UniversityHangzhou 310020, Zhejiang, China
| |
Collapse
|
10
|
Garcia-Peterson LM, Li X. Trending topics of SIRT1 in tumorigenicity. Biochim Biophys Acta Gen Subj 2021; 1865:129952. [PMID: 34147543 DOI: 10.1016/j.bbagen.2021.129952] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/09/2021] [Accepted: 06/15/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Carcinogenesis is governed by a series of genetic alterations and epigenetic changes that lead to aberrant patterns in neoplastic cells. Sirtuin-1(SIRT1), an NAD+-dependent protein deacetylase, is capable of deacetylating histones and non-histone substrates that regulate various physiological activities during tumorigenesis. Recent studies have identified the role of SIRT1 in different stages of cancer, including genome instability, tumor initiation, proliferation, metabolism, and therapeutic response. However, the action of SIRT1 has been reported to be both oncogenic and tumor suppressive during carcinogenesis. Consequently, the biological functions of SIRT1 in cancer remain controversial. SCOPE OF REVIEW We highlight the most recent findings on SIRT1 in different stages of tumorigenesis, and update the current status of SIRT1 small molecule modulators in clinical application of cancer treatment. MAJOR CONCLUSION By targeting both tumor suppressors and oncogenic proteins, SIRT1 has a bifunctional role at different stages of tumorigenesis. The impact of SIRT1 on tumorigenesis is also distinct at different stages and is dependent on its dosages. SIRT1 suppresses tumor initiation through its functions in promoting DNA repair, increasing genome stability, and inhibiting inflammation at the pre-cancer stage. However, SIRT1 enhances tumor proliferation, survival, and drug resistance through its roles in anti-apoptosis, pro-tumor metabolism, and anti-inflammation (inhibition of anti-tumor immunity) at the stages of tumor progression, metastasis, and relapse. Consequently, both SIRT1 inhibitors and activators have been explored for cancer treatment. GENERAL SIGNIFICANCE Better understanding the dose- and stage-dependent roles of SIRT1 in each cancer type can provide new avenues of exploration for therapy development.
Collapse
Affiliation(s)
- Liz M Garcia-Peterson
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Xiaoling Li
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
11
|
Katayoshi T, Nakajo T, Tsuji-Naito K. Restoring NAD + by NAMPT is essential for the SIRT1/p53-mediated survival of UVA- and UVB-irradiated epidermal keratinocytes. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2021; 221:112238. [PMID: 34130091 DOI: 10.1016/j.jphotobiol.2021.112238] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 05/06/2021] [Accepted: 06/01/2021] [Indexed: 12/18/2022]
Abstract
Nicotinamide adenine dinucleotide (NAD+) is a crucial coenzyme in energy production. The imbalance of NAD+ synthesis has been found to trigger age-related diseases, such as metabolic disorders, cancer, and neurodegenerative diseases. Also, UV irradiation induces NAD+ depletion in the skin. In mammals, nicotinamide phosphoribosyltransferase (NAMPT) is the rate-limiting enzyme in the NAD+ salvage pathway and essential for NAD+ homeostasis. However, but few studies have focused on the role of NAMPT in response to UV irradiation. Here, we show that NAMPT prevents NAD+ depletion in epidermal keratinocytes to protect against the mild-dose UVA and UVB (UVA/B)-induced proliferation defects. We showed that poly(ADP-ribose) polymerase (PARP) inhibitor rescued the NAD+ depletion in UVA/B-irradiated human keratinocytes, confirming that PAPR transiently exhausts cellular NAD+ to repair DNA damage. Notably, the treatment with a NAMPT inhibitor exacerbated the UVA/B-induced loss of energy production and cell viability. Moreover, the NAMPT inhibitor abrogated the sirtuin-1 (SIRT1)-mediated deacetylation of p53 and significantly inhibited the proliferation of UVA/B-irradiated cells, suggesting that the NAMPT-NAD+-SIRT1 axis regulates p53 functions upon UVA/B stress. The supplementation with NAD+ intermediates, nicotinamide mononucleotide and nicotinamide riboside, rescued the UVA/B-induced phenotypes in the absence of NAMPT activity. Therefore, NAD+ homeostasis is likely essential for the protection of keratinocytes from UV stress in mild doses. Since the skin is continuously exposed to UVA/B irradiation, understanding the protective role of NAMPT in UV stress will help prevent and treat skin photoaging.
Collapse
Affiliation(s)
- Takeshi Katayoshi
- DHC Corporation Laboratories, Division 2, 2-42 Hamada, Mihama-ku, Chiba 261-0025, Japan.
| | - Takahisa Nakajo
- DHC Corporation Laboratories, Division 2, 2-42 Hamada, Mihama-ku, Chiba 261-0025, Japan
| | - Kentaro Tsuji-Naito
- DHC Corporation Laboratories, Division 2, 2-42 Hamada, Mihama-ku, Chiba 261-0025, Japan
| |
Collapse
|
12
|
Cui YH, Yang S, Wei J, Shea CR, Zhong W, Wang F, Shah P, Kibriya MG, Cui X, Ahsan H, He C, He YY. Autophagy of the m 6A mRNA demethylase FTO is impaired by low-level arsenic exposure to promote tumorigenesis. Nat Commun 2021; 12:2183. [PMID: 33846348 PMCID: PMC8041927 DOI: 10.1038/s41467-021-22469-6] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 03/16/2021] [Indexed: 12/14/2022] Open
Abstract
Here we show that FTO as an N6-methyladenosine (m6A) RNA demethylase is degraded by selective autophagy, which is impaired by low-level arsenic exposure to promote tumorigenesis. We found that in arsenic-associated human skin lesions, FTO is upregulated, while m6A RNA methylation is downregulated. In keratinocytes, chronic relevant low-level arsenic exposure upregulated FTO, downregulated m6A RNA methylation, and induced malignant transformation and tumorigenesis. FTO deletion inhibited arsenic-induced tumorigenesis. Moreover, in mice, epidermis-specific FTO deletion prevented skin tumorigenesis induced by arsenic and UVB irradiation. Targeting FTO genetically or pharmacologically inhibits the tumorigenicity of arsenic-transformed tumor cells. We identified NEDD4L as the m6A-modified gene target of FTO. Finally, arsenic stabilizes FTO protein through inhibiting p62-mediated selective autophagy. FTO upregulation can in turn inhibit autophagy, leading to a positive feedback loop to maintain FTO accumulation. Our study reveals FTO-mediated dysregulation of mRNA m6A methylation as an epitranscriptomic mechanism to promote arsenic tumorigenicity.
Collapse
Affiliation(s)
- Yan-Hong Cui
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, USA
| | - Seungwon Yang
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, USA
| | - Jiangbo Wei
- Departments of Chemistry, Department of Biochemistry and Molecular Biology, Institute for Biophysical Dynamics, University of Chicago, Chicago, IL, USA
| | - Christopher R Shea
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, USA
| | - Wen Zhong
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, USA
- Department of Radiation Oncology, 4th Affiliated Hospital, China Medical University, Shenyang, China
| | - Fang Wang
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, USA
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, China
| | - Palak Shah
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Muhammad G Kibriya
- Institute for Population and Precision Health, Department of Public Health Sciences, The University of Chicago, Chicago, IL, USA
| | - Xiaolong Cui
- Departments of Chemistry, Department of Biochemistry and Molecular Biology, Institute for Biophysical Dynamics, University of Chicago, Chicago, IL, USA
| | - Habibul Ahsan
- Institute for Population and Precision Health, Department of Public Health Sciences, The University of Chicago, Chicago, IL, USA
| | - Chuan He
- Departments of Chemistry, Department of Biochemistry and Molecular Biology, Institute for Biophysical Dynamics, University of Chicago, Chicago, IL, USA
- Howard Hughes Medical Institute, University of Chicago, Chicago, IL, USA
| | - Yu-Ying He
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
13
|
Sirtuin 1 and Skin: Implications in Intrinsic and Extrinsic Aging-A Systematic Review. Cells 2021; 10:cells10040813. [PMID: 33917352 PMCID: PMC8067363 DOI: 10.3390/cells10040813] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 03/28/2021] [Accepted: 04/02/2021] [Indexed: 02/06/2023] Open
Abstract
Skin, as the outermost organ of the body, is constantly exposed to both intrinsic and extrinsic causative factors of aging. Intrinsic aging is related to compromised cellular proliferative capacity, and may be accelerated by harmful environmental influences with the greatest significance of ultraviolet radiation exposure, contributing not only to premature aging, but also to skin carcinogenesis. The overall skin cancer burden and steadily increasing global antiaging market provide an incentive for searching novel targets to improve skin resistance against external injury. Sirtuin 1, initially linked to extension of yeast and rodent lifespan, plays a key role in epigenetic modification of proteins, histones, and chromatin by which regulates the expression of genes implicated in the oxidative stress response and apoptosis. The spectrum of cellular pathways regulated by sirtuin 1 suggests its beneficial impact on skin aging. However, the data on its role in carcinogenesis remains controversial. The aim of this review was to discuss the relevance of sirtuin 1 in skin aging, in the context of intrinsic factors, related to genetic premature aging syndromes, as well as extrinsic modifiable ones, with the assessment of its future application. PubMed were searched from inception to 4 January 2021 for relevant papers with further search carried out on ClinicalTrials.gov. The systematic review included 46 eligible original articles. The evidence from numerous studies proves sirtuin 1 significance in both chronological and premature aging as well as its dual role in cancer development. Several botanical compounds hold the potential to improve skin aging symptoms.
Collapse
|
14
|
Russell-Goldman E, Murphy GF. The Pathobiology of Skin Aging: New Insights into an Old Dilemma. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:1356-1369. [PMID: 32246919 PMCID: PMC7481755 DOI: 10.1016/j.ajpath.2020.03.007] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 02/19/2020] [Accepted: 03/05/2020] [Indexed: 02/07/2023]
Abstract
Long considered both physiologic and inevitable, skin aging is a degenerative phenomenon whereby both intrinsic and environmental factors conspire to produce an authentic disease. The consequences of this disorder are many and varied, ranging from atrophy and fragility to defective repair to deficient immunity and vulnerability to certain infections. The pathobiologic basis for skin aging remains poorly understood. At a cellular level, stem cell dysfunction and attrition appear to be key events, and both genetic and epigenetic factors are involved in a complex interplay that over time results in deterioration of our main protective interface with the external environment. Past and current understanding of the cellular and molecular intricacies of skin aging provide a foundation for future approaches designed to thwart the aging phenotype. Herein, the authors provide a review of current insights into skin aging, including the mechanisms of skin aging, the role of stem cells in skin aging and the implications of skin aging for the microbiome and for the development of cancer. Conquest of the oft overlooked disease of skin aging should have broad implications that transcend the integument and inform novel approaches to retarding aging and age-related dysfunction in those internal organs that youthful skin was designed to envelop and safeguard.
Collapse
Affiliation(s)
- Eleanor Russell-Goldman
- Program in Dermatopathology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - George F Murphy
- Program in Dermatopathology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
15
|
PARP1 Inhibition Augments UVB-Mediated Mitochondrial Changes-Implications for UV-Induced DNA Repair and Photocarcinogenesis. Cancers (Basel) 2019; 12:cancers12010005. [PMID: 31861350 PMCID: PMC7016756 DOI: 10.3390/cancers12010005] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 12/07/2019] [Accepted: 12/10/2019] [Indexed: 01/02/2023] Open
Abstract
Keratinocytes provide the first line of defense of the human body against carcinogenic ultraviolet (UV) radiation. Acute and chronic UVB-mediated cellular responses were widely studied. However, little is known about the role of mitochondrial regulation in UVB-induced DNA damage. Here, we show that poly (ADP-ribose) polymerase 1 (PARP1) and ataxia-telangiectasia-mutated (ATM) kinase, two tumor suppressors, are important regulators in mitochondrial alterations induced by UVB. Our study demonstrates that PARP inhibition by ABT-888 upon UVB treatment exacerbated cyclobutane pyrimidine dimers (CPD) accumulation, cell cycle block and cell death and reduced cell proliferation in premalignant skin keratinocytes. Furthermore, in human keratinocytes UVB enhanced oxidative phosphorylation (OXPHOS) and autophagy which were further induced upon PARP inhibition. Immunoblot analysis showed that these cellular responses to PARP inhibition upon UVB irradiation strongly alter the phosphorylation level of ATM, adenosine monophosphate-activated kinase (AMPK), p53, protein kinase B (AKT), and mammalian target of rapamycin (mTOR) proteins. Furthermore, chemical inhibition of ATM led to significant reduction in AMPK, p53, AKT, and mTOR activation suggesting the central role of ATM in the UVB-mediated mitochondrial changes. Our results suggest a possible link between UVB-induced DNA damage and metabolic adaptations of mitochondria and reveal the OXPHOS-regulating role of autophagy which is dependent on key metabolic and DNA damage regulators downstream of PARP1 and ATM.
Collapse
|
16
|
Signaling Pathways, Chemical and Biological Modulators of Nucleotide Excision Repair: The Faithful Shield against UV Genotoxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4654206. [PMID: 31485292 PMCID: PMC6702832 DOI: 10.1155/2019/4654206] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/10/2019] [Indexed: 12/28/2022]
Abstract
The continuous exposure of the human body's cells to radiation and genotoxic stresses leads to the accumulation of DNA lesions. Fortunately, our body has several effective repair mechanisms, among which is nucleotide excision repair (NER), to counteract these lesions. NER includes both global genome repair (GG-NER) and transcription-coupled repair (TC-NER). Deficiencies in the NER pathway underlie the development of several DNA repair diseases, such as xeroderma pigmentosum (XP), Cockayne syndrome (CS), and trichothiodystrophy (TTD). Deficiencies in GG-NER and TC-NER render individuals to become prone to cancer and neurological disorders, respectively. Therefore, NER regulation is of interest in fine-tuning these risks. Distinct signaling cascades including the NFE2L2 (NRF2), AHR, PI3K/AKT1, MAPK, and CSNK2A1 pathways can modulate NER function. In addition, several chemical and biological compounds have proven success in regulating NER's activity. These modulators, particularly the positive ones, could therefore provide potential treatments for genetic DNA repair-based diseases. Negative modulators, nonetheless, can help sensitize cells to killing by genotoxic chemicals. In this review, we will summarize and discuss the major upstream signaling pathways and molecules that could modulate the NER's activity.
Collapse
|
17
|
Lago JC, Puzzi MB. The effect of aging in primary human dermal fibroblasts. PLoS One 2019; 14:e0219165. [PMID: 31269075 PMCID: PMC6608952 DOI: 10.1371/journal.pone.0219165] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 06/17/2019] [Indexed: 12/04/2022] Open
Abstract
Skin aging is a complex process, and alterations in human skin due to aging have distinct characteristic as compared to other organs. The aging of dermal cells and the biological mechanisms involved in this process are key areas to understand skin aging. A large number of biological mechanisms, such as decreasing of protein synthesis of extracellular matrix or increasing of degradation, are known to be altered through skin aging. However, environmental influence can accelerate this characteristic phenotype. In this study, we analyzed primary human dermal fibroblasts in three different in-vitro aging models—UVB irradiation and accelerated proliferation of human dermal fibroblasts from young donors as well as from elderly donors—for the gene expression of COL1A1, COL1A2, COL3A1, COL4A1, COL7A1, MMP1, MMP2, MMP3, MMP7, MMP8, MMP9, MMP10, MMP12, MMP13, MMP14, TIMP1, TIMP2, TIMP3, TIMP4, IL1B, IL1A, IL6, IL8, IL10, PTGS2, TP53, CASP3, LMNA, SIRT1. We compared the gene expression levels with young control. Furthermore, the behavior of skin fibroblasts was also evaluated using cell growth rate. The findings reveal that the gene expression levels in skin fibroblasts was altered in the process of aging in all three in-vitro aging models, and the cell growth rate was reduced, suggesting that these methods can be employed to understand skin aging mechanisms as well as drug discovery screening method.
Collapse
Affiliation(s)
- Juliana Carvalhães Lago
- Department of Dermatology, School of Medical Sciences, Laboratory of Skin Cell Cultures-Pediatric Research Center, University of Campinas – UNICAMP, Campinas, São Paulo, Brazil
- * E-mail:
| | - Maria Beatriz Puzzi
- Department of Dermatology, School of Medical Sciences, Laboratory of Skin Cell Cultures-Pediatric Research Center, University of Campinas – UNICAMP, Campinas, São Paulo, Brazil
| |
Collapse
|
18
|
Zhao B, Li X, Zhou L, Wang Y, Shang P. SIRT1: a potential tumour biomarker and therapeutic target. J Drug Target 2019; 27:1046-1052. [PMID: 31056963 DOI: 10.1080/1061186x.2019.1605519] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
SIRT1, which is highly homologous to yeast silent information regulator 2, has recently garnered tremendous attention because of its various regulatory effects in several pathological conditions. Numerous studies have found that SIRT1 is highly expressed in a broad range of tumours compared with the paracancerous tissue. However, the role of SIRT1 in malignancies has yet to be systematically elucidated, and its use as a promising biomarker or therapeutic target for tumours has not been well-reported. Herein, we focus on the roles of SIRT1 in cancers and summarise the potential use of SIRT1 as a promising tumour biomarker or therapeutic target.
Collapse
Affiliation(s)
- Bin Zhao
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen , Shenzhen , China.,School of Life Science, Northwestern Polytechnical University , Xi'an , China.,Key Laboratory for Space Bioscience and Biotechnology, School of Life Science, Institute of Special Environmental Biophysics, Northwestern Polytechnical University , Xi'an , China
| | - Xin Li
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen , Shenzhen , China.,School of Life Science, Northwestern Polytechnical University , Xi'an , China.,Key Laboratory for Space Bioscience and Biotechnology, School of Life Science, Institute of Special Environmental Biophysics, Northwestern Polytechnical University , Xi'an , China
| | - Liangfu Zhou
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen , Shenzhen , China.,School of Life Science, Northwestern Polytechnical University , Xi'an , China.,Key Laboratory for Space Bioscience and Biotechnology, School of Life Science, Institute of Special Environmental Biophysics, Northwestern Polytechnical University , Xi'an , China
| | - Ye Wang
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen , Shenzhen , China.,School of Life Science, Northwestern Polytechnical University , Xi'an , China.,Key Laboratory for Space Bioscience and Biotechnology, School of Life Science, Institute of Special Environmental Biophysics, Northwestern Polytechnical University , Xi'an , China
| | - Peng Shang
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen , Shenzhen , China.,Key Laboratory for Space Bioscience and Biotechnology, School of Life Science, Institute of Special Environmental Biophysics, Northwestern Polytechnical University , Xi'an , China
| |
Collapse
|
19
|
Anti-Wrinkling and Anti-Melanogenic Effect of Pradosia mutisii Methanol Extract. Int J Mol Sci 2019; 20:ijms20051043. [PMID: 30818884 PMCID: PMC6429126 DOI: 10.3390/ijms20051043] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 02/21/2019] [Accepted: 02/23/2019] [Indexed: 02/06/2023] Open
Abstract
Ultraviolet (UV) exposure causes skin photoaging leading to skin wrinkling and sagging via production of reactive oxygen species (ROS). For this reason, protection from photoaging is an important feature in cosmeceutical and dermatological products. Natural product-derived biomaterials are highly desired as future possible ingredients, because these biomaterials are often safe and effective. In this study, we aimed to characterize the skin protective activity of Pradosia mutisii, traditionally used to treat sunburn and erythema. We determined the free radical scavenging, anti-melanogenic, and moisturizing effects of a methanol extract of Pradosia mutisii (Pm-ME) in keratinocytes (HaCaT cells), melanocytes (B16F10 cells), and fibroblasts (human dermal fibroblasts (HDFs)) at non-cytotoxic concentrations. Pradosia mutisii methanol extract contains coumaric acid as a major component, and the extract exhibited protective activity against UVB- and H2O2-induced cytotoxicity. This extract also suppressed the expression of metalloproteinases (MMPs) and cyclooxygenase (COX)-2 in HaCaT cells. A reduction of Sirt-1 expression under UVB- and H2O2-treated conditions was recovered in HaCaT cells by Pm-ME. This extract displayed significant free radical scavenging activity according to the 2,2′-azino-bis (3-ethylbenzothiazoline-6-sulphonic acid) diammonium salt (ABTS) assay. The Pm-ME also upregulated the expression levels of hyaluronic acid synthase (HAS) and transglutaminase-1 (TGM-1) in HaCaT cells, indicating a putative moisturizing activity. Interestingly, the expression of collagen type 1 (Col1A1) gene and its promoter activity, as assessed by a reporter gene assay, were found to be increased in HDF and HEK293 cells. Similarly, Pm-ME helped recover collagen levels after UVB and H2O2 treatment in HDFs as well as decreased the synthesis and secretion of melanin from B16F10 melanoma cells, which may indicate a beneficial whitening cosmetic value. The p38 inhibitor SB203580 and the JNK inhibitor SP600125 suppressed MMP-9 and COX-2 expression in H2O2-treated HaCaT cells. Similarly, the ERK inhibitor U0126 inhibited HAS-2 in Pm-ME/H2O2-treated HaCaT cells. These findings suggested that inhibition of JNK and p38 and activation of ERK could be targeted by Pm-ME. Therefore, Pm-ME may exert anti-photoaging and anti-melanogenic properties via the regulation of mitogen-activated protein kinase, which could be beneficial in the cosmeceutical industry.
Collapse
|
20
|
Lu Y, Tao F, Zhou MT, Tang KF. The signaling pathways that mediate the anti-cancer effects of caloric restriction. Pharmacol Res 2019; 141:512-520. [PMID: 30641278 DOI: 10.1016/j.phrs.2019.01.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 12/31/2018] [Accepted: 01/11/2019] [Indexed: 02/07/2023]
Abstract
Caloric restriction (CR) has been shown to promote longevity and ameliorate aging-associated diseases, including cancer. Extensive research over recent decades has revealed that CR reduces IGF-1/PI3K/AKT signaling and increases sirtuin signaling. We recently found that CR also enhances ALDOA/DNA-PK/p53 signaling. In the present review, we summarize the molecular mechanisms underlying the modulation of the IGF-1/PI3K/AKT pathway, sirtuin signaling, and the ALDOA/DNA-PK/p53 pathway by CR. We also summarize the evidence concerning the roles of these signaling pathways in carcinogenesis, and discuss how they are regulated by CR. Finally, we discuss the crosstalk between these signaling pathways.
Collapse
Affiliation(s)
- Yiyi Lu
- Department of Dermato-Venereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China
| | - Fengxing Tao
- Department of Dermato-Venereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, China
| | - Meng-Tao Zhou
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, Zhejiang, China.
| | - Kai-Fu Tang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, Zhejiang, China; Digestive Cancer Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325015, Zhejiang, China.
| |
Collapse
|
21
|
Epigenetic Regulation of Skin Cells in Natural Aging and Premature Aging Diseases. Cells 2018; 7:cells7120268. [PMID: 30545089 PMCID: PMC6315602 DOI: 10.3390/cells7120268] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 12/07/2018] [Accepted: 12/11/2018] [Indexed: 02/06/2023] Open
Abstract
Skin undergoes continuous renewal throughout an individual’s lifetime relying on stem cell functionality. However, a decline of the skin regenerative potential occurs with age. The accumulation of senescent cells over time probably reduces tissue regeneration and contributes to skin aging. Keratinocytes and dermal fibroblasts undergo senescence in response to several intrinsic or extrinsic stresses, including telomere shortening, overproduction of reactive oxygen species, diet, and sunlight exposure. Epigenetic mechanisms directly regulate skin homeostasis and regeneration, but they also mark cell senescence and the natural and pathological aging processes. Progeroid syndromes represent a group of clinical and genetically heterogeneous pathologies characterized by the accelerated aging of various tissues and organs, including skin. Skin cells from progeroid patients display molecular hallmarks that mimic those associated with naturally occurring aging. Thus, investigations on progeroid syndromes strongly contribute to disclose the causal mechanisms that underlie the aging process. In the present review, we discuss the role of epigenetic pathways in skin cell regulation during physiologic and premature aging.
Collapse
|
22
|
Jarrett SG, Carter KM, Bautista RM, He D, Wang C, D'Orazio JA. Sirtuin 1-mediated deacetylation of XPA DNA repair protein enhances its interaction with ATR protein and promotes cAMP-induced DNA repair of UV damage. J Biol Chem 2018; 293:19025-19037. [PMID: 30327428 DOI: 10.1074/jbc.ra118.003940] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 10/12/2018] [Indexed: 12/31/2022] Open
Abstract
Blunted melanocortin 1 receptor (MC1R) signaling promotes melanocyte genomic instability in part by attenuating cAMP-mediated DNA repair responses, particularly nucleotide excision repair (NER), which recognizes and clears mutagenic photodamage. cAMP-enhanced NER is mediated by interactions between the ataxia telangiectasia-mutated and Rad3-related (ATR) and xeroderma pigmentosum complementation group A (XPA) proteins. We now report a critical role for sirtuin 1 (SIRT1) in regulating ATR-mediated phosphorylation of XPA. SIRT1 deacetylates XPA at residues Lys-63, Lys-67, and Lys-215 to promote interactions with ATR. Mutant XPA containing acetylation mimetics at residues Lys-63, Lys-67, and Lys-215 exhibit blunted UV-dependent ATR-XPA interactions even in the presence of cAMP signals. ATR-mediated phosphorylation of XPA on Ser-196 enhances cAMP-mediated optimization of NER and is promoted by SIRT1-mediated deacetylation of XPA on Lys-63, Lys-67, and Lys-215. Interference with ATR-mediated XPA phosphorylation at Ser-196 by persistent acetylation of XPA at Lys-63, Lys-67, and Lys-215 delays repair of UV-induced DNA damage and attenuates cAMP-enhanced NER. Our study identifies a regulatory ATR-SIRT1-XPA axis in cAMP-mediated regulation melanocyte genomic stability, involving SIRT1-mediated deacetylation (Lys-63, Lys-67, and Lys-215) and ATR-dependent phosphorylation (Ser-196) post-translational modifications of the core NER factor XPA.
Collapse
Affiliation(s)
- Stuart G Jarrett
- From the Markey Cancer Center and .,the Departments of Toxicology and Cancer Biology
| | | | | | - Daheng He
- From the Markey Cancer Center and.,Biostatistics and Bioinformatics, and
| | - Chi Wang
- From the Markey Cancer Center and.,Biostatistics and Bioinformatics, and
| | - John A D'Orazio
- From the Markey Cancer Center and .,the Departments of Toxicology and Cancer Biology.,Pediatrics, University of Kentucky College of Medicine, Lexington, Kentucky 40536
| |
Collapse
|
23
|
Hong YH, Kim D, Nam G, Yoo S, Han SY, Jeong SG, Kim E, Jeong D, Yoon K, Kim S, Park J, Cho JY. Photoaging protective effects of BIOGF1K, a compound-K-rich fraction prepared from Panax ginseng. J Ginseng Res 2018; 42:81-89. [PMID: 29348726 PMCID: PMC5766695 DOI: 10.1016/j.jgr.2017.01.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 12/25/2016] [Accepted: 01/02/2017] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND BIOGF1K, a compound-K-rich fraction, has been shown to display anti-inflammatory activity. Although Panax ginseng is widely used for the prevention of photoaging events induced by UVB irradiation, the effect of BIOGF1K on photoaging has not yet been examined. In this study, we investigated the effects of BIOGF1K on UVB-induced photoaging events. METHODS We analyzed the ability of BIOGF1K to prevent UVB-induced apoptosis, enhance matrix metalloproteinase (MMP) expression, upregulate anti-inflammatory activity, reduce sirtuin 1 expression, and melanin production using reverse transcription-polymerase chain reaction, melanin content assay, tyrosinase assay, and flow cytometry. We also evaluated the effects of BIOGF1K on the activator protein-1 signaling pathway, which plays an important role in photoaging, by immunoblot analysis and luciferase reporter gene assays. RESULTS Treatment of UVB-irradiated NIH3T3 fibroblasts with BIOGF1K prevented UVB-induced cell death, inhibited apoptosis, suppressed morphological changes, reduced melanin secretion, restored the levels of type I procollagen and sirtuin 1, and prevented mRNA upregulation of MMP-1, MMP-2, and cyclo-oxygenase-2; these effects all occurred in a dose-dependent manner. In addition, BIOGF1K markedly reduced activator-protein-1-mediated luciferase activity and decreased the activity of mitogen-activated protein kinases (extracellular response kinase, p38, and C-Jun N-terminal kinase). CONCLUSION Our results strongly suggest that BIOGF1K has anti-photoaging activity and that BIOGF1K could be used in anti-aging cosmeceutical preparations.
Collapse
Affiliation(s)
- Yo Han Hong
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Donghyun Kim
- Amorepacific Research and Development Unit, Yongin, Republic of Korea
| | - Gibaeg Nam
- Amorepacific Research and Development Unit, Yongin, Republic of Korea
| | - Sulgi Yoo
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Sang Yun Han
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Seong-Gu Jeong
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Eunji Kim
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Deok Jeong
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Keejung Yoon
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| | - Sunggyu Kim
- Research and Business Foundation, Sungkyunkwan University, Suwon, Republic of Korea
| | - Junseong Park
- Amorepacific Research and Development Unit, Yongin, Republic of Korea
| | - Jae Youl Cho
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
24
|
Shah P, Qiang L, Yang S, Soltani K, He YY. Regulation of XPC deubiquitination by USP11 in repair of UV-induced DNA damage. Oncotarget 2017; 8:96522-96535. [PMID: 29228550 PMCID: PMC5722502 DOI: 10.18632/oncotarget.22105] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 10/13/2017] [Indexed: 12/31/2022] Open
Abstract
Nucleotide excision repair (NER) is the most versatile DNA repair pathway for removing DNA damage caused by UV radiation and many environmental carcinogens. NER is essential for suppressing tumorigenesis in the skin, lungs and brain. Although the core NER proteins have been identified and characterized, molecular regulation of NER remains poorly understood. Here we show that ubiquitin-specific peptidase 11 (USP11) positively regulates NER by deubiquitinating xeroderma pigmentosum complementation group C (XPC) and promoting its retention at the DNA damage sites. In addition, UV irradiation induces both USP11 recruitment to the chromatin and USP11 interaction with XPC in an XPC-ubiquitination-dependent manner. Furthermore, we found that USP11 is down-regulated in chronically UV-exposed mouse skin and in skin tumors from mice and humans. Our findings indicate that USP11 plays an important role in maintaining NER capacity, and suggest that USP11 acts as a tumor suppressor via its role in DNA repair.
Collapse
Affiliation(s)
- Palak Shah
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, USA
- Committee on Molecular Pathogenesis and Molecular Medicine, University of Chicago, Chicago, IL, USA
| | - Lei Qiang
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, USA
| | - Seungwon Yang
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, USA
| | - Keyoumars Soltani
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, USA
| | - Yu-Ying He
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, USA
- Committee on Molecular Pathogenesis and Molecular Medicine, University of Chicago, Chicago, IL, USA
| |
Collapse
|
25
|
Smith KJ, Boyer JA, Muku GE, Murray IA, Gowda K, Desai D, Amin SG, Glick AB, Perdew GH. Editor's Highlight: Ah Receptor Activation Potentiates Neutrophil Chemoattractant (C-X-C Motif) Ligand 5 Expression in Keratinocytes and Skin. Toxicol Sci 2017; 160:83-94. [PMID: 28973351 PMCID: PMC5837612 DOI: 10.1093/toxsci/kfx160] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Chemokines are components of the skin microenvironment, which enable immune cell chemotaxis. Traditionally, transcription factors involved in inflammatory signaling (eg, NFκB) are important mediators of chemokine expression. To what extent xenobiotics and their associated receptors control chemokine expression is poorly understood. The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor known to mediate physiological responses in the skin through the regulation of genes involved in xenobiotic metabolism, epidermal differentiation, and immunity. Here, we demonstrate that AHR activation within primary mouse keratinocytes regulates the expression of a neutrophil directing chemokine (C-X-C motif) ligand 5 (Cxcl5). AHR-mediated regulation of Cxcl5 is because of direct transcriptional activity upon treatment with AHR agonists such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Additionally, AHR mediates enhanced induction of Cxcl5 upon exposure to an agonist and the inflammatory cytokine interleukin 1 beta. This synergy is confined primarily to keratinocytes, as dermal fibroblasts did not achieve the same level of combinatorial induction. AHR-specific antagonists were able to reduce basal and induced levels of Cxcl5, demonstrating the potential for pharmacological intervention. Exposure of C57BL/6 J mice to ultraviolet (UV) light followed by topical treatment with the AHR agonist formylindolo(3,2-b)carbazole (FICZ) significantly induced Cxcl5 expression in skin compared with UV alone, and this response was absent in Ahr-/- mice. These results establish AHR as an important mediator of Cxcl5, with implications for the treatment of inflammatory skin diseases.
Collapse
Affiliation(s)
- Kayla J. Smith
- The Graduate Program in Biochemistry, Microbiology, and Molecular Biology, Department of Biochemistry and Molecular Biology
| | - Jacob A. Boyer
- Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, Pennsylvania 16802
| | - Gulsum E. Muku
- Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, Pennsylvania 16802
| | - Iain A. Murray
- Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, Pennsylvania 16802
| | - Krishne Gowda
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - Dhimant Desai
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - Shantu G. Amin
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - Adam B. Glick
- Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, Pennsylvania 16802
| | - Gary H. Perdew
- Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, Pennsylvania 16802
| |
Collapse
|
26
|
Qiang L, Sample A, Liu H, Wu X, He YY. Epidermal SIRT1 regulates inflammation, cell migration, and wound healing. Sci Rep 2017; 7:14110. [PMID: 29074993 PMCID: PMC5658409 DOI: 10.1038/s41598-017-14371-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 10/09/2017] [Indexed: 01/12/2023] Open
Abstract
Sirtuins (SIRT1-7) are NAD-dependent proteins with the enzymatic activity of deacetylases and ADP ribosyltransferases. SIRT1 is the proto member of the proteins in the mammalian sirtuin family and plays multiple roles in aging and disease. Using mice with epidermis-specific SIRT1 deletion, we show that SIRT1 is required for efficient wound healing. SIRT1 deficiency in the epidermis inhibited the regeneration of both the epidermis and the dermal stroma. SIRT1 loss altered the production of many cytokines, inhibited the recruitment of macrophages, neutrophils, and mast cells, the recruitment and activation of fibroblasts, and angiogenesis in the granulation tissue. In keratinocytes, SIRT1 knockdown inhibited EMT, cell migration, and TGF-β signaling. For the first time, using skin-specific mouse model, we demonstrate that epidermal SIRT1 plays a crucial role in wound repair. These findings are novel in understanding how wound healing is regulated. Our findings provide in vivo and in vitro evidence that SIRT1 in the epidermis regulates cell migration, redox response, inflammation, epidermis re-epithelialization, granulation formation, and proper wound healing in mice.
Collapse
Affiliation(s)
- Lei Qiang
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, 60637, USA. .,State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| | - Ashley Sample
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, 60637, USA.,Committee on Cancer Biology, University of Chicago, Chicago, IL, 60637, USA
| | - Han Liu
- Ben May Department of Cancer Research, University of Chicago, Chicago, IL, 60637, USA
| | - Xiaoyang Wu
- Committee on Cancer Biology, University of Chicago, Chicago, IL, 60637, USA.,Ben May Department of Cancer Research, University of Chicago, Chicago, IL, 60637, USA
| | - Yu-Ying He
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, 60637, USA. .,Committee on Cancer Biology, University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
27
|
Garcia-Peterson LM, Wilking-Busch MJ, Ndiaye MA, Philippe CGA, Setaluri V, Ahmad N. Sirtuins in Skin and Skin Cancers. Skin Pharmacol Physiol 2017; 30:216-224. [PMID: 28704830 PMCID: PMC5568696 DOI: 10.1159/000477417] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 05/09/2017] [Indexed: 12/21/2022]
Abstract
The sirtuins are a family of proteins that comprise class III of the histone deacetylases. These NAD+-dependent proteins have been found to be intricately involved in a variety of important and skin-relevant cellular functions and processes, including aging, UV damage response, oxidative stress, and wound repair. In addition, recent research is unraveling the role of sirtuins in a variety of skin diseases, including melanoma and nonmelanoma skin cancers. In this review, we provide a discussion on the potential roles and implications of different sirtuins in skin-specific cellular processes, which may have relevance to skin health and skin diseases. Based on the available literature, the sirtuins appear to be important targets in the management of a variety of skin diseases from cosmetic (e.g., skin aging) to fatal conditions (e.g., melanoma).
Collapse
Affiliation(s)
| | | | - Mary Ann Ndiaye
- Department of Dermatology, University of Wisconsin, Madison, Wisconsin, USA
| | | | - Vijayasaradhi Setaluri
- Department of Dermatology, University of Wisconsin, Madison, Wisconsin, USA
- William S. Middleton VA Medical Center, Madison, Wisconsin, USA
| | - Nihal Ahmad
- Department of Dermatology, University of Wisconsin, Madison, Wisconsin, USA
- William S. Middleton VA Medical Center, Madison, Wisconsin, USA
| |
Collapse
|
28
|
Nikolova T, Kiweler N, Krämer OH. Interstrand Crosslink Repair as a Target for HDAC Inhibition. Trends Pharmacol Sci 2017; 38:822-836. [PMID: 28687272 DOI: 10.1016/j.tips.2017.05.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 05/29/2017] [Accepted: 05/31/2017] [Indexed: 12/29/2022]
Abstract
DNA interstrand crosslinks (ICLs) covalently connect complementary DNA strands. Consequently, DNA replication and transcription are hampered, DNA damage responses (DDR) are initiated, and cell death is triggered. Therefore, drugs inducing ICLs are effective against rapidly growing cancer cells. However, tumors engage a complicated enzymatic machinery to repair and survive ICLs. Several factors, including the post-translational acetylation/deacetylation of lysine residues within proteins, control this network. Histone deacetylases (HDACs) modulate the expression and functions of DNA repair proteins which remove ICLs and control the accessibility of chromatin. Accordingly, histone deacetylase inhibitors (HDACi) are small, pharmacologically and clinically relevant molecules that sensitize cancer cells to ICL inducers. We discuss the mechanism of ICL repair and targets of HDACi within this pathway.
Collapse
Affiliation(s)
- Teodora Nikolova
- Institute of Toxicology, University Medical Center, Obere Zahlbacher Strasse 67, 55131 Mainz, Germany.
| | - Nicole Kiweler
- Institute of Toxicology, University Medical Center, Obere Zahlbacher Strasse 67, 55131 Mainz, Germany
| | - Oliver H Krämer
- Institute of Toxicology, University Medical Center, Obere Zahlbacher Strasse 67, 55131 Mainz, Germany.
| |
Collapse
|
29
|
Ren NSX, Ji M, Tokar EJ, Busch EL, Xu X, Lewis D, Li X, Jin A, Zhang Y, Wu WKK, Huang W, Li L, Fargo DC, Keku TO, Sandler RS, Li X. Haploinsufficiency of SIRT1 Enhances Glutamine Metabolism and Promotes Cancer Development. Curr Biol 2017; 27:483-494. [PMID: 28162896 DOI: 10.1016/j.cub.2016.12.047] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 10/31/2016] [Accepted: 12/20/2016] [Indexed: 01/05/2023]
Abstract
SIRT1, the most conserved mammalian NAD+-dependent protein deacetylase, plays a vital role in the regulation of metabolism, stress responses, and genome stability. However, the role of SIRT1 in the multi-step process leading to transformation and/or tumorigenesis, as either a tumor suppressor or tumor promoter, is complex and may be dependent upon the context in which SIRT1 activity is altered, and the role of SIRT1 in tumor metabolism is unknown. Here, we demonstrate that SIRT1 dose-dependently regulates cellular glutamine metabolism and apoptosis, which in turn differentially impact cell proliferation and cancer development. Heterozygous deletion of Sirt1 induces c-Myc expression, enhancing glutamine metabolism and subsequent proliferation, autophagy, stress resistance, and cancer formation. In contrast, homozygous deletion of Sirt1 triggers cellular apoptotic pathways, increases cell death, diminishes autophagy, and reduces cancer formation. Consistent with the observed dose dependence in cells, intestine-specific Sirt1 heterozygous mice have enhanced intestinal tumor formation, whereas intestine-specific Sirt1 homozygous knockout mice have reduced development of colon cancer. Furthermore, SIRT1 reduction, but not deletion, is associated with human colorectal tumors, and colorectal cancer patients with low protein expression of SIRT1 have a poor prognosis. Taken together, our findings indicate that the dose-dependent regulation of tumor metabolism and possibly apoptosis by SIRT1 mechanistically contribute to the observed dual roles of SIRT1 in tumorigenesis. Our study highlights the importance of maintenance of a suitable SIRT1 dosage for metabolic and tissue homeostasis, which will have important implications in SIRT1-small-molecule-activator/inhibitor-based therapeutic strategies for cancers.
Collapse
Affiliation(s)
- Natalie S X Ren
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Ming Ji
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Erik J Tokar
- Stem Cell Toxicology Group, National Toxicology Program, Research Triangle Park, NC 27709, USA
| | - Evan L Busch
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Xiaojiang Xu
- Integrative Bioinformatics Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - DeAsia Lewis
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Xiangchun Li
- Department of Anaesthesia and Intensive Care, Institute of Digestive Disease and State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
| | - Aiwen Jin
- Department of Radiation Oncology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yanping Zhang
- Department of Radiation Oncology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - William K K Wu
- Department of Anaesthesia and Intensive Care, Institute of Digestive Disease and State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
| | - Weichun Huang
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Leping Li
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - David C Fargo
- Integrative Bioinformatics Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Temitope O Keku
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Robert S Sandler
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Xiaoling Li
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
30
|
Qiang L, Sample A, Shea CR, Soltani K, Macleod KF, He YY. Autophagy gene ATG7 regulates ultraviolet radiation-induced inflammation and skin tumorigenesis. Autophagy 2017; 13:2086-2103. [PMID: 28933598 PMCID: PMC5788558 DOI: 10.1080/15548627.2017.1380757] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 08/17/2017] [Accepted: 09/13/2017] [Indexed: 12/26/2022] Open
Abstract
Macroautophagy (hereafter autophagy) is a cellular "self-eating" process that is implicated in many human cancers, where it can act to either promote or suppress tumorigenesis. However, the role of autophagy in regulation of inflammation during tumorigenesis remains unclear. Here we show that autophagy is induced in the epidermis by ultraviolet (UV) irradiation and autophagy gene Atg7 promoted UV-induced inflammation and skin tumorigenesis. Atg7 regulated UV-induced cytokine expression and secretion, and promoted Ptgs2/Cox-2 expression through both a CREB1/CREB-dependent cell autonomous mechanism and an IL1B/IL1β-dependent non-cell autonomous mechanism. Adding PGE2 increased UV-induced skin inflammation and tumorigenesis, reversing the epidermal phenotype in mice with Atg7 deletion in keratinocytes. Similar to ATG7 knockdown in human keratinocytes, ATG5 knockdown inhibited UVB-induced expression of PTGS2 and cytokines. Furthermore, ATG7 loss increased the activation of the AMPK pathway and the phosphorylation of CRTC1, and led to endoplasmic reticulum (ER) accumulation and reduction of ER stress. Inducing ER stress and inhibiting calcium influx into the ER by thapsigargin reverses the inflammation and tumorigenesis phenotype in mice with epidermal Atg7 deletion. Taken together, these findings demonstrate that deleting autophagy gene Atg7 leads to a suppression of carcinogen-induced protumorigenic inflammatory microenvironment and tumorigenesis of the epithelium.
Collapse
Affiliation(s)
- Lei Qiang
- Section of Dermatology, Department of Medicine, University of Chicago, Chicago, Illinois, USA
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ashley Sample
- Section of Dermatology, Department of Medicine, University of Chicago, Chicago, Illinois, USA
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois, USA
| | - Christopher R. Shea
- Section of Dermatology, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Keyoumars Soltani
- Section of Dermatology, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Kay F. Macleod
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois, USA
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois, USA
| | - Yu-Ying He
- Section of Dermatology, Department of Medicine, University of Chicago, Chicago, Illinois, USA
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
31
|
Wu X, Cao N, Fenech M, Wang X. Role of Sirtuins in Maintenance of Genomic Stability: Relevance to Cancer and Healthy Aging. DNA Cell Biol 2016; 35:542-575. [DOI: 10.1089/dna.2016.3280] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Affiliation(s)
- Xiayu Wu
- School of Life Sciences, The Engineering Research Center of Sustainable Development and Utilization of Biomass Energy, Ministry of Education, Yunnan Normal University, Kunming, Yunnan, China
| | - Neng Cao
- School of Life Sciences, The Engineering Research Center of Sustainable Development and Utilization of Biomass Energy, Ministry of Education, Yunnan Normal University, Kunming, Yunnan, China
| | - Michael Fenech
- Genome Health and Personalized Nutrition, Commonwealth Scientific and Industrial Research Organization Food and Nutrition, Adelaide, South Australia, Australia
| | - Xu Wang
- School of Life Sciences, The Engineering Research Center of Sustainable Development and Utilization of Biomass Energy, Ministry of Education, Yunnan Normal University, Kunming, Yunnan, China
| |
Collapse
|
32
|
Liu L, Liu C, Zhang Q, Shen J, Zhang H, Shan J, Duan G, Guo D, Chen X, Cheng J, Xu Y, Yang Z, Yao C, Lai M, Qian C. SIRT1-mediated transcriptional regulation of SOX2 is important for self-renewal of liver cancer stem cells. Hepatology 2016; 64:814-27. [PMID: 27312708 DOI: 10.1002/hep.28690] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 05/18/2016] [Accepted: 05/27/2016] [Indexed: 12/21/2022]
Abstract
UNLABELLED Hepatocellular carcinoma (HCC) is a highly aggressive liver tumor containing cancer stem cells (CSCs), which participate in tumor invasion, therapeutic resistance, and tumor relapse leading to poor outcome and limited therapeutic options. Histone deacetylatase sirtuin 1 (SIRT1) has been shown to be up-regulated in human cancers; however, its role in liver CSCs is unknown. In this study, we explored the biological functions of SIRT1 in liver CSCs. Our data show that SIRT1 is highly expressed in liver CSCs and decreases during differentiation. In addition, high levels of SIRT1 predict a decreased probability of survival in patients with HCC. SIRT1 is responsible for the maintenance of self-renewal and tumorigenicity of liver CSCs, and overexpression of exogenous SIRT1 can restore self-renewal of non-CSCs. We demonstrated that SOX2 is a main downstream regulator of SIRT1-mediated self-renewal and tumorigenicity potential of liver CSCs. Mechanistically, SIRT1 regulates transcription of the SOX2 gene by way of chromatin-based epigenetic changes, which are dependent on DNA methylation. This effect is achieved by alternation of histone modification and interaction with DNA methyltransferase 3A, resulting in hypermethylation of SOX2 promoter. Furthermore, we demonstrated that insulin growth factor signaling plays an important role in maintaining SIRT1 expression through increased SIRT1 protein stability. CONCLUSIONS These findings highlight the importance of SIRT1 in the biology of liver CSCs and suggest that SIRT1 may serve as a molecular target for HCC therapy. (Hepatology 2016;64:814-827).
Collapse
Affiliation(s)
- Limei Liu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Chungang Liu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China.,Department of Pathology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qianzhen Zhang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Junjie Shen
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Heng Zhang
- Institute of Urology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Juanjuan Shan
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Guangjie Duan
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Deyu Guo
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Xuejiao Chen
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jiamin Cheng
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Yanmin Xu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Zhi Yang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Chao Yao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Maode Lai
- Department of Pathology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Cheng Qian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
33
|
Zhao B, Shah P, Qiang L, He TC, Budanov A, He YY. Distinct Role of Sesn2 in Response to UVB-Induced DNA Damage and UVA-Induced Oxidative Stress in Melanocytes. Photochem Photobiol 2016; 93:375-381. [PMID: 27463837 DOI: 10.1111/php.12624] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 07/12/2016] [Indexed: 02/06/2023]
Abstract
Ultraviolet (UV) radiation, including both UVB and UVA irradiation, is the major risk factor for causing skin cancer including melanoma. Recently, we have shown that Sesn2, a member of the evolutionarily conserved stress-inducible protein family Sestrins (Sesn), is upregulated in human melanomas as compared to melanocytes in normal human skin, suggesting an oncogenic role of Sesn2. However, the role of Sesn2 in UVB and UVA response is unknown. Here, we demonstrated that both UVB and UVA induce Sesn2 upregulation in melanocytes and melanoma cells. UVB induces Sesn2 expression through the p53 and AKT3 pathways. Sesn2 negatively regulates UVB-induced DNA damage repair. In comparison, UVA induces Sesn2 upregulation through mitochondria but not Nrf2. Sesn2 ablation increased UVA-induced Nrf2 induction and inhibits UVA-induced ROS production, indicating that Sesn2 acts as an upstream regulator of Nrf2. These findings suggest previously unrecognized mechanisms in melanocyte response to UVB and UVA irradiation and potentially in melanoma formation.
Collapse
Affiliation(s)
- Baozhong Zhao
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL
| | - Palak Shah
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL
| | - Lei Qiang
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL
| | - Tong-Chuan He
- Department of Orthopaedic Surgery & Rehabilitation Medicine, University of Chicago, Chicago, IL
| | - Andrey Budanov
- Department of Human Molecular Genetics, Virginia Commonwealth University, Richmond, VA
| | - Yu-Ying He
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL
| |
Collapse
|
34
|
Gyöngyösi N, Lőrincz K, Keszeg A, Haluszka D, Bánvölgyi A, Tátrai E, Kárpáti S, Wikonkál NM. Photosensitivity of murine skin greatly depends on the genetic background: clinically relevant dose as a new measure to replace minimal erythema dose in mouse studies. Exp Dermatol 2016; 25:519-25. [DOI: 10.1111/exd.12984] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2016] [Indexed: 01/22/2023]
Affiliation(s)
- Nóra Gyöngyösi
- Department of Dermatology, Dermatooncology and Venerology; Semmelweis University School of Medicine; Budapest Hungary
| | - Kende Lőrincz
- Department of Dermatology, Dermatooncology and Venerology; Semmelweis University School of Medicine; Budapest Hungary
| | - András Keszeg
- Department of Dermatology, Dermatooncology and Venerology; Semmelweis University School of Medicine; Budapest Hungary
| | - Dóra Haluszka
- Department of Dermatology, Dermatooncology and Venerology; Semmelweis University School of Medicine; Budapest Hungary
| | - András Bánvölgyi
- Department of Dermatology, Dermatooncology and Venerology; Semmelweis University School of Medicine; Budapest Hungary
| | - Erika Tátrai
- Department of Ophthalmology; Semmelweis University School of Medicine; Budapest Hungary
| | - Sarolta Kárpáti
- Department of Dermatology, Dermatooncology and Venerology; Semmelweis University School of Medicine; Budapest Hungary
| | - Norbert M. Wikonkál
- Department of Dermatology, Dermatooncology and Venerology; Semmelweis University School of Medicine; Budapest Hungary
| |
Collapse
|
35
|
Zhao B, Qiang L, Joseph J, Kalyanaraman B, Viollet B, He YY. Mitochondrial dysfunction activates the AMPK signaling and autophagy to promote cell survival. Genes Dis 2016; 3:82-87. [PMID: 28066797 PMCID: PMC5215801 DOI: 10.1016/j.gendis.2015.12.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 12/21/2015] [Indexed: 12/25/2022] Open
Abstract
Autophagy is a cellular self-eating process essential for stress response and maintaining tissue homeostasis by lysosomal degradation of unwanted or damaged proteins and organelles. Here, we show that cells with defective mitochondria induce autophagy to promote cell survival through activating the AMPK pathway. Loss of mitochondrial complex III protein cytochrome b activates the AMPK signaling and induced autophagy. Inhibiting mitochondria energetics by mitochondria-targeted agents activates the AMPK signaling and induced autophagy. Genetic inhibition of AMPK inhibits autophagy induction in cells with defective mitochondria, while genetic inhibition of autophagy has no effect on AMPK activation. Mitochondria dysfunction has no effect of DNA repair of UV-induced DNA damage. However, mitochondria dysfunction sensitizes cells to apoptosis induced by UV radiation. Genetic inhibition of autophagy or AMPK sensitized cells to apoptosis in cells with defective mitochondria. Our results demonstrate that AMPK and autophagy senses mitochondria dysfunction and serves as a mechanism for survival. Our findings may provide new insights into the interplay between mitochondria function and autophagy process in maintaining tissue homeostasis, and suggest that this interaction may play important roles in diseases such as cancer and neurodegeneration.
Collapse
Affiliation(s)
- Baozhong Zhao
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, USA
| | - Lei Qiang
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, USA
| | - Joy Joseph
- Free Radical Research Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Benoit Viollet
- INSERM U567, CNRS UMR8104, Department of Endocrinology, Metabolism and Cancer, Institut Cochin, Université Paris 5, 75014 Paris, France
| | - Yu-Ying He
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL, USA
| |
Collapse
|
36
|
Wahedi HM, Lee TH, Moon EY, Kim SY. Juglone up-regulates sirt1 in skin cells under normal and UVB irradiated conditions. J Dermatol Sci 2015; 81:210-2. [PMID: 26747057 DOI: 10.1016/j.jdermsci.2015.12.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 11/10/2015] [Accepted: 12/14/2015] [Indexed: 01/04/2023]
Affiliation(s)
- Hussain Mustatab Wahedi
- College of Pharmacy, Gachon University, #191 Hambakmoero, Yeonsu-gu, Incheon 406-799, Republic of Korea
| | - Taek Hwan Lee
- College of Pharmacy, Yonsei University, #162-1 Songdo-dong, Yeonsu-gu, Incheon 406-840, Republic of Korea
| | - Eun-Yi Moon
- Department of Bioscience and Biotechnology, Sejong University, Seoul 143-747, Republic of Korea
| | - Sun Yeou Kim
- College of Pharmacy, Gachon University, #191 Hambakmoero, Yeonsu-gu, Incheon 406-799, Republic of Korea; Gachon Institute of Pharmaceutical Science, Gachon University, #191 Hambakmoe-ro, Yeonsu-gu, Incheon 406-799, Republic of Korea; Gachon Medical Research Institute, Gil Medical Center, Inchon 405-760, Republic of Korea.
| |
Collapse
|
37
|
Qiang L, Zhao B, Shah P, Sample A, Yang S, He YY. Autophagy positively regulates DNA damage recognition by nucleotide excision repair. Autophagy 2015; 12:357-68. [PMID: 26565512 DOI: 10.1080/15548627.2015.1110667] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Macroautophagy (hereafter autophagy) is a cellular catabolic process that is essential for maintaining tissue homeostasis and regulating various normal and pathologic processes in human diseases including cancer. One cancer-driving process is accumulation of genetic mutations due to impaired DNA damage repair, including nucleotide excision repair. Here we show that autophagy positively regulates nucleotide excision repair through enhancing DNA damage recognition by the DNA damage sensor proteins XPC and DDB2 via 2 pathways. First, autophagy deficiency downregulates the transcription of XPC through TWIST1-dependent activation of the transcription repressor complex E2F4-RBL2. Second, autophagy deficiency impairs the recruitment of DDB2 to ultraviolet radiation (UV)-induced DNA damage sites through TWIST1-mediated inhibition of EP300. In mice, the pharmacological autophagy inhibitor Spautin-1 promotes UVB-induced tumorigenesis, whereas the autophagy inducer rapamycin reduces UVB-induced tumorigenesis. These findings demonstrate the crucial role of autophagy in maintaining proper nucleotide excision repair in mammalian cells and suggest a previously unrecognized tumor-suppressive mechanism of autophagy in cancer.
Collapse
Affiliation(s)
- Lei Qiang
- a Department of Medicine, Section of Dermatology , University of Chicago , Chicago , IL , USA
| | - Baozhong Zhao
- a Department of Medicine, Section of Dermatology , University of Chicago , Chicago , IL , USA
| | - Palak Shah
- a Department of Medicine, Section of Dermatology , University of Chicago , Chicago , IL , USA
| | - Ashley Sample
- a Department of Medicine, Section of Dermatology , University of Chicago , Chicago , IL , USA
| | - Seungwon Yang
- a Department of Medicine, Section of Dermatology , University of Chicago , Chicago , IL , USA
| | - Yu-Ying He
- a Department of Medicine, Section of Dermatology , University of Chicago , Chicago , IL , USA
| |
Collapse
|
38
|
Talero E, García-Mauriño S, Ávila-Román J, Rodríguez-Luna A, Alcaide A, Motilva V. Bioactive Compounds Isolated from Microalgae in Chronic Inflammation and Cancer. Mar Drugs 2015; 13:6152-209. [PMID: 26437418 PMCID: PMC4626684 DOI: 10.3390/md13106152] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 09/09/2015] [Accepted: 09/15/2015] [Indexed: 12/12/2022] Open
Abstract
The risk of onset of cancer is influenced by poorly controlled chronic inflammatory processes. Inflammatory diseases related to cancer development include inflammatory bowel disease, which can lead to colon cancer, or actinic keratosis, associated with chronic exposure to ultraviolet light, which can progress to squamous cell carcinoma. Chronic inflammatory states expose these patients to a number of signals with tumorigenic effects, including nuclear factor kappa B (NF-κB) and mitogen-activated protein kinases (MAPK) activation, pro-inflammatory cytokines and prostaglandins release and ROS production. In addition, the participation of inflammasomes, autophagy and sirtuins has been demonstrated in pathological processes such as inflammation and cancer. Chemoprevention consists in the use of drugs, vitamins, or nutritional supplements to reduce the risk of developing or having a recurrence of cancer. Numerous in vitro and animal studies have established the potential colon and skin cancer chemopreventive properties of substances from marine environment, including microalgae species and their products (carotenoids, fatty acids, glycolipids, polysaccharides and proteins). This review summarizes the main mechanisms of actions of these compounds in the chemoprevention of these cancers. These actions include suppression of cell proliferation, induction of apoptosis, stimulation of antimetastatic and antiangiogenic responses and increased antioxidant and anti-inflammatory activity.
Collapse
Affiliation(s)
- Elena Talero
- Department of Pharmacology, Faculty of Pharmacy, University of Seville, Seville 41012, Spain.
| | - Sofía García-Mauriño
- Department of Plant Biology and Ecology, Faculty of Biology, University of Seville, Seville 41012, Spain.
| | - Javier Ávila-Román
- Department of Pharmacology, Faculty of Pharmacy, University of Seville, Seville 41012, Spain.
| | - Azahara Rodríguez-Luna
- Department of Pharmacology, Faculty of Pharmacy, University of Seville, Seville 41012, Spain.
| | - Antonio Alcaide
- Department of Pharmacology, Faculty of Pharmacy, University of Seville, Seville 41012, Spain.
| | - Virginia Motilva
- Department of Pharmacology, Faculty of Pharmacy, University of Seville, Seville 41012, Spain.
| |
Collapse
|
39
|
Piao MJ, Hewage SRKM, Han X, Kang KA, Kang HK, Lee NH, Hyun JW. Protective Effect of Diphlorethohydroxycarmalol against Ultraviolet B Radiation-Induced DNA Damage by Inducing the Nucleotide Excision Repair System in HaCaT Human Keratinocytes. Mar Drugs 2015; 13:5629-41. [PMID: 26404324 PMCID: PMC4584344 DOI: 10.3390/md13095629] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 08/26/2015] [Accepted: 08/26/2015] [Indexed: 12/02/2022] Open
Abstract
We investigated the protective properties of diphlorethohydroxycarmalol (DPHC), a phlorotannin, against ultraviolet B (UVB) radiation-induced cyclobutane pyrimidine dimers (CPDs) in HaCaT human keratinocytes. The nucleotide excision repair (NER) system is the pathway by which cells identify and repair bulky, helix-distorting DNA lesions such as ultraviolet (UV) radiation-induced CPDs and 6-4 photoproducts. CPDs levels were elevated in UVB-exposed cells; however, this increase was reduced by DPHC. Expression levels of xeroderma pigmentosum complementation group C (XPC) and excision repair cross-complementing 1 (ERCC1), which are essential components of the NER pathway, were induced in DPHC-treated cells. Expression of XPC and ERCC1 were reduced following UVB exposure, whereas DPHC treatment partially restored the levels of both proteins. DPHC also increased expression of transcription factor specificity protein 1 (SP1) and sirtuin 1, an up-regulator of XPC, in UVB-exposed cells. DPHC restored binding of the SP1 to the XPC promoter, which is reduced in UVB-exposed cells. These results indicate that DPHC can protect cells against UVB-induced DNA damage by inducing the NER system.
Collapse
Affiliation(s)
- Mei Jing Piao
- School of Medicine and Institute for Nuclear Science and Technology, Jeju National University, Jeju 63243, Korea.
| | | | - Xia Han
- School of Medicine and Institute for Nuclear Science and Technology, Jeju National University, Jeju 63243, Korea.
| | - Kyoung Ah Kang
- School of Medicine and Institute for Nuclear Science and Technology, Jeju National University, Jeju 63243, Korea.
| | - Hee Kyoung Kang
- School of Medicine and Institute for Nuclear Science and Technology, Jeju National University, Jeju 63243, Korea.
| | - Nam Ho Lee
- Department of Chemistry, College of Natural Sciences, Jeju National University, Jeju 63243, Korea.
| | - Jin Won Hyun
- School of Medicine and Institute for Nuclear Science and Technology, Jeju National University, Jeju 63243, Korea.
| |
Collapse
|
40
|
Ming M, Han W, Zhao B, Sundaresan NR, Deng CX, Gupta MP, He YY. SIRT6 promotes COX-2 expression and acts as an oncogene in skin cancer. Cancer Res 2014; 74:5925-33. [PMID: 25320180 DOI: 10.1158/0008-5472.can-14-1308] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
SIRT6 is a SIR2 family member that regulates multiple molecular pathways involved in metabolism, genomic stability, and aging. It has been proposed previously that SIRT6 is a tumor suppressor in cancer. Here, we challenge this concept by presenting evidence that skin-specific deletion of SIRT6 in the mouse inhibits skin tumorigenesis. SIRT6 promoted expression of COX-2 by repressing AMPK signaling, thereby increasing cell proliferation and survival in the skin epidermis. SIRT6 expression in skin keratinocytes was increased by exposure to UVB light through activation of the AKT pathway. Clinically, we found that SIRT6 was upregulated in human skin squamous cell carcinoma. Taken together, our results provide evidence that SIRT6 functions as an oncogene in the epidermis and suggest greater complexity to its role in epithelial carcinogenesis.
Collapse
Affiliation(s)
- Mei Ming
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, Illinois
| | - Weinong Han
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, Illinois
| | - Baozhong Zhao
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, Illinois
| | - Nagalingam R Sundaresan
- Department of Surgery, Committee on Cellular and Molecular Physiology, University of Chicago, Chicago, Illinois. Division of Biological Sciences, Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
| | - Chu-Xia Deng
- National Institute of Diabetes, Digestive and Kidney Diseases, US NIH, Bethesda, Maryland
| | - Mahesh P Gupta
- Department of Surgery, Committee on Cellular and Molecular Physiology, University of Chicago, Chicago, Illinois
| | - Yu-Ying He
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, Illinois.
| |
Collapse
|
41
|
Lu J, Zhang L, Chen X, Lu Q, Yang Y, Liu J, Ma X. SIRT1 counteracted the activation of STAT3 and NF-κB to repress the gastric cancer growth. Int J Clin Exp Med 2014; 7:5050-5058. [PMID: 25664004 PMCID: PMC4307451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Accepted: 11/25/2014] [Indexed: 06/04/2023]
Abstract
Sirtuin-1 (SIRT1) possesses apparently dual roles in regulation of tumor. Previous reports have documented the crosstalk between SIRT1 with signal transducer and activator of transcription 3 (STAT3) and nuclear factor kappa-B (NF-κB) signaling in leukemia, lymphoma and myeloma. In this study, the purpose was to survey the regulatory effects of SIRT1 on gastric cancer (GC) cells (AGS and MKN-45) and the relationships between SIRT1 and activation of STAT3 and NF-κB in GC cells. We found the SIRT1 activator (resveratrol RSV) contributed to the repression of viability and increase of senescence, which were rescued by SIRT1 inhibitor (nicotinamide NA) and SIRT1 depletion by CCK-8 assay and SA-β-gal assay respectively. Further study found SIRT1 activation (RSV supplement) not only inhibited the activation of STAT3 including STAT3 mRNA level, c-myc mRNA level phosphorylated STAT3 (pSTAT3) proteins and acetylizad STAT3 (acSTAT3) proteins, but also repression of pNF-κB p65 and acNF-κB p65. NA reversed the effects of RSV. In addition, either RSV or NA application could not change the cellular viability and senescence in MKN-45 cells with STAT3 knockdown or NF-κB knockdown. Overall, our findings suggested SIRT1 activation could induced the loss of viability and increases of senescence in GC in vitro. Moreover, our observations revealed SIRT1 displayed growth inhibitory activity in GC cells highly associated with causing repression of activation of STAT3 and NF-κB proteins via deacetylation.
Collapse
Affiliation(s)
- Juanjuan Lu
- Department of Digestive Disease, Gansu Provincial Hospital 160 Donggang West Road, Lanzhou 730000, China
| | - Liping Zhang
- Department of Digestive Disease, Gansu Provincial Hospital 160 Donggang West Road, Lanzhou 730000, China
| | - Xiang Chen
- Department of Digestive Disease, Gansu Provincial Hospital 160 Donggang West Road, Lanzhou 730000, China
| | - Qiming Lu
- Department of Digestive Disease, Gansu Provincial Hospital 160 Donggang West Road, Lanzhou 730000, China
| | - Yuxia Yang
- Department of Digestive Disease, Gansu Provincial Hospital 160 Donggang West Road, Lanzhou 730000, China
| | - Jingping Liu
- Department of Digestive Disease, Gansu Provincial Hospital 160 Donggang West Road, Lanzhou 730000, China
| | - Xin Ma
- Department of Digestive Disease, Gansu Provincial Hospital 160 Donggang West Road, Lanzhou 730000, China
| |
Collapse
|
42
|
Clark-Knowles KV, Dewar-Darch D, Jardine KE, McBurney MW. Modulation of tumorigenesis by dietary intervention is not mediated by SIRT1 catalytic activity. PLoS One 2014; 9:e112406. [PMID: 25380034 PMCID: PMC4224430 DOI: 10.1371/journal.pone.0112406] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 10/14/2014] [Indexed: 12/31/2022] Open
Abstract
The protein deacetylase SIRT1 is involved in the regulation of a large number of cellular processes that are thought to be required for cancer initiation and progression. Both SIRT1 activity and tumorigenesis can be influenced by dietary fat and polyphenolics. We set out to determine whether dietary modulations of tumorigenesis are mediated by SIRT1 catalytic functions. We introduced a mammary gland tumor-inducing transgene, MMTV-PyMT, into stocks of mice bearing a H355Y point mutation in the Sirt1 gene that abolishes SIRT1 catalytic activity. Tumor latency was reduced in animals fed a high fat diet but this effect was not dependent on SIRT1 activity. Resveratrol had little effect on tumor formation except in animals heterozygous for the mutant Sirt1 gene. We conclude that the effects of these dietary interventions on tumorigenesis are not mediated by modulation of SIRT1 catalytic activity.
Collapse
MESH Headings
- Analysis of Variance
- Animals
- Antigens, Polyomavirus Transforming/genetics
- Antineoplastic Agents, Phytogenic/pharmacology
- Biocatalysis
- Cell Transformation, Neoplastic/drug effects
- Cell Transformation, Neoplastic/genetics
- Diet, High-Fat
- Heterozygote
- Male
- Mammary Neoplasms, Experimental/diet therapy
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/metabolism
- Mammary Tumor Virus, Mouse/genetics
- Mice, Transgenic
- Point Mutation
- Resveratrol
- Sirtuin 1/genetics
- Sirtuin 1/metabolism
- Stilbenes/pharmacology
- Tumor Burden/drug effects
- Tumor Burden/genetics
Collapse
Affiliation(s)
| | - Danielle Dewar-Darch
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Karen E. Jardine
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Michael W. McBurney
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
43
|
Albert MC, Brinkmann K, Kashkar H. Noxa and cancer therapy: Tuning up the mitochondrial death machinery in response to chemotherapy. Mol Cell Oncol 2014; 1:e29906. [PMID: 27308315 PMCID: PMC4905168 DOI: 10.4161/mco.29906] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 06/24/2014] [Accepted: 07/02/2014] [Indexed: 12/12/2022]
Abstract
Biochemical analyses have characterized the BH3-only protein family member Noxa as a “sensitizer” with weak pro-apoptotic activity. Investigations into cancer cell responses to chemotherapeutic agents have identified Noxa as a pivotal factor mediating the cytotoxic effect of a plethora of anticancer treatments independent of its own pro-apoptotic activity. Accumulating evidence now suggests that tumor cells exert a number of strategies to counteract Noxa function by exploiting diverse cellular regulatory circuits that normally govern Noxa expression during cellular stress responses. Here, we summarize data concerning the role of Noxa in cancer chemosensitivity and highlight the potential of this enigmatic BH3-only protein family member in current and novel anticancer therapies.
Collapse
Affiliation(s)
- Marie-Christine Albert
- Centre for Molecular Medicine Cologne (CMMC); Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD); University of Cologne, Köln, Germany; Institute for Medical Microbiology, Immunology and Hygiene (IMMIH); University of Cologne, Köln, Germany
| | - Kerstin Brinkmann
- Centre for Molecular Medicine Cologne (CMMC); Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD); University of Cologne, Köln, Germany; Institute for Medical Microbiology, Immunology and Hygiene (IMMIH); University of Cologne, Köln, Germany
| | - Hamid Kashkar
- Centre for Molecular Medicine Cologne (CMMC); Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD); University of Cologne, Köln, Germany; Institute for Medical Microbiology, Immunology and Hygiene (IMMIH); University of Cologne, Köln, Germany
| |
Collapse
|