1
|
Simón Serrano S, Tavecchio M, Mallik J, Grönberg A, Elmér E, Kifagi C, Gallay P, Hansson MJ, Massoumi R. Synergistic Effects of Sanglifehrin-Based Cyclophilin Inhibitor NV651 with Cisplatin in Hepatocellular Carcinoma. Cancers (Basel) 2022; 14:cancers14194553. [PMID: 36230472 PMCID: PMC9559492 DOI: 10.3390/cancers14194553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/08/2022] [Accepted: 09/15/2022] [Indexed: 11/16/2022] Open
Abstract
Hepatocellular carcinoma (HCC), commonly diagnosed at an advanced stage, is the most common primary liver cancer. Owing to a lack of effective HCC treatments and the commonly acquired chemoresistance, novel therapies need to be investigated. Cyclophilins-intracellular proteins with peptidyl-prolyl isomerase activity-have been shown to play a key role in therapy resistance and cell proliferation. Here, we aimed to evaluate changes in the gene expression of HCC cells caused by cyclophilin inhibition in order to explore suitable combination treatment approaches, including the use of chemoagents, such as cisplatin. Our results show that the novel cyclophilin inhibitor NV651 decreases the expression of genes involved in several pathways related to the cancer cell cycle and DNA repair. We evaluated the potential synergistic effect of NV651 in combination with other treatments used against HCC in cisplatin-sensitive cells. NV651 showed a synergistic effect in inhibiting cell proliferation, with a significant increase in intrinsic apoptosis in combination with the DNA crosslinking agent cisplatin. This combination also affected cell cycle progression and reduced the capacity of the cell to repair DNA in comparison with a single treatment with cisplatin. Based on these results, we believe that the combination of cisplatin and NV651 may provide a novel approach to HCC treatment.
Collapse
Affiliation(s)
- Sonia Simón Serrano
- Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, SE-223 63 Lund, Sweden
- Abliva AB, Medicon Village, Scheelevägen 2, SE-233 81 Lund, Sweden
| | - Michele Tavecchio
- Abliva AB, Medicon Village, Scheelevägen 2, SE-233 81 Lund, Sweden
- Mitochondrial Medicine, Department of Clinical Sciences, Lund University, BMC A13, SE-221 84 Lund, Sweden
| | - Josef Mallik
- Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, SE-223 63 Lund, Sweden
| | - Alvar Grönberg
- Abliva AB, Medicon Village, Scheelevägen 2, SE-233 81 Lund, Sweden
| | - Eskil Elmér
- Abliva AB, Medicon Village, Scheelevägen 2, SE-233 81 Lund, Sweden
- Mitochondrial Medicine, Department of Clinical Sciences, Lund University, BMC A13, SE-221 84 Lund, Sweden
| | - Chamseddine Kifagi
- NGS & OMICS Data Analysis (NODA) Consulting, Flöjtvägen 10b, SE-224 68 Lund, Sweden
| | - Philippe Gallay
- Department of Immunology & Microbial Science, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Magnus Joakim Hansson
- Abliva AB, Medicon Village, Scheelevägen 2, SE-233 81 Lund, Sweden
- Mitochondrial Medicine, Department of Clinical Sciences, Lund University, BMC A13, SE-221 84 Lund, Sweden
| | - Ramin Massoumi
- Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, SE-223 63 Lund, Sweden
- Correspondence: ; Tel.: +46-46-222-64-30
| |
Collapse
|
2
|
Zhang J, Zhang L, Nie J, Lin Y, Li Y, Xu W, Zhao JY, Zhao SM, Wang C. Calcineurin inactivation inhibits pyruvate dehydrogenase complex activity and induces the Warburg effect. Oncogene 2021; 40:6692-6702. [PMID: 34667275 DOI: 10.1038/s41388-021-02065-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 09/24/2021] [Accepted: 10/05/2021] [Indexed: 12/25/2022]
Abstract
Calcineurin is a calcium- and calmodulin-dependent serine/threonine protein phosphatase that connects the Ca2+-dependent signalling to multiple cellular responses. Calcineurin inhibitors (CNIs) have been widely used to suppress immune response in allograft patients. However, CNIs significantly increase cancer incidence in transplant recipients compared with the general population. Accumulating evidence suggests that CNIs may promote the malignant transformation of cancer cells in addition to its role in immunosuppression, but the underlying mechanisms remain poorly understood. Here, we show that calcineurin interacts with pyruvate dehydrogenase complex (PDC), a mitochondrial gatekeeper enzyme that connects two key metabolic pathways of cells, glycolysis and the tricarboxylic acid cycle. Mitochondrial-localized calcineurin dephosphorylates PDHA1 at Ser232, Ser293 and Ser300, and thus enhances PDC enzymatic activity, remodels cellular glycolysis and oxidative phosphorylation, and suppresses cancer cell proliferation. Hypoxia attenuates mitochondrial translocation of calcineurin to promote PDC inactivation. Moreover, CNIs promote metabolic remodelling and the Warburg effect by blocking calcineurin-mediated PDC activation in cancer cells. Our findings indicate that calcineurin is a critical regulator of mitochondrial metabolism and suggest that CNIs may promote tumorigenesis through inhibition of the calcineurin-PDC pathway.
Collapse
Affiliation(s)
- Jianong Zhang
- Obstetrics & Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, Key Laboratory of Reproduction Regulation of NPFPC (SIPPR, IRD), School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Liang Zhang
- Obstetrics & Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, Key Laboratory of Reproduction Regulation of NPFPC (SIPPR, IRD), School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Ji Nie
- Obstetrics & Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, Key Laboratory of Reproduction Regulation of NPFPC (SIPPR, IRD), School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Yan Lin
- Obstetrics & Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, Key Laboratory of Reproduction Regulation of NPFPC (SIPPR, IRD), School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Yao Li
- Obstetrics & Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, Key Laboratory of Reproduction Regulation of NPFPC (SIPPR, IRD), School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Wei Xu
- Shanghai Fifth People's Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Jian-Yuan Zhao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Shi-Min Zhao
- Obstetrics & Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, Key Laboratory of Reproduction Regulation of NPFPC (SIPPR, IRD), School of Life Sciences, Fudan University, Shanghai, 200438, China.
| | - Chenji Wang
- Obstetrics & Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, Key Laboratory of Reproduction Regulation of NPFPC (SIPPR, IRD), School of Life Sciences, Fudan University, Shanghai, 200438, China.
| |
Collapse
|
3
|
Abstract
Benefits of solid organ transplantation in end stage organ diseases are indisputable. Malignancy is a feared complication of solid organ transplantation and is a leading cause of mortality in patients with organ transplantation. Iatrogenic immunosuppression to prevent graft rejection plays a crucial role in the cancer development in solid organ transplant recipients. Chronic exposure to immunosuppression increases the malignancy burden through deregulation of host immune defense mechanisms and unchecked proliferation of oncogenic viruses and malignancies associated with these viruses. Vigorous screening of candidates undergoing transplant evaluation for malignancies, careful assessment of donors, and vigilant monitoring of transplant recipients are necessary to prevent, detect, and manage this life-threatening complication.
Collapse
|
4
|
Anwar S, Almatroudi A, Alsahli MA, Khan MA, Khan AA, Rahmani AH. Natural Products: Implication in Cancer Prevention and Treatment through Modulating Various Biological Activities. Anticancer Agents Med Chem 2021; 20:2025-2040. [PMID: 32628596 DOI: 10.2174/1871520620666200705220307] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 04/20/2020] [Accepted: 04/27/2020] [Indexed: 12/14/2022]
Abstract
Cancer is one of the most leading causes of death worldwide. It is one of the primary global diseases that cause morbidity and mortality in millions of people. It is usually caused by different carcinogenic agents that damage the genetic material and alter the cell signaling pathways. Carcinogens are classified into two groups as genotoxic and non-genotoxic agents. Genotoxic carcinogens are capable of directly altering the genetic material, while the non-genotoxic carcinogens are capable of producing cancer by some secondary mechanisms not related to direct gene damage. There is undoubtedly the greatest need to utilize some novel natural products as anticancer agents, as these are within reach everywhere. Interventions by some natural products aimed at decreasing the levels and conditions of these risk factors can reduce the frequency of cancer incidences. Cancer is conventionally treated by surgery, radiation therapy and chemotherapy, but such treatments may be fast-acting and causes adverse effects on normal tissues. Alternative and innovative methods of cancer treatment with the least side effects and improved efficiency are being encouraged. In this review, we discuss the different risk factors of cancer development, conventional and innovative strategies of its management and provide a brief review of the most recognized natural products used as anticancer agents globally.
Collapse
Affiliation(s)
- Shehwaz Anwar
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 52571, Saudi Arabia
| | - Ahmad Almatroudi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 52571, Saudi Arabia
| | - Mohammed A Alsahli
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 52571, Saudi Arabia
| | - Masood A Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraidah, Saudi Arabia
| | - Amjad A Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraidah, Saudi Arabia
| | - Arshad H Rahmani
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 52571, Saudi Arabia
| |
Collapse
|
5
|
Hou Y, Chen K, Liao R, Li Y, Yang H, Gong J. LINC01419-mediated epigenetic silencing of ZIC1 promotes metastasis in hepatocellular carcinoma through the PI3K/Akt signaling pathway. J Transl Med 2021; 101:570-587. [PMID: 33772101 DOI: 10.1038/s41374-021-00539-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 01/04/2021] [Accepted: 01/07/2021] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a rapidly growing tumor characterized by a high potential for vascular invasion and metastasis. The purpose of our study is to explore the regulation mechanism of long noncoding RNA (lncRNA) LINC01419 on cell-cycle distribution and metastasis in hepatocellular carcinoma (HCC) by regulating zinc finger of the cerebellum (ZIC1) through PI3K/Akt signaling pathway. Bioinformatics analysis and dual-luciferase reporter assay were used to analyze LINC01419 and related genes in HCC, and their expression in HCC tissues and adjacent normal tissues were determined by reverse transcription quantitative polymerase chain reaction (RT-qPCR) and western blot. Then, HCC cell lines were subjected to the construction of LINC01419/ZIC1 overexpression/knockdown cells utilizing lentiviral vectors. RIP and ChIP assays were applied to identify the LINC01419-binding protein. BSP and MSP assays were used to determine gene methylation. According to the results, LINC01419 was highly expressed in HCC tissues and cells, while ZIC1 was poorly expressed. LINC01419 targeted and downregulated ZIC1 expression. Furthermore, LINC01419 increased the methylation of ZIC1 promoter and repressed ZIC1 expression. PI3K/Akt signaling pathway was activated by LINC01419 overexpression and ZIC1 knockdown, under which conditions, the HCC cell self-renewal and proliferation were promoted while cell apoptosis was attenuated, accompanied by accelerated formation and metastasis of xenografted tumors in mice. In conclusion, LINC01419 enhances the methylation of ZIC1 promoter, inhibits ZIC1 expression, and activates the PI3K/Akt signaling pathway, thereby enhancing the malignant phenotypes of HCC cells in vitro as well as tumor formation and metastasis in vivo.
Collapse
Affiliation(s)
- Yifu Hou
- Organ Transplant Center and Third Department of Hepatobiliary and Pancreatic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, PR China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, PR China
| | - Kai Chen
- Organ Transplant Center and Third Department of Hepatobiliary and Pancreatic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, PR China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, PR China
| | - Rui Liao
- Department of Hepatobiliary, Southwest Medical University, Luzhou, PR China
| | - Youzan Li
- Department of Hepatobiliary, Southwest Medical University, Luzhou, PR China
| | - Hongji Yang
- Organ Transplant Center and Third Department of Hepatobiliary and Pancreatic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, PR China.
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, PR China.
| | - Jun Gong
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, PR China.
- Second Department of Hepatobiliary and Pancreatic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, PR China.
| |
Collapse
|
6
|
Gu Y, Wang C, Chen S, Tang J, Guo X, Hu W, Cui A, Zhang D, Yu K, Chen M. A Critical Role of Peptidylprolyl Isomerase A Pseudogene 22/microRNA-197-3p/Peptidylprolyl Isomerase A Axis in Hepatocellular Carcinoma. Front Genet 2021; 12:604461. [PMID: 33790943 PMCID: PMC8006304 DOI: 10.3389/fgene.2021.604461] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 02/26/2021] [Indexed: 12/09/2022] Open
Abstract
The burden of hepatocellular carcinoma (HCC) worldwide is increasing over time, while the underlying molecular mechanism of HCC development is still under exploration. Pseudogenes are classified as a special type of long non-coding RNAs (lncRNAs), and they played a vital role in regulating tumor-associated gene expression. Here, we report that a pseudogene peptidylprolyl isomerase A pseudogene 22 (PPIAP22) and its parental gene peptidylprolyl isomerase A (PPIA) were upregulated in HCC and were associated with the clinical outcomes of HCC. Further investigation revealed that PPIAP22 might upregulate the expression of PPIA through sponging microRNA (miR)-197-3p, behaving as competing endogenous RNA (ceRNA). PPIA could participate in the development of HCC by regulating mRNA metabolic process and tumor immunity based on the functional enrichment analysis. We also found a strong correlation between the expression levels of PPIA and the immune cell infiltration or the expression of chemokines, especially macrophage, C-C motif chemokine ligand 15 (CCL15), and C-X-C motif chemokine ligand 12 (CXCL12). Our findings demonstrate that the PPIAP22/miR-197-3p/PPIA axis plays a vital role in the progression of HCC by increasing the malignancy of tumor cells and regulating the immune cell infiltration, especially macrophage, through CCL15-CCR1 or CXCL12-CXCR4/CXCR7 pathways.
Collapse
Affiliation(s)
- Yuwei Gu
- Department of Infectious Diseases, Huashan Hospital, Shanghai, China
| | - Chao Wang
- Emergency Department, Huashan Hospital, Shanghai, China
| | - Shengsen Chen
- Department of Endoscopy, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Jia Tang
- Department of Infectious Diseases, Huashan Hospital, Shanghai, China
| | - Xiaoxiao Guo
- Department of Infectious Diseases, Huashan Hospital, Shanghai, China
| | - Wei Hu
- Shanghai Medical College of Fudan University, Shanghai, China
| | - An Cui
- Department of Infectious Diseases, Huashan Hospital, Shanghai, China
| | - Dian Zhang
- Emergency Department, Huashan Hospital, Shanghai, China
| | - Kangkang Yu
- Department of Infectious Diseases, Huashan Hospital, Shanghai, China
| | - Mingquan Chen
- Department of Infectious Diseases, Huashan Hospital, Shanghai, China.,Emergency Department, Huashan Hospital, Shanghai, China
| |
Collapse
|
7
|
Chen JC, Ko JC, Taso YC, Cheng HH, Chen TY, Yen TC, Lin YW. Downregulation of Xeroderma Pigmentosum Complementation Group C Expression by 17-Allylamino-17-Demethoxygeldanamycin Enhances Bevacizumab-Induced Cytotoxicity in Human Lung Cancer Cells. Pharmacology 2020; 106:154-168. [PMID: 33202406 DOI: 10.1159/000509052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 05/29/2020] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Xeroderma pigmentosum complementation group C (XPC) protein is an important DNA damage recognition factor involved in nucleotide excision repair and regulation of non-small-cell lung cancer (NSCLC) cell proliferation and viability. 17-Allylamino-17-demethoxygeldanamycin (17-AAG) blocks ATP binding to heat shock protein 90 (Hsp90), resulting in destabilization of Hsp90-client protein complexes. Vascular endothelial growth factor (VEGF) is a potent angiogenic growth factor expressed by many types of tumors. Bevacizumab (Avastin) is a humanized monoclonal antibody against human VEGF used as an antiangiogenesis agent in the therapy of many cancers, proving successful in increasing objective tumor response rate and prolonging overall survival in NSCLC patients. METHODS After the bevacizumab and/or 17-AAG treatment, the expressions of XPC mRNA were determined by quantitative real-time PCR analysis. Protein levels of XPC and phospho-AKT were determined by Western blot analysis. We used specific XPC small interfering RNA and PI3K inhibitor (LY294002) to examine the role of the AKT-XPC signal in regulating the chemosensitivity of bevacizumab and 17-AAG. Cell viability was assessed by the MTS assay and trypan blue exclusion assay. RESULTS In this study, bevacizumab decreased XPC expression in human lung squamous cell carcinoma H520 and H1703 cells via AKT inactivation. Enhancement of AKT activity by transfection with constitutively active AKT vectors increased XPC expression and cell survival after treatment with bevacizumab. In addition, 17-AAG synergistically enhanced bevacizumab-induced cytotoxicity and cell growth inhibition in H520 and H1703 cells, associated with downregulation of XPC expression and inactivation of AKT. DISCUSSION/CONCLUSION Together, these results may provide a rationale to combine bevacizumab with Hsp90 inhibitors in future to enhance therapeutic effects for lung cancer.
Collapse
Affiliation(s)
- Jyh-Cheng Chen
- Department of Food Science, National Chiayi University, Chiayi, Taiwan
| | - Jen-Chung Ko
- Department of Internal Medicine, National Taiwan University Hospital, Hsin-Chu Branch, Hsin-Chu, Taiwan
| | - Yong-Cing Taso
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi, Taiwan
| | - Hsiang-Hung Cheng
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi, Taiwan
| | - Tzu-Ying Chen
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi, Taiwan
| | - Ting-Chuan Yen
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi, Taiwan
| | - Yun-Wei Lin
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi, Taiwan,
| |
Collapse
|
8
|
Ume AC, Pugh JM, Kemp MG, Williams CR. Calcineurin inhibitor (CNI)-associated skin cancers: New insights on exploring mechanisms by which CNIs downregulate DNA repair machinery. PHOTODERMATOLOGY, PHOTOIMMUNOLOGY & PHOTOMEDICINE 2020; 36:433-440. [PMID: 32786098 PMCID: PMC11042075 DOI: 10.1111/phpp.12600] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/22/2020] [Accepted: 08/02/2020] [Indexed: 12/31/2022]
Abstract
The use of the calcineurin inhibitors (CNI) cyclosporine (CsA) and tacrolimus remains a cornerstone in post-transplantation immunosuppression. Although these immunosuppressive agents have revolutionized the field of transplantation medicine, its increased skin cancer risk poses a major concern. A key contributor to this phenomenon is a reduced capacity to repair DNA damage caused by exposure to ultraviolet (UV) wavelengths of sunlight. CNIs decrease DNA repair by mechanisms that remain to be fully explored. Though CsA is known to decrease the abundance of key DNA repair enzymes, less is known about how tacrolimus yields this effect. CNIs hold the capacity to inhibit both of the main catalytic calcineurin isoforms (CnAα and CnAβ). However, it is unknown which isoform regulates UV-induced DNA repair, which is the focus of this review. It is with hope that this insight spurs investigative efforts that conclusively addresses these gaps in knowledge. Additionally, this research also raises the possibility that newer CNIs can be developed that effectively blunt the immune response while mitigating the incidence of skin cancers with immunosuppression.
Collapse
Affiliation(s)
- Adaku C. Ume
- Department of Neuroscience, Cell Biology & Physiology, College of Science and Mathematics, Wright State University Boonshoft School of Medicine, Dayton, Ohio
| | - Jennifer M. Pugh
- Department of Neuroscience, Cell Biology & Physiology, College of Science and Mathematics, Wright State University Boonshoft School of Medicine, Dayton, Ohio
| | - Michael G. Kemp
- Department of Pharmacology & Toxicology, Wright State University Boonshoft School of Medicine, Dayton, Ohio
| | - Clintoria R. Williams
- Department of Neuroscience, Cell Biology & Physiology, College of Science and Mathematics, Wright State University Boonshoft School of Medicine, Dayton, Ohio
| |
Collapse
|
9
|
Corchado-Cobos R, García-Sancha N, González-Sarmiento R, Pérez-Losada J, Cañueto J. Cutaneous Squamous Cell Carcinoma: From Biology to Therapy. Int J Mol Sci 2020; 21:ijms21082956. [PMID: 32331425 PMCID: PMC7216042 DOI: 10.3390/ijms21082956] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/18/2020] [Accepted: 04/20/2020] [Indexed: 12/13/2022] Open
Abstract
Cutaneous squamous cell carcinoma (CSCC) is the second most frequent cancer in humans and its incidence continues to rise. Although CSCC usually display a benign clinical behavior, it can be both locally invasive and metastatic. The signaling pathways involved in CSCC development have given rise to targetable molecules in recent decades. In addition, the high mutational burden and increased risk of CSCC in patients under immunosuppression were part of the rationale for developing the immunotherapy for CSCC that has changed the therapeutic landscape. This review focuses on the molecular basis of CSCC and the current biology-based approaches of targeted therapies and immune checkpoint inhibitors. Another purpose of this review is to explore the landscape of drugs that may induce or contribute to the development of CSCC. Beginning with the pathogenetic basis of these drug-induced CSCCs, we move on to consider potential therapeutic opportunities for overcoming this adverse effect.
Collapse
Affiliation(s)
- Roberto Corchado-Cobos
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC)-Centro de Investigación del cáncer (CIC)-CSIC, Laboratory 7, 37007 Salamanca, Spain; (R.C.-C.); (N.G.-S.); (J.P.-L.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Complejo Asistencial Universitario de Salamanca, Hospital Virgen de la Vega, 37007 Salamanca, Spain;
| | - Natalia García-Sancha
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC)-Centro de Investigación del cáncer (CIC)-CSIC, Laboratory 7, 37007 Salamanca, Spain; (R.C.-C.); (N.G.-S.); (J.P.-L.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Complejo Asistencial Universitario de Salamanca, Hospital Virgen de la Vega, 37007 Salamanca, Spain;
| | - Rogelio González-Sarmiento
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Complejo Asistencial Universitario de Salamanca, Hospital Virgen de la Vega, 37007 Salamanca, Spain;
- Molecular Medicine Unit, Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
| | - Jesús Pérez-Losada
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC)-Centro de Investigación del cáncer (CIC)-CSIC, Laboratory 7, 37007 Salamanca, Spain; (R.C.-C.); (N.G.-S.); (J.P.-L.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Complejo Asistencial Universitario de Salamanca, Hospital Virgen de la Vega, 37007 Salamanca, Spain;
| | - Javier Cañueto
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC)-Centro de Investigación del cáncer (CIC)-CSIC, Laboratory 7, 37007 Salamanca, Spain; (R.C.-C.); (N.G.-S.); (J.P.-L.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Complejo Asistencial Universitario de Salamanca, Hospital Virgen de la Vega, 37007 Salamanca, Spain;
- Department of Dermatology, Complejo Asistencial Universitario de Salamanca, 37007 Salamanca, Spain
- Correspondence: ; Tel.: +34-923-291-100 (ext. 55574)
| |
Collapse
|
10
|
Shah P, Zhao B, Qiang L, He YY. Phosphorylation of xeroderma pigmentosum group C regulates ultraviolet-induced DNA damage repair. Nucleic Acids Res 2019; 46:5050-5060. [PMID: 29660033 PMCID: PMC6007576 DOI: 10.1093/nar/gky239] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 03/21/2018] [Indexed: 12/18/2022] Open
Abstract
Nucleotide excision repair (NER) is the most versatile DNA repair system that removes bulky DNA damage induced by various endogenous and exogenous factors, including UV radiation. Defects in NER can lead to the xeroderma pigmentosum (XP) syndrome, mainly characterized by increased carcinogenesis in the skin. The function of NER factors, including xeroderma pigmentosum group C (XPC), can be regulated by post-translational modifications such as ubiquitination. However, the role of phosphorylation in XPC function remains unknown. Here, we show that phosphorylation of XPC acts as a novel post-translational regulatory mechanism of the NER pathway. We show that XPC is phosphorylated at serine 94. Moreover, after UVB irradiation, XPC phosphorylation regulates recruitment of ubiquitinated XPC and its downstream NER factors to the chromatin. In addition, upon evaluating the predicted kinases for XPC phosphorylation, we found that casein kinase II (CK2) promotes NER. Furthermore, CK2 kinase mediates XPC phosphorylation at serine 94, and also promotes recruitment of ubiquitinated XPC to the chromatin after UVB irradiation. Our findings have identified XPC phosphorylation as a new mechanism for regulating NER following UV-induced DNA damage.
Collapse
Affiliation(s)
- Palak Shah
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL 60637, USA.,Committee on Molecular Pathogenesis and Molecular Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Baozhong Zhao
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL 60637, USA
| | - Lei Qiang
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL 60637, USA.,School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210008, China
| | - Yu-Ying He
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL 60637, USA.,Committee on Molecular Pathogenesis and Molecular Medicine, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
11
|
Chen JC, Wu CH, Peng YS, Zheng HY, Lin YC, Ma PF, Yen TC, Chen TY, Lin YW. Astaxanthin enhances erlotinib-induced cytotoxicity by p38 MAPK mediated xeroderma pigmentosum complementation group C (XPC) down-regulation in human lung cancer cells. Toxicol Res (Camb) 2018; 7:1247-1256. [PMID: 30555679 DOI: 10.1039/c7tx00292k] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 09/07/2018] [Indexed: 12/11/2022] Open
Abstract
Astaxanthin has been demonstrated to exhibit a wide range of beneficial effects that include anti-cancer and anti-inflammatory properties. Xeroderma pigmentosum complementation group C (XPC) protein is an important DNA damage recognition factor in nucleotide excision repair and is involved in regulating non-small cell lung cancer (NSCLC) cell proliferation and viability. Erlotinib (TarcevaR) is a selective epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor that has demonstrated clinical activity in NSCLC cells. However, whether astaxanthin and erlotinib could induce synergistic cytotoxicity in NSCLC cells through modulating XPC expression is unknown. In this study, we found that p38 MAPK activation by astaxanthin decreased XPC expression in two human lung adenocarcinoma A549 and H1975 cells. Inactivation of p38 MAPK by pharmacological inhibitor SB203580 or the specific small interfering RNA (siRNA) rescued the astaxanthin-reduced XPC mRNA and protein levels. Enforced expression of XPC cDNA or inhibiting the p38 MAPK activity reduced the cytotoxicity and cell growth inhibition of astaxanthin. In contrast, knockdown of XPC using siRNA enhanced the cytotoxic effects of astaxanthin. Moreover, astaxanthin synergistically enhanced cytotoxicity and cell growth inhibition of erlotinib in NSCLC cells, which were associated with the down-regulation of XPC expression and activation of p38 MAPK. Our findings suggested that the astaxanthin induced p38 MAPK mediated XPC down-regulation enhanced the erlotinib-induced cytotoxicity in A549 and H1975 cells.
Collapse
Affiliation(s)
- Jyh-Cheng Chen
- Department of Food Science , National Chiayi University , Chiayi , Taiwan
| | - Chia-Hung Wu
- Department of Biochemical Science and Technology , National Chiayi University , Chiayi , Taiwan . ; ; Tel: +886-5-271-7770
| | - Yi-Shuan Peng
- Department of Biochemical Science and Technology , National Chiayi University , Chiayi , Taiwan . ; ; Tel: +886-5-271-7770
| | - Hao-Yu Zheng
- Department of Biochemical Science and Technology , National Chiayi University , Chiayi , Taiwan . ; ; Tel: +886-5-271-7770
| | - Yuan-Cheng Lin
- Department of Biochemical Science and Technology , National Chiayi University , Chiayi , Taiwan . ; ; Tel: +886-5-271-7770
| | - Peng-Fang Ma
- Department of Biochemical Science and Technology , National Chiayi University , Chiayi , Taiwan . ; ; Tel: +886-5-271-7770
| | - Ting-Chuan Yen
- Department of Biochemical Science and Technology , National Chiayi University , Chiayi , Taiwan . ; ; Tel: +886-5-271-7770
| | - Tzu-Ying Chen
- Department of Biochemical Science and Technology , National Chiayi University , Chiayi , Taiwan . ; ; Tel: +886-5-271-7770
| | - Yun-Wei Lin
- Department of Biochemical Science and Technology , National Chiayi University , Chiayi , Taiwan . ; ; Tel: +886-5-271-7770
| |
Collapse
|
12
|
Marzejon M, Kosowska M, Majchrowicz D, Bułło-Piontecka B, Wąsowicz M, Jędrzejewska-Szczerska M. Optical-Spectrometry-Based Method for Immunosuppressant Medicine Level Detection in Aqueous Solutions. SENSORS (BASEL, SWITZERLAND) 2018; 18:E2001. [PMID: 29932121 PMCID: PMC6068911 DOI: 10.3390/s18072001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 06/19/2018] [Accepted: 06/20/2018] [Indexed: 01/28/2023]
Abstract
In this paper, an investigation into detecting immunosuppressive medicine in aqueous solutions using a spectrometry-based technique is described. Using optical transmissive spectrometry, absorbance measurements in the spectra range from 250 nm to 1000 nm were carried out for different cyclosporine A (CsA) concentrations in aqueous solutions. The experiment was conducted for samples both with and without interferent substances—glucose and sodium chloride. Using a dedicated algorithm, the measured data was analyzed and a high correlation coefficient R² = 0.8647 was achieved. The experiment showed that the described technique allowed for the detection of various CsA concentration levels in a selective, label-free and simple way. This method could be used in medicine, veterinary medicine and laboratory diagnostics.
Collapse
Affiliation(s)
- Marcin Marzejon
- Department of Metrology and Optoelectronics, Faculty of Electronics, Telecommunications and Informatics, Gdańsk University of Technology, 11/12 Narutowicza Street, 80-233 Gdansk, Poland.
| | - Monika Kosowska
- Department of Metrology and Optoelectronics, Faculty of Electronics, Telecommunications and Informatics, Gdańsk University of Technology, 11/12 Narutowicza Street, 80-233 Gdansk, Poland.
| | - Daria Majchrowicz
- Department of Metrology and Optoelectronics, Faculty of Electronics, Telecommunications and Informatics, Gdańsk University of Technology, 11/12 Narutowicza Street, 80-233 Gdansk, Poland.
| | - Barbara Bułło-Piontecka
- Department of Nephrology, Transplantology and Internal Diseases, Faculty of Medicine, Medical University of Gdańsk, 3a Marii Skłodowskiej-Curie Street, 80-210 Gdansk, Poland.
| | - Michał Wąsowicz
- Department of Morphological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences, 159 Nowoursynowska Street, 02-776 Warszawa, Poland.
| | - Małgorzata Jędrzejewska-Szczerska
- Department of Metrology and Optoelectronics, Faculty of Electronics, Telecommunications and Informatics, Gdańsk University of Technology, 11/12 Narutowicza Street, 80-233 Gdansk, Poland.
| |
Collapse
|
13
|
Zhao X, Xia C, Wang X, Wang H, Xin M, Yu L, Liang Y. Cyclophilin J PPIase Inhibitors Derived from 2,3-Quinoxaline-6 Amine Exhibit Antitumor Activity. Front Pharmacol 2018. [PMID: 29520233 PMCID: PMC5826973 DOI: 10.3389/fphar.2018.00126] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cyclophilin J (CyPJ), also called peptidylprolyl isomerase like 3, has been identified as a novel member of the cyclophilin family. Our previous research has resolved the three-dimensional structure of CyPJ and demonstrated the peptidylprolyl cis–trans isomerase (PPIase) activity of CyPJ, which can be inhibited by the common immunosuppressive drug cyclosporine A (CsA). Importantly, CyPJ is upregulated in hepatocellular carcinoma (HCC) and promotes tumor growth; CyPJ inhibition by CsA- or siRNA-based knockdown results in a remarkable suppression of HCC. These findings suggest that CyPJ may be a potential therapeutic target for HCC, and discovery of relevant inhibitors may facilitate development of a novel CyPJ-based targeting therapy. However, apart from the common inhibitor CsA, CyPJ has yet to be investigated as a target for cancer therapy. Here, we report structure-based identification of novel small molecule non-peptidic CyPJ inhibitors and their potential as antitumor lead compounds. Based on computer-aided virtual screening, in silico, and subsequently surface plasmon resonance analysis, 19 potential inhibitors of CyPJ were identified and selected for further evaluation of PPIase CyPJ inhibition in vitro. Thirteen out of 19 compounds exhibited notable inhibition against PPIase activity. Among them, the compound ZX-J-19, with a quinoxaline nucleus, showed potential for tumor inhibition; thus, we selected it for further structure–activity optimization. A total of 22 chemical derivatives with 2,3-substituted quinoxaline-6-amine modifications were designed and successfully synthesized. At least 2 out of the 22 derivatives, such as ZX-J-19j and ZX-J-19l, demonstrated remarkable inhibition of tumor cell growth, comparable to CsA but much stronger than 5-fluorouracil. These results indicate that these two small molecules represent novel potential lead compounds for CyPJ-based antitumor drug development.
Collapse
Affiliation(s)
- Xuemei Zhao
- College of Pharmacy, Taishan Medical University, Tai'an, China
| | - Chengcai Xia
- College of Pharmacy, Taishan Medical University, Tai'an, China
| | - Xiaodan Wang
- College of Pharmacy, Taishan Medical University, Tai'an, China
| | - Hao Wang
- College of Pharmacy, Taishan Medical University, Tai'an, China
| | - Ming Xin
- College of Pharmacy, Taishan Medical University, Tai'an, China
| | - Long Yu
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Yulong Liang
- College of Pharmacy, Taishan Medical University, Tai'an, China.,Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
14
|
Abstract
Merkel cell carcinoma (MCC) is a rare and aggressive skin cancer with a neuroendocrine phenotype. Incidence varies according to the geographic regions but is overall increasing. Different risk factors have been identified namely advanced age, immunosuppression, and ultraviolet light exposure. An association between MCC and polyomavirus infection is known. However, the exact mechanism that leads to carcinogenesis is yet to be fully understood. Surgery when feasible is the recommended treatment for localized disease, followed by adjuvant radiation or chemoradiation. In the metastatic setting, chemotherapy has been the standard treatment. However, two recently published trials with immune checkpoint inhibitors in first and second line showed promising results with a tolerable safety profile and these might become the standard therapy shortly. Somatostatin receptors are expressed in many MCC but such expression is not associated with disease severity. Presently there are no biomarkers predictive of response that could help to better select patients to these new therapies, and additional research is essential.
Collapse
Affiliation(s)
- Teresa Amaral
- Center for Dermatooncology, Department of Dermatology, University Hospital Tuebingen, Liebermeisterstr. 25, 72076, Tuebingen, Germany
- Portuguese Air Force Health Direction, Paço do Lumiar, 1649-020, Lisbon, Portugal
| | - Ulrike Leiter
- Center for Dermatooncology, Department of Dermatology, University Hospital Tuebingen, Liebermeisterstr. 25, 72076, Tuebingen, Germany
| | - Claus Garbe
- Center for Dermatooncology, Department of Dermatology, University Hospital Tuebingen, Liebermeisterstr. 25, 72076, Tuebingen, Germany.
| |
Collapse
|
15
|
Madeleine M, Patel N, Plasmeijer E, Engels E, Bouwes Bavinck J, Toland A, Green A. Epidemiology of keratinocyte carcinomas after organ transplantation. Br J Dermatol 2017; 177:1208-1216. [DOI: 10.1111/bjd.15931] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2017] [Indexed: 12/14/2022]
Affiliation(s)
| | - N.S. Patel
- University of South Florida Tampa FL U.S.A
| | - E.I. Plasmeijer
- QIMR Berghofer Medical Research Institute Queensland Australia
| | | | | | - A.E. Toland
- The Ohio State University Medical Center Columbus OH U.S.A
| | - A.C. Green
- QIMR Berghofer Medical Research Institute Queensland Australia
- CRUK Manchester Institute and University of Manchester Manchester Academic Health Sciences Centre Manchester U.K
| | | |
Collapse
|
16
|
Zamoiski RD, Yanik E, Gibson TM, Cahoon EK, Madeleine MM, Lynch CF, Gustafson S, Goodman MT, Skeans M, Israni AK, Engels EA, Morton LM. Risk of Second Malignancies in Solid Organ Transplant Recipients Who Develop Keratinocyte Cancers. Cancer Res 2017; 77:4196-4203. [PMID: 28615224 PMCID: PMC5540772 DOI: 10.1158/0008-5472.can-16-3291] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Revised: 03/06/2017] [Accepted: 06/05/2017] [Indexed: 01/07/2023]
Abstract
Solid organ transplant recipients have increased risk for developing keratinocyte cancers, including cutaneous squamous cell carcinoma (SCC) and basal cell carcinoma (BCC), in part as a result of immunosuppressive medications administered to prevent graft rejection. In the general population, keratinocyte cancers are associated with increased risks of subsequent malignancy, however, the risk in organ transplant populations has not been evaluated. We addressed this question by linking the U.S. Scientific Registry of Transplant Recipients, which includes data on keratinocyte cancer occurrence, with 15 state cancer registries. Risk of developing malignancies after keratinocyte cancer was assessed among 118,440 Caucasian solid organ transplant recipients using multivariate Cox regression models. Cutaneous SCC occurrence (n = 6,169) was associated with 1.44-fold increased risk [95% confidence interval (CI), 1.31-1.59] for developing later malignancies. Risks were particularly elevated for non-cutaneous SCC, including those of the oral cavity/pharynx (HR, 5.60; 95% CI, 4.18-7.50) and lung (HR, 1.66; 95% CI, 1.16-2.31). Cutaneous SCC was also associated with increased risk of human papillomavirus-related cancers, including anal cancer (HR, 2.77; 95% CI, 1.29-5.96) and female genital cancers (HR, 3.43; 95% CI, 1.44-8.19). In contrast, BCC (n = 3,669) was not associated with overall risk of later malignancy (HR, 0.98; 95% CI, 0.87-1.12), including any SCC. Our results suggest that transplant recipients with cutaneous SCC, but not BCC, have an increased risk of developing other SCC. These findings somewhat differ from those for the general population and suggest a shared etiology for cutaneous SCC and other SCC in the setting of immunosuppression. Cutaneous SCC occurrence after transplantation could serve as a marker for elevated malignancy risk. Cancer Res; 77(15); 4196-203. ©2017 AACR.
Collapse
Affiliation(s)
- Rachel D Zamoiski
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Department of Health and Human Services, Bethesda, Maryland
| | - Elizabeth Yanik
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Department of Health and Human Services, Bethesda, Maryland
| | - Todd M Gibson
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Elizabeth K Cahoon
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Department of Health and Human Services, Bethesda, Maryland
| | - Margaret M Madeleine
- Public Health Science, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Charles F Lynch
- Department of Epidemiology, University of Iowa, Iowa City, Iowa
| | - Sally Gustafson
- Scientific Registry of Transplant Recipients, Minneapolis, Minnesota
| | - Marc T Goodman
- Cancer Prevention and Control Program, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Melissa Skeans
- Scientific Registry of Transplant Recipients, Minneapolis, Minnesota
| | - Ajay K Israni
- Department of Epidemiology and Community Health, University of Minnesota, Minneapolis, Minnesota
| | - Eric A Engels
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Department of Health and Human Services, Bethesda, Maryland
| | - Lindsay M Morton
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Department of Health and Human Services, Bethesda, Maryland.
| |
Collapse
|
17
|
Engels EA. Cancer in Solid Organ Transplant Recipients: There Is Still Much to Learn and Do. Am J Transplant 2017; 17:1967-1969. [PMID: 28394489 PMCID: PMC6309190 DOI: 10.1111/ajt.14140] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 11/15/2016] [Accepted: 11/19/2016] [Indexed: 01/25/2023]
|
18
|
Yanik EL, Pfeiffer RM, Freedman DM, Weinstock MA, Cahoon EK, Arron ST, Chaloux M, Connolly MK, Nagarajan P, Engels EA. Spectrum of Immune-Related Conditions Associated with Risk of Keratinocyte Cancers among Elderly Adults in the United States. Cancer Epidemiol Biomarkers Prev 2017; 26:998-1007. [PMID: 28377416 DOI: 10.1158/1055-9965.epi-17-0003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 01/27/2017] [Accepted: 03/03/2017] [Indexed: 11/16/2022] Open
Abstract
Background: Elevated keratinocyte carcinoma risk is present with several immune-related conditions, e.g., solid organ transplantation and non-Hodgkin lymphoma. Because many immune-related conditions are rare, their relationships with keratinocyte carcinoma have not been studied.Methods: We used Medicare claims to identify cutaneous squamous cell carcinoma (SCC) and basal cell carcinoma (BCC) cases in 2012, and controls matched on sex and age. All subjects were aged 65 to 95 years, of white race, and had attended ≥1 dermatologist visit in 2010-2011. Immune-related conditions were identified during 1999-2011 using Medicare claims. Associations were estimated with logistic regression, with statistical significance determined after Bonferroni correction for multiple comparisons.Results: We included 258,683 SCC and 304,903 BCC cases. Of 47 immune-related conditions, 21 and 9 were associated with increased SCC and BCC risk, respectively. We identified strongly elevated keratinocyte carcinoma risk with solid organ transplantation (SCC OR = 5.35; BCC OR = 1.94) and non-Hodgkin lymphoma (SCC OR = 1.62; BCC OR = 1.25). We identified associations with common conditions, e.g., rheumatoid arthritis [SCC OR = 1.06, 95% confidence interval (95% CI), 1.04-1.09] and Crohn's disease (SCC OR = 1.33, 95% CI, 1.27-1.39; BCC OR = 1.10, 95% CI, 1.05-1.15), and rare or poorly characterized conditions, e.g., granulomatosis with polyangiitis (SCC OR = 1.88; 95% CI, 1.61-2.19), autoimmune hepatitis (SCC OR = 1.81; 95% CI, 1.52-2.16), and deficiency of humoral immunity (SCC OR = 1.51, 95% CI, 1.41-1.61; BCC OR = 1.22, 95% CI, 1.14-1.31). Most conditions were more positively associated with SCC than BCC. Associations were generally consistent regardless of prior keratinocyte carcinoma history.Conclusions: Many immune-related conditions are associated with elevated keratinocyte carcinoma risk and appear more tightly linked to SCC.Impact: Immunosuppression or immunosuppressive treatment may increase keratinocyte carcinoma risk, particularly SCC. Cancer Epidemiol Biomarkers Prev; 26(7); 998-1007. ©2017 AACR.
Collapse
Affiliation(s)
- Elizabeth L Yanik
- Department of Orthopaedic Surgery, Washington University, St. Louis, Missouri.
| | - Ruth M Pfeiffer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - D Michal Freedman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Martin A Weinstock
- Department of Dermatology and Department of Epidemiology, Brown University, Providence, Rhode Island
| | - Elizabeth K Cahoon
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Sarah T Arron
- Department of Dermatology, University of California, San Francisco, California
| | | | - M Kari Connolly
- Department of Dermatology, University of California, San Francisco, California
| | | | - Eric A Engels
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
19
|
Sun CC, Li SJ, Yuan ZP, Li DJ. MicroRNA-346 facilitates cell growth and metastasis, and suppresses cell apoptosis in human non-small cell lung cancer by regulation of XPC/ERK/Snail/E-cadherin pathway. Aging (Albany NY) 2016; 8:2509-2524. [PMID: 27777383 PMCID: PMC5115903 DOI: 10.18632/aging.101080] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Accepted: 10/04/2016] [Indexed: 01/02/2023]
Abstract
Determinants of growth and metastasis in cancer remain of great interest to define. MicroRNAs (miRNAs) have frequently emerged as tumor metastatic regulator by acting on multiple signaling pathways. Here we report the definition of miR-346 as a novel oncogenic microRNA that facilitates non-small cell lung cancer (NSCLC) cell growth and metastasis. XPC, an important DNA damage recognition factor in nucleotide excision repair was defined as a target for down-regulation by miR-346, functioning through direct interaction with the 3'-UTR of XPC mRNA. Blocking miR-346 by an antagomiR was sufficient to inhibit NSCLC cell growth and metastasis, an effect that could be phenol-copied by RNAi-mediated silencing of XPC. In vivo studies established that miR-346 overexpression was sufficient to promote tumor growth by A549 cells in xenografts mice, relative to control cells. Overall, our results defined miR-346 as an oncogenic miRNA in NSCLC, the levels of which contributed to tumor growth and invasive aggressiveness.
Collapse
Affiliation(s)
- Cheng-Cao Sun
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, 430071 Wuhan, P. R. China
| | - Shu-Jun Li
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, 430071 Wuhan, P. R. China
- Wuhan Hospital for the Prevention and Treatment of Occupational Diseases, 430071 Wuhan, P. R. China
| | - Zhan-Peng Yuan
- Department of Toxicology, School of Public Health, Wuhan University, 430071 Wuhan, P. R. China
| | - De-Jia Li
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, 430071 Wuhan, P. R. China
| |
Collapse
|
20
|
Cui T, Srivastava AK, Han C, Yang L, Zhao R, Zou N, Qu M, Duan W, Zhang X, Wang QE. XPC inhibits NSCLC cell proliferation and migration by enhancing E-Cadherin expression. Oncotarget 2016; 6:10060-72. [PMID: 25871391 PMCID: PMC4496340 DOI: 10.18632/oncotarget.3542] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Accepted: 02/13/2015] [Indexed: 02/07/2023] Open
Abstract
Xeroderma pigmentosum complementation group C (XPC) protein is an important DNA damage recognition factor in nucleotide excision repair. Deletion of XPC is associated with early stages of human lung carcinogenesis, and reduced XPC mRNA levels predict poor patient outcome for non-small cell lung cancer (NSCLC). However, the mechanisms linking loss of XPC expression and poor prognosis in lung cancer are still unclear. Here, we report evidence that XPC silencing drives proliferation and migration of NSCLC cells by down-regulating E-Cadherin. XPC knockdown enhanced proliferation and migration while decreasing E-Cadherin expression in NSCLC cells with an epithelial phenotype. Restoration of E-Cadherin in these cells suppressed XPC knockdown-induced cell growth both in vitro and in vivo. Mechanistic studies showed that the loss of XPC repressed E-Cadherin expression by activating the ERK pathway and upregulating Snail expression. Our findings indicate that XPC silencing-induced reduction of E-Cadherin expression contributes, at least in part, to the poor outcome of NSCLC patients with low XPC expression.
Collapse
Affiliation(s)
- Tiantian Cui
- Department of Radiology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Amit Kumar Srivastava
- Department of Radiology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Chunhua Han
- Department of Radiology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Linlin Yang
- Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Ran Zhao
- Department of Radiology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Ning Zou
- Department of Radiology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Meihua Qu
- Department of Radiology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Wenrui Duan
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Xiaoli Zhang
- Center for Biostatistics, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Qi-En Wang
- Department of Radiology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
21
|
BERTUCCELLI GIUSEPPE, ZERBINATI NICOLA, MARCELLINO MASSIMILIANO, NANDA KUMAR NAVALPURSHANMUGAM, HE FANG, TSEPAKOLENKO VLADIMIR, CERVI JOSEPH, LORENZETTI ALDO, MAROTTA FRANCESCO. Effect of a quality-controlled fermented nutraceutical on skin aging markers: An antioxidant-control, double-blind study. Exp Ther Med 2016; 11:909-916. [PMID: 26998011 PMCID: PMC4774357 DOI: 10.3892/etm.2016.3011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 09/28/2015] [Indexed: 02/07/2023] Open
Abstract
The aim of the present study was to determine whether oral supplementation with a fermented papaya preparation (FPP-treated group) or an antioxidant cocktail (antioxidant-control group, composed of 10 mg trans-resveratrol, 60 µg selenium, 10 mg vitamin E and 50 mg vitamin C) was able to improve the skin antioxidant capacity and the expression of key skin genes, while promoting skin antiaging effects. The study enrolled 60 healthy non-smoker males and females aged 40-65 years, all of whom showed clinical signs of skin aging. The subjects were randomly divided into two matched groups, and were administered FPP or antioxidant treatment of a 4.5 g/day sachet sublingually twice a day for 90 days in a double-blind fashion. The parameters investigated were: Skin surface, brown spots, skin evenness, skin moisturization, elasticity (face), redox balance, nitric oxide (NO) concentration, and the expression levels of key genes (outer forearm sample). As compared with the baseline (day 0) and antioxidant-control values, FPP-treated subjects showed a significant improvement in skin evenness, moisturization and elasticity. The two treatments improved the MDA and SOD skin concentrations, but only the FPP-treated group showed a higher SOD level and a significant NO increase, along with significant upregulation of acquaporin-3 and downregulation of the potentially pro-aging/carcinogenetic cyclophilin-A and CD147 genes (P<0.05). Progerin was unaffected in both treatment groups. In conclusion, these findings suggest that orally-administered FPP showed a consistent biological and gene-regulatory improvement in the skin, as was also demonstrated in previous experimental and clinical trials testing other tissues, while common oral antioxidants had only a minor effect.
Collapse
Affiliation(s)
| | - NICOLA ZERBINATI
- Dermatology Unit, CMP-Medical Center and Laboratories, Pavia 27100, Italy
| | | | - NAVALPUR SHANMUGAM NANDA KUMAR
- Department of Pediatrics, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - FANG HE
- Department of Nutrition and Food Hygiene, West China School of Public Health, Sichuan University, Chengdu, Sichuan 610065, P.R. China
| | - VLADIMIR TSEPAKOLENKO
- Virtus Medical Center, Ukraine Institute of Plastic Surgery and Dermatology, Odessa 68355, Ukraine
| | - JOSEPH CERVI
- ReGenera Research Group for Aging Intervention, Milan 20154, Italy
| | - ALDO LORENZETTI
- ReGenera Research Group for Aging Intervention, Milan 20154, Italy
| | - FRANCESCO MAROTTA
- ReGenera Research Group for Aging Intervention, Milan 20154, Italy
- Correspondence to: Professor Francesco Marotta, ReGenera Research Group for Aging-Intervention, 12 Piazza Firenze, Milan 20154, Italy, E-mail:
| |
Collapse
|
22
|
Robbins HA, Clarke CA, Arron ST, Tatalovich Z, Kahn AR, Hernandez BY, Paddock L, Yanik EL, Lynch CF, Kasiske BL, Snyder J, Engels EA. Melanoma Risk and Survival among Organ Transplant Recipients. J Invest Dermatol 2015; 135:2657-2665. [PMID: 26270022 PMCID: PMC4640996 DOI: 10.1038/jid.2015.312] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 04/05/2015] [Accepted: 06/10/2015] [Indexed: 01/07/2023]
Abstract
Solid organ transplant recipients, who are medically immunosuppressed to prevent graft rejection, have increased melanoma risk, but risk factors and outcomes are incompletely documented. We evaluated melanoma incidence among 139,991 non-Hispanic white transplants using linked US transplant-cancer registry data (1987-2010). We used standardized incidence ratios (SIRs) to compare incidence with the general population and incidence rate ratios (IRRs) from multivariable Poisson models to assess risk factors. Separately, we compared post-melanoma survival among transplant recipients (n=182) and non-recipients (n=131,358) using multivariable Cox models. Among transplant recipients, risk of invasive melanoma (n=519) was elevated (SIR=2.20, 95% CI 2.01-2.39), especially for regional stage tumors (SIR=4.11, 95% CI 3.27-5.09). Risk of localized tumors was stable over time after transplantation but higher with azathioprine maintenance therapy (IRR=1.35, 95% CI 1.03-1.77). Risk of regional/distant stage tumors peaked within 4 years following transplantation and increased with polyclonal antibody induction therapy (IRR=1.65, 95% CI 1.02-2.67). Melanoma-specific mortality was higher among transplant recipients than non-recipients (hazard ratio 2.98, 95% CI 2.26-3.93). Melanoma exhibits increased incidence and aggressive behavior under transplant-related immunosuppression. Some localized melanomas may result from azathioprine, which acts synergistically with UV radiation, whereas T-cell-depleting induction therapies may promote late-stage tumors. Our findings support sun safety practices and skin screening for transplant recipients.
Collapse
Affiliation(s)
- Hilary A Robbins
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland, USA.
| | - Christina A Clarke
- Cancer Prevention Institute of California, Fremont, California, USA; Department of Health Research and Policy, Stanford University School of Medicine and Stanford Cancer Institute, Palo Alto, California, USA
| | - Sarah T Arron
- Department of Dermatology, University of California, San Francisco, San Francisco, California, USA
| | - Zaria Tatalovich
- Division of Cancer Control and Population Sciences, National Cancer Institute, National Institutes of Health, Rockville, Maryland, USA
| | - Amy R Kahn
- New York State Cancer Registry, Albany, New York, USA
| | - Brenda Y Hernandez
- University of Hawaii Cancer Center, University of Hawaii, Honolulu, Hawaii, USA
| | - Lisa Paddock
- New Jersey State Cancer Registry, Trenton, New Jersey, USA; Rutgers School of Public Health, Piscataway, New Jersey, USA
| | - Elizabeth L Yanik
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland, USA
| | - Charles F Lynch
- Department of Epidemiology, University of Iowa, Iowa City, Iowa, USA
| | - Bertram L Kasiske
- Department of Medicine, Hennepin County Medical Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jon Snyder
- Scientific Registry of Transplant Recipients, Minneapolis Medical Research Foundation, Minneapolis, Minnesota, USA
| | - Eric A Engels
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland, USA
| |
Collapse
|
23
|
Yanik EL, Siddiqui K, Engels EA. Sirolimus effects on cancer incidence after kidney transplantation: a meta-analysis. Cancer Med 2015; 4:1448-59. [PMID: 26108799 PMCID: PMC4567030 DOI: 10.1002/cam4.487] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 05/21/2015] [Accepted: 05/27/2015] [Indexed: 12/21/2022] Open
Abstract
Sirolimus, an immunosuppressant option for kidney transplant recipients, may reduce cancer risk by interrupting the mammalian target of rapamycin pathway. However, studies of sirolimus and cancer incidence in kidney recipients have not been definitive, and have had limited ability to examine specific cancer types. The literature was systematically reviewed to identify randomized controlled trials (RCTs) and observational studies of kidney recipients that compared sirolimus users to sirolimus nonusers. Meta-analytic methods were used to obtain pooled estimates of the association between sirolimus use and incidence of total cancer and specific cancer types. Estimates were stratified by study type (RCT vs. observational) and use of cyclosporine (an immunosuppressant that affects DNA repair). Twenty RCTs and two observational studies were eligible for meta-analysis, including 39,039 kidney recipients overall. Sirolimus use was associated with lower overall cancer incidence (incidence rate ratio [IRR] = 0.71, 95% CI = 0.56-0.90), driven by a reduction in incidence of nonmelanoma skin cancer (NMSC, IRR = 0.49, 95% CI = 0.32-0.76). The protective effect of sirolimus on NMSC risk was most notable in studies comparing sirolimus against cyclosporine (IRR = 0.19, 95% CI = 0.04-0.84). After excluding NMSCs, there was no overall association between sirolimus and incidence of other cancers (IRR = 1.06, 95% CI = 0.69-1.63). However, sirolimus use had associations with lower kidney cancer incidence (IRR = 0.40, 95% CI = 0.20-0.81), and higher prostate cancer incidence (IRR = 1.85, 95% CI = 1.17-2.91). Among kidney recipients, sirolimus users have lower NMSC risk, which may be partly due to removal of cyclosporine. Sirolimus may also reduce kidney cancer risk but did not appear protective for other cancers, and it may actually increase prostate cancer risk.
Collapse
Affiliation(s)
- Elizabeth L Yanik
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Kulsoom Siddiqui
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland.,Brown School of Social Work, Washington University in St. Louis, St. Louis, Missouri
| | - Eric A Engels
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
24
|
Shah P, He YY. Molecular regulation of UV-induced DNA repair. Photochem Photobiol 2015; 91:254-64. [PMID: 25534312 DOI: 10.1111/php.12406] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 12/09/2014] [Indexed: 12/21/2022]
Abstract
Ultraviolet (UV) radiation from sunlight is a major etiologic factor for skin cancer, the most prevalent cancer in the United States, as well as premature skin aging. In particular, UVB radiation causes formation of specific DNA damage photoproducts between pyrimidine bases. These DNA damage photoproducts are repaired by a process called nucleotide excision repair, also known as UV-induced DNA repair. When left unrepaired, UVB-induced DNA damage leads to accumulation of mutations, predisposing people to carcinogenesis as well as to premature aging. Genetic loss of nucleotide excision repair leads to severe disorders, namely, xeroderma pigmentosum (XP), trichothiodystrophy (TTD) and Cockayne syndrome (CS), which are associated with predisposition to skin carcinogenesis at a young age as well as developmental and neurological conditions. Regulation of nucleotide excision repair is an attractive avenue to preventing or reversing these detrimental consequences of impaired nucleotide excision repair. Here, we review recent studies on molecular mechanisms regulating nucleotide excision repair by extracellular cues and intracellular signaling pathways, with a special focus on the molecular regulation of individual repair factors.
Collapse
Affiliation(s)
- Palak Shah
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL
| | | |
Collapse
|
25
|
Clarke CA, Robbins HA, Tatalovich Z, Lynch CF, Pawlish KS, Finch JL, Hernandez BY, Fraumeni JF, Madeleine MM, Engels EA. Risk of merkel cell carcinoma after solid organ transplantation. J Natl Cancer Inst 2015; 107:dju382. [PMID: 25575645 DOI: 10.1093/jnci/dju382] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Solid organ transplant recipients have elevated risks of virus-related cancers, in part because of long-term immunosuppression. Merkel cell carcinoma (MCC) is an aggressive skin cancer recently found to have a viral origin, but little is known regarding the occurrence of MCC after transplant. METHODS We linked the US Scientific Registry of Transplant Recipients with data from 15 population-based cancer registries to ascertain MCC occurrence among 189498 solid organ transplant recipients from 1987 to 2009. Risks for MCC following transplantation were compared with the general population using standardized incidence ratios, and Poisson regression was used to compare incidence rates according to key patient and transplant characteristics. All statistical tests were two-sided. RESULTS After solid organ transplantation, overall risk of MCC was increased 23.8-fold (95% confidence interval = 19.6 to 28.7, n = 110). Adjusted risks were highest among older recipients, increased with time since transplantation, and varied by organ type (all P ≤ .007). Azathioprine, cyclosporine, and mTOR inhibitors given for maintenance immunosuppression increased risk, and non-Hispanic white recipients on cyclosporine and azathioprine experienced increasing MCC risk with lower latitude of residence (ie, higher ultraviolet radiation exposure, P = .012). CONCLUSIONS MCC risk is sharply elevated after solid organ transplant, likely resulting from long-term immunosuppression. Immunosuppressive medications may act synergistically with ultraviolet radiation to increase risk.
Collapse
Affiliation(s)
- Christina A Clarke
- Cancer Prevention Institute of California, Fremont, CA (CAC); Department of Health Research and Policy, Stanford University School of Medicine and Stanford Cancer Institute, Palo Alto, CA (CAC); Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD (HAR, ZT, JFFJr, EAE); Department of Epidemiology, University of Iowa, Iowa City, IA (CFL); New Jersey State Cancer Registry, New Jersey Department of Health, Trenton, NJ (KSP); Colorado Central Cancer Registry, Colorado Department of Public Health and Environment, Denver, CO (JLF); University of Hawaii Cancer Center, Honolulu, HI (BYH); Program in Epidemiology, Fred Hutchinson Cancer Research Center, Seattle, WA (MMM); Department of Epidemiology, University of Washington, Seattle, WA (MMM).
| | - Hilary A Robbins
- Cancer Prevention Institute of California, Fremont, CA (CAC); Department of Health Research and Policy, Stanford University School of Medicine and Stanford Cancer Institute, Palo Alto, CA (CAC); Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD (HAR, ZT, JFFJr, EAE); Department of Epidemiology, University of Iowa, Iowa City, IA (CFL); New Jersey State Cancer Registry, New Jersey Department of Health, Trenton, NJ (KSP); Colorado Central Cancer Registry, Colorado Department of Public Health and Environment, Denver, CO (JLF); University of Hawaii Cancer Center, Honolulu, HI (BYH); Program in Epidemiology, Fred Hutchinson Cancer Research Center, Seattle, WA (MMM); Department of Epidemiology, University of Washington, Seattle, WA (MMM)
| | - Zaria Tatalovich
- Cancer Prevention Institute of California, Fremont, CA (CAC); Department of Health Research and Policy, Stanford University School of Medicine and Stanford Cancer Institute, Palo Alto, CA (CAC); Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD (HAR, ZT, JFFJr, EAE); Department of Epidemiology, University of Iowa, Iowa City, IA (CFL); New Jersey State Cancer Registry, New Jersey Department of Health, Trenton, NJ (KSP); Colorado Central Cancer Registry, Colorado Department of Public Health and Environment, Denver, CO (JLF); University of Hawaii Cancer Center, Honolulu, HI (BYH); Program in Epidemiology, Fred Hutchinson Cancer Research Center, Seattle, WA (MMM); Department of Epidemiology, University of Washington, Seattle, WA (MMM)
| | - Charles F Lynch
- Cancer Prevention Institute of California, Fremont, CA (CAC); Department of Health Research and Policy, Stanford University School of Medicine and Stanford Cancer Institute, Palo Alto, CA (CAC); Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD (HAR, ZT, JFFJr, EAE); Department of Epidemiology, University of Iowa, Iowa City, IA (CFL); New Jersey State Cancer Registry, New Jersey Department of Health, Trenton, NJ (KSP); Colorado Central Cancer Registry, Colorado Department of Public Health and Environment, Denver, CO (JLF); University of Hawaii Cancer Center, Honolulu, HI (BYH); Program in Epidemiology, Fred Hutchinson Cancer Research Center, Seattle, WA (MMM); Department of Epidemiology, University of Washington, Seattle, WA (MMM)
| | - Karen S Pawlish
- Cancer Prevention Institute of California, Fremont, CA (CAC); Department of Health Research and Policy, Stanford University School of Medicine and Stanford Cancer Institute, Palo Alto, CA (CAC); Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD (HAR, ZT, JFFJr, EAE); Department of Epidemiology, University of Iowa, Iowa City, IA (CFL); New Jersey State Cancer Registry, New Jersey Department of Health, Trenton, NJ (KSP); Colorado Central Cancer Registry, Colorado Department of Public Health and Environment, Denver, CO (JLF); University of Hawaii Cancer Center, Honolulu, HI (BYH); Program in Epidemiology, Fred Hutchinson Cancer Research Center, Seattle, WA (MMM); Department of Epidemiology, University of Washington, Seattle, WA (MMM)
| | - Jack L Finch
- Cancer Prevention Institute of California, Fremont, CA (CAC); Department of Health Research and Policy, Stanford University School of Medicine and Stanford Cancer Institute, Palo Alto, CA (CAC); Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD (HAR, ZT, JFFJr, EAE); Department of Epidemiology, University of Iowa, Iowa City, IA (CFL); New Jersey State Cancer Registry, New Jersey Department of Health, Trenton, NJ (KSP); Colorado Central Cancer Registry, Colorado Department of Public Health and Environment, Denver, CO (JLF); University of Hawaii Cancer Center, Honolulu, HI (BYH); Program in Epidemiology, Fred Hutchinson Cancer Research Center, Seattle, WA (MMM); Department of Epidemiology, University of Washington, Seattle, WA (MMM)
| | - Brenda Y Hernandez
- Cancer Prevention Institute of California, Fremont, CA (CAC); Department of Health Research and Policy, Stanford University School of Medicine and Stanford Cancer Institute, Palo Alto, CA (CAC); Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD (HAR, ZT, JFFJr, EAE); Department of Epidemiology, University of Iowa, Iowa City, IA (CFL); New Jersey State Cancer Registry, New Jersey Department of Health, Trenton, NJ (KSP); Colorado Central Cancer Registry, Colorado Department of Public Health and Environment, Denver, CO (JLF); University of Hawaii Cancer Center, Honolulu, HI (BYH); Program in Epidemiology, Fred Hutchinson Cancer Research Center, Seattle, WA (MMM); Department of Epidemiology, University of Washington, Seattle, WA (MMM)
| | - Joseph F Fraumeni
- Cancer Prevention Institute of California, Fremont, CA (CAC); Department of Health Research and Policy, Stanford University School of Medicine and Stanford Cancer Institute, Palo Alto, CA (CAC); Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD (HAR, ZT, JFFJr, EAE); Department of Epidemiology, University of Iowa, Iowa City, IA (CFL); New Jersey State Cancer Registry, New Jersey Department of Health, Trenton, NJ (KSP); Colorado Central Cancer Registry, Colorado Department of Public Health and Environment, Denver, CO (JLF); University of Hawaii Cancer Center, Honolulu, HI (BYH); Program in Epidemiology, Fred Hutchinson Cancer Research Center, Seattle, WA (MMM); Department of Epidemiology, University of Washington, Seattle, WA (MMM)
| | - Margaret M Madeleine
- Cancer Prevention Institute of California, Fremont, CA (CAC); Department of Health Research and Policy, Stanford University School of Medicine and Stanford Cancer Institute, Palo Alto, CA (CAC); Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD (HAR, ZT, JFFJr, EAE); Department of Epidemiology, University of Iowa, Iowa City, IA (CFL); New Jersey State Cancer Registry, New Jersey Department of Health, Trenton, NJ (KSP); Colorado Central Cancer Registry, Colorado Department of Public Health and Environment, Denver, CO (JLF); University of Hawaii Cancer Center, Honolulu, HI (BYH); Program in Epidemiology, Fred Hutchinson Cancer Research Center, Seattle, WA (MMM); Department of Epidemiology, University of Washington, Seattle, WA (MMM)
| | - Eric A Engels
- Cancer Prevention Institute of California, Fremont, CA (CAC); Department of Health Research and Policy, Stanford University School of Medicine and Stanford Cancer Institute, Palo Alto, CA (CAC); Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD (HAR, ZT, JFFJr, EAE); Department of Epidemiology, University of Iowa, Iowa City, IA (CFL); New Jersey State Cancer Registry, New Jersey Department of Health, Trenton, NJ (KSP); Colorado Central Cancer Registry, Colorado Department of Public Health and Environment, Denver, CO (JLF); University of Hawaii Cancer Center, Honolulu, HI (BYH); Program in Epidemiology, Fred Hutchinson Cancer Research Center, Seattle, WA (MMM); Department of Epidemiology, University of Washington, Seattle, WA (MMM)
| |
Collapse
|
26
|
Qiu J, Wang X, Meng X, Zheng Y, Li G, Ma J, Ye G, Li Y, Li J. Attenuated NER expressions of XPF and XPC associated with smoking are involved in the recurrence of bladder cancer. PLoS One 2014; 9:e115224. [PMID: 25535740 PMCID: PMC4275238 DOI: 10.1371/journal.pone.0115224] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 11/20/2014] [Indexed: 01/01/2023] Open
Abstract
The varied NER genes and smoking are two important risk factors of bladder cancer, but the mechanism of the NER protein and smoking in cancer progression, however, remains unclear. In this report, we compared the expressions of NER genes in 79 bladder cancer tissues with or without any recurrence by real-time PCR and then analyzed the varied NER genes by immunochemistry in 219 bladder cancer tissue samples. Based on the clinical data, we analyzed the clinical value of varied NER genes and smoking in 219 bladder cancers by the Kaplan-Meier method and Cox proportional hazards regression. We found the expressions of the NER gene XPF and XPC were significantly lower in bladder cancer tissues with a recurrence compared with those without a recurrence at mRNA level. Also, the patients with the XPF and XPC defect had a statistically significant lower median recurrence-free survival time than those without the XPF and XPC defect, and smoking can make this difference more remarkable. Our results suggest that XPF and XPC expression may be a potential predictive factor for bladder cancer, and smoking can not only influence the recurrence of bladder cancer as a single factor but also aggravate the results of the XPF defect and XPC defect.
Collapse
Affiliation(s)
- Jianhong Qiu
- Department of Urology, Bethune International Peace Hospital, Shijiazhuang, China
| | - Xiangwei Wang
- Department of Urology, The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China
| | - Xiaodong Meng
- Department of Urology, Bethune International Peace Hospital, Shijiazhuang, China
| | - Yan Zheng
- Department of Urology, Bethune International Peace Hospital, Shijiazhuang, China
| | - Gang Li
- Department of Urology, Bethune International Peace Hospital, Shijiazhuang, China
| | - Jiyao Ma
- Department of Urology, Bethune International Peace Hospital, Shijiazhuang, China
| | - Gang Ye
- Department of Urology, The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China
| | - Yong Li
- Department of Urology, Bethune International Peace Hospital, Shijiazhuang, China
| | - Jie Li
- Department of Urology, Bethune International Peace Hospital, Shijiazhuang, China
- * E-mail:
| |
Collapse
|
27
|
Kim I, He YY. Ultraviolet radiation-induced non-melanoma skin cancer: Regulation of DNA damage repair and inflammation. Genes Dis 2014; 1:188-198. [PMID: 25642450 PMCID: PMC4307792 DOI: 10.1016/j.gendis.2014.08.005] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 08/22/2014] [Indexed: 01/01/2023] Open
Abstract
Exposure to ultraviolet (UV) radiation is associated with approximately 65% of melanoma cases, and 90% of non-melanoma skin cancers (NMSC), including basal cell carcinoma (BCC) and squamous cell carcinoma (SCC). While the incidence of most other malignancies has either stabilized or declined, that of NMSC has increased and is developing even in younger age groups. NMSCs account for nearly 15,000 deaths, 3.5 million new cases, and more than 3 billion dollars a year in medical costs in the United States alone, representing a major public health concern. As sun protection efforts have not been proven effective, targeted chemoprevention strategies are much needed. Skin carcinogenesis by DNA damage is considered a predominant paradigm for UV toxicity. Exposure to UV radiation can activate various oncogenes while inactivating tumor suppressor genes, resulting in inappropriate survival and proliferation of keratinocytes that harbor these damages. Moreover, increasing evidence demonstrate that inflammatory responses by the immune cells within the tumor microenvironment also contribute significantly to skin tumorigenesis. Initiation and progression of skin carcinogenesis mediated by UV radiation involve complex pathways, including those of apoptosis, proliferation, autophagy, DNA repair, checkpoint signaling, metabolism, and inflammation. In this review, we highlight the recent advances in two of these key molecular processes that result in UV-mediated skin carcinogenesis. In particular, we discuss 1) pathways that regulate DNA damage repair and 2) the regulation of the inflammatory process its crosstalk with DNA repair potentially leading to non-melanoma skin carcinogenesis.
Collapse
Affiliation(s)
- InYoung Kim
- Pritzker School of Medicine, University of Chicago, Chicago, IL, USA
| | - Yu-Ying He
- Section of Dermatology, Department of Medicine, University of Chicago, Chicago, IL, USA
| |
Collapse
|
28
|
Feng MX, Li JP, Wang H, Shen YX, Tan LJ, Wang Q, Wang YQ. Clinical significance of cyclophilin a expression in esophageal squamous cell carcinoma. Genet Test Mol Biomarkers 2014; 19:182-6. [PMID: 25420107 DOI: 10.1089/gtmb.2014.0067] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE To explore the expression of cyclophilin A (CypA) in esophageal tissues and its clinical significance. METHOD Expression of CypA was detected in 236 esophageal cancer tissues and 236 normal tissues by using an immunohistochemical method, and the relationship between CypA expression and clinical outcomes was observed. RESULTS There were 166 patients with high expression of CypA (70.23%) and a higher expression in 69.3% of males and 73.3% in females. The CypA expression was irrelevant to age, tumor location, lymph node metastasis, and tumor differentiation degree. The Kaplan-Meier survival curve analysis showed that the expression of CypA was associated with the prognosis of patients with esophageal squamous cell carcinoma. CONCLUSION The poor prognosis of esophageal cancer patients was associated with high expression of CypA.
Collapse
Affiliation(s)
- Ming-Xiang Feng
- 1 Department of Thoracic Surgery, Zhongshan Hospital, Fudan University , Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
29
|
Ming M, Zhao B, Qiang L, He YY. Effect of immunosuppressants tacrolimus and mycophenolate mofetil on the keratinocyte UVB response. Photochem Photobiol 2014; 91:242-7. [PMID: 25039758 DOI: 10.1111/php.12318] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 07/11/2014] [Indexed: 01/10/2023]
Abstract
Nonmelanoma skin cancer, derived from epidermal keratinocytes, is the most common malignancy in organ transplant recipients, causes serious morbidity and mortality, and is strongly associated with solar ultraviolet (UV) exposure. Preventing and treating skin cancer in these individuals has been extraordinarily challenging. Following organ transplantation, the immunosuppressants are used to prevent graft rejection. Until now, immunosuppression has been assumed to be the major factor leading to skin cancer in this setting. However, the mechanism of skin carcinogenesis in organ transplant recipients has not been understood to date; specifically, it remains unknown whether these cancers are immunosuppression-dependent or -independent. In particular, it remains poorly understood what is the mechanistic carcinogenic action of the newer generation of immunosuppressants including tacrolimus and mycophenolate mofetil (MMF). Here, we show that tacrolimus and MMF impairs UVB-induced DNA damage repair and apoptosis in human epidermal keratinocytes. In addition, tacrolimus inhibits UVB-induced checkpoint signaling. However, MMF had no effect. Our findings have demonstrated that tacrolimus and MMF compromises proper UVB response in keratinocytes, suggesting an immunosuppression-independent mechanism in the tumor-promoting action of these immunosuppressants.
Collapse
Affiliation(s)
- Mei Ming
- Section of Dermatology, Department of Medicine, University of Chicago, Chicago, IL
| | | | | | | |
Collapse
|
30
|
Wheless L, Jacks S, Mooneyham Potter KA, Leach BC, Cook J. Skin cancer in organ transplant recipients: more than the immune system. J Am Acad Dermatol 2014; 71:359-65. [PMID: 24725477 DOI: 10.1016/j.jaad.2014.02.039] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 02/08/2014] [Accepted: 02/11/2014] [Indexed: 02/08/2023]
Abstract
Organ transplant recipients (OTRs) are at increased risk of developing nonmelanoma skin cancers. This has long been thought to be caused by immunosuppression and viral infection. However, skin cancer risk among individuals with AIDS or iatrogenic immunodeficiency does not approach the levels seen in OTRs, suggesting other factors play a critical role in oncogenesis. In clinical trials of OTRs, switching from calcineurin inhibitors to mammalian target of rapamycin inhibitors consistently led to a significant reduction in the risk of developing new skin cancers. New evidence suggests calcineurin inhibitors interfere with p53 signaling and nucleotide excision repair. These two pathways are associated with nonmelanoma skin cancer, and squamous cell carcinoma in particular. This finding may help explain the predominance of squamous cell carcinoma over basal cell carcinoma in this population. Mammalian target of rapamycin inhibitors do not appear to impact these pathways. Immunosuppression, viral infection, and impaired DNA repair and p53 signaling all interact in OTRs to create a phenotype of extreme risk for nonmelanoma skin cancer.
Collapse
Affiliation(s)
- Lee Wheless
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, South Carolina; Department of Medicine, Medical University of South Carolina, Charleston, South Carolina; Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Sarah Jacks
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina; Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Kathryn Anne Mooneyham Potter
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina; Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Brian C Leach
- Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, South Carolina.
| | - Joel Cook
- Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
31
|
Hepatocellular carcinoma and other malignancies in autoimmune hepatitis. Dig Dis Sci 2013; 58:1459-76. [PMID: 23306849 DOI: 10.1007/s10620-012-2525-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 12/03/2012] [Indexed: 12/13/2022]
Abstract
Hepatocellular carcinoma and extrahepatic malignancies can complicate the course of autoimmune hepatitis, and these occurrences may increase in frequency as the survival of patients with cirrhosis is extended and the prospect of new nonstandard immune-modifying intervention is realized. The frequency of hepatocellular carcinoma in patients with autoimmune hepatitis and cirrhosis is 1-9 %, and annual occurrence in patients with cirrhosis is 1.1-1.9 %. The standardized incidence ratio for hepatocellular carcinoma in autoimmune hepatitis is 23.3 (95 % confidence interval (CI) 7.5-54.3) in Sweden, and the standardized mortality ratio for hepatobiliary cancer is 42.3 (95 % CI 20.3-77.9) in New Zealand. The principal risk factor is long-standing cirrhosis, and patients at risk are characterized mainly by cirrhosis for ≥ 10 years, manifestations of portal hypertension, persistent liver inflammation, and immunosuppressive therapy for ≥ 3 years. Multiple molecular disturbances, including the accumulation of senescent hepatocytes because of telomere shortening, step-wise accumulation of chromosomal injuries, and aberrations in transcription factors and genes, may contribute to the risk. Extraheptic malignancies of diverse cell types occur in 5 % in an unpredictable fashion. The standardized incidence ratio is 2.7 (95 % CI 1.8-3.9) in New Zealand, and non-melanoma skin cancers are most common. Outcomes are related to the nature and stage of the tumor at diagnosis. Surveillance recommendations have not been promulgated, but hepatic ultrasonography every six months in patients with cirrhosis is a consideration. Routine health screening measures for other malignancies should be applied diligently.
Collapse
|