1
|
Kabatnik S, Post F, Drici L, Bartels AS, Strauss MT, Zheng X, Madsen GI, Mund A, Rosenberger FA, Moreira J, Mann M. Spatial characterization and stratification of colorectal adenomas by deep visual proteomics. iScience 2024; 27:110620. [PMID: 39252972 PMCID: PMC11381895 DOI: 10.1016/j.isci.2024.110620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 05/13/2024] [Accepted: 07/26/2024] [Indexed: 09/11/2024] Open
Abstract
Colorectal adenomas (CRAs) are potential precursor lesions to adenocarcinomas, currently classified by morphological features. We aimed to establish a molecular feature-based risk allocation framework toward improved patient stratification. Deep visual proteomics (DVP) is an approach that combines image-based artificial intelligence with automated microdissection and ultra-high sensitive mass spectrometry. Here, we used DVP on formalin-fixed, paraffin-embedded (FFPE) CRA tissues from nine male patients, immunohistologically stained for caudal-type homeobox 2 (CDX2), a protein implicated in colorectal cancer, enabling the characterization of cellular heterogeneity within distinct tissue regions and across patients. DVP identified DMBT1, MARCKS, and CD99 as protein markers linked to recurrence, suggesting their potential for risk assessment. It also detected a metabolic shift to anaerobic glycolysis in cells with high CDX2 expression. Our findings underscore the potential of spatial proteomics to refine early stage detection and contribute to personalized patient management strategies and provided novel insights into metabolic reprogramming.
Collapse
Affiliation(s)
- Sonja Kabatnik
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Science, University of Copenhagen, Copenhagen, Denmark
| | - Frederik Post
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Science, University of Copenhagen, Copenhagen, Denmark
| | - Lylia Drici
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Science, University of Copenhagen, Copenhagen, Denmark
| | - Annette Snejbjerg Bartels
- Precision Cancer Medicine Laboratory, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Maximilian T Strauss
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Science, University of Copenhagen, Copenhagen, Denmark
| | - Xiang Zheng
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Science, University of Copenhagen, Copenhagen, Denmark
| | - Gunvor I Madsen
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Andreas Mund
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Science, University of Copenhagen, Copenhagen, Denmark
| | - Florian A Rosenberger
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - José Moreira
- Precision Cancer Medicine Laboratory, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Matthias Mann
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health Science, University of Copenhagen, Copenhagen, Denmark
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| |
Collapse
|
2
|
Keggenhoff FL, Castven D, Becker D, Stojkovic S, Castven J, Zimpel C, Straub BK, Gerber T, Langer H, Hähnel P, Kindler T, Fahrer J, O'Rourke CJ, Ehmer U, Saborowski A, Ma L, Wang XW, Gaiser T, Matter MS, Sina C, Derer S, Lee JS, Roessler S, Kaina B, Andersen JB, Galle PR, Marquardt JU. PARP-1 selectively impairs KRAS-driven phenotypic and molecular features in intrahepatic cholangiocarcinoma. Gut 2024; 73:1712-1724. [PMID: 38857989 PMCID: PMC11420749 DOI: 10.1136/gutjnl-2023-331237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 05/24/2024] [Indexed: 06/12/2024]
Abstract
OBJECTIVE Intrahepatic cholangiocarcinoma (iCCA) is the second most common primary liver cancer with limited therapeutic options. KRAS mutations are among the most abundant genetic alterations in iCCA associated with poor clinical outcome and treatment response. Recent findings indicate that Poly(ADP-ribose)polymerase1 (PARP-1) is implicated in KRAS-driven cancers, but its exact role in cholangiocarcinogenesis remains undefined. DESIGN PARP-1 inhibition was performed in patient-derived and established iCCA cells using RNAi, CRISPR/Cas9 and pharmacological inhibition in KRAS-mutant, non-mutant cells. In addition, Parp-1 knockout mice were combined with iCCA induction by hydrodynamic tail vein injection to evaluate an impact on phenotypic and molecular features of Kras-driven and Kras-wildtype iCCA. Clinical implications were confirmed in authentic human iCCA. RESULTS PARP-1 was significantly enhanced in KRAS-mutant human iCCA. PARP-1-based interventions preferentially impaired cell viability and tumourigenicity in human KRAS-mutant cell lines. Consistently, loss of Parp-1 provoked distinct phenotype in Kras/Tp53-induced versus Akt/Nicd-induced iCCA and abolished Kras-dependent cholangiocarcinogenesis. Transcriptome analyses confirmed preferential impairment of DNA damage response pathways and replicative stress response mediated by CHK1. Consistently, inhibition of CHK1 effectively reversed PARP-1 mediated effects. Finally, Parp-1 depletion induced molecular switch of KRAS-mutant iCCA recapitulating good prognostic human iCCA patients. CONCLUSION Our findings identify the novel prognostic and therapeutic role of PARP-1 in iCCA patients with activation of oncogenic KRAS signalling.
Collapse
Affiliation(s)
- Friederike L Keggenhoff
- First Department of Internal Medicine, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Darko Castven
- Department of Medicine I, University Medical Center Schleswig Holstein Campus Lübeck, Lübeck, Germany
| | - Diana Becker
- First Department of Internal Medicine, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Stojan Stojkovic
- Department of Medicine I, University Medical Center Schleswig Holstein Campus Lübeck, Lübeck, Germany
| | - Jovana Castven
- Department of Medicine I, University Medical Center Schleswig Holstein Campus Lübeck, Lübeck, Germany
| | - Carolin Zimpel
- Department of Medicine I, University Medical Center Schleswig Holstein Campus Lübeck, Lübeck, Germany
| | - Beate K Straub
- Institute of Pathology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
- Tissue Biobank of the University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Tiemo Gerber
- Institute of Pathology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Harald Langer
- Cardiology Angiology, University Medical Centre, Mannheim, Germany
| | - Patricia Hähnel
- Department of Hematology, Medical Oncology and Pneumology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Thomas Kindler
- Department of Hematology, Medical Oncology and Pneumology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Jörg Fahrer
- Department of Chemistry, RPTU Kaiserslautern-Landau, Kaiserslautern, Germany
| | - Colm J O'Rourke
- Department of Health and Medical Sciences, University of Copenhagen Biotech Research & Innovation Centre, Kobenhavn, Denmark
| | - Ursula Ehmer
- Department of Internal Medicine II, Klinikum rechts der Isar der Technischen Universitat, München, Germany
| | - Anna Saborowski
- Department of Gastroenterology, Hepatology, and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Lichun Ma
- Laboratory of Human Carcinogenesis, National Cancer Institute, Center for Cancer Research, Bethesda, Maryland, USA
| | - Xin Wei Wang
- Laboratory of Human Carcinogenesis, National Cancer Institute, Center for Cancer Research, Bethesda, Maryland, USA
- Liver Cancer Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Timo Gaiser
- Institute of Pathology, University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Matthias S Matter
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Christian Sina
- Institute of Nutritional Medicine, University Medical Center Schleswig Holstein Campus Lübeck, Lübeck, Germany
| | - Stefanie Derer
- Institute of Nutritional Medicine, University Medical Center Schleswig Holstein Campus Lübeck, Lübeck, Germany
| | - Ju-Seog Lee
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Stephanie Roessler
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Bernd Kaina
- Department of Toxicology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Jesper B Andersen
- Department of Health and Medical Sciences, University of Copenhagen Biotech Research & Innovation Centre, Kobenhavn, Denmark
| | - Peter R Galle
- First Department of Internal Medicine, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Jens U Marquardt
- Department of Medicine I, University Medical Center Schleswig Holstein Campus Lübeck, Lübeck, Germany
| |
Collapse
|
3
|
Albaqami WF, Alshamrani AA, Almubarak AA, Alotaibi FE, Alotaibi BJ, Alanazi AM, Alotaibi MR, Alhoshani A, As Sobeai HM. Genetic and Epigenetic Biomarkers Associated with Early Relapse in Pediatric Acute Lymphoblastic Leukemia: A Focused Bioinformatics Study on DNA-Repair Genes. Biomedicines 2024; 12:1766. [PMID: 39200230 PMCID: PMC11351110 DOI: 10.3390/biomedicines12081766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 07/28/2024] [Accepted: 08/01/2024] [Indexed: 09/02/2024] Open
Abstract
Genomic instability is one of the main drivers of tumorigenesis and the development of hematological malignancies. Cancer cells can remedy chemotherapeutic-induced DNA damage by upregulating DNA-repair genes and ultimately inducing therapy resistance. Nevertheless, the association between the DNA-repair genes, drug resistance, and disease relapse has not been well characterized in acute lymphoblastic leukemia (ALL). This study aimed to explore the role of the DNA-repair machinery and the molecular mechanisms by which it is regulated in early- and late-relapsing pediatric ALL patients. We performed secondary data analysis on the Therapeutically Applicable Research to Generate Effective Treatments (TARGET)-ALL expansion phase II trial of 198 relapsed pediatric precursor B-cell ALL. Comprehensive genetic and epigenetic investigations of 147 DNA-repair genes were conducted in the study. Gene expression was assessed using Microarray and RNA-sequencing platforms. Genomic alternations, methylation status, and miRNA transcriptome were investigated for the candidate DNA-repair genes. We identified three DNA-repair genes, ALKBH3, NHEJ1, and PARP1, that were upregulated in early relapsers compared to late relapsers (p < 0.05). Such upregulation at diagnosis was significantly associated with disease-free survival and overall survival in precursor-B-ALL (p < 0.05). Moreover, PARP1 upregulation accompanied a significant downregulation of its targeting miRNA, miR-1301-3p (p = 0.0152), which was strongly linked with poorer disease-free and overall survivals. Upregulation of DNA-repair genes, PARP1 in particular, increases the likelihood of early relapse of precursor-B-ALL in children. The observation that PARP1 was upregulated in early relapsers relative to late relapsers might serve as a valid rationale for proposing alternative treatment approaches, such as using PARP inhibitors with chemotherapy.
Collapse
Affiliation(s)
- Walaa F. Albaqami
- Department of Science, Prince Sultan Military College of Health Sciences, Dhahran 31932, Saudi Arabia;
| | - Ali A. Alshamrani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.A.A.); (F.E.A.); (B.J.A.); (A.M.A.); (M.R.A.); (A.A.)
| | - Ali A. Almubarak
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.A.A.); (F.E.A.); (B.J.A.); (A.M.A.); (M.R.A.); (A.A.)
| | - Faris E. Alotaibi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.A.A.); (F.E.A.); (B.J.A.); (A.M.A.); (M.R.A.); (A.A.)
| | - Basil Jamal Alotaibi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.A.A.); (F.E.A.); (B.J.A.); (A.M.A.); (M.R.A.); (A.A.)
| | - Abdulrahman M. Alanazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.A.A.); (F.E.A.); (B.J.A.); (A.M.A.); (M.R.A.); (A.A.)
- Pharmaceutical Care Division, King Faisal Specialist Hospital & Research Centre, Madinah 42523, Saudi Arabia
| | - Moureq R. Alotaibi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.A.A.); (F.E.A.); (B.J.A.); (A.M.A.); (M.R.A.); (A.A.)
| | - Ali Alhoshani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.A.A.); (F.E.A.); (B.J.A.); (A.M.A.); (M.R.A.); (A.A.)
| | - Homood M. As Sobeai
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.A.A.); (F.E.A.); (B.J.A.); (A.M.A.); (M.R.A.); (A.A.)
| |
Collapse
|
4
|
Singla S, Jena G. Studies on the mechanism of local and extra-intestinal tissue manifestations in AOM-DSS-induced carcinogenesis in BALB/c mice: role of PARP-1, NLRP3, and autophagy. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:4321-4337. [PMID: 38091080 DOI: 10.1007/s00210-023-02878-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/28/2023] [Indexed: 05/23/2024]
Abstract
Colitis-associated colorectal cancer (CACC) is one of the devastating complications of long-term inflammatory bowel disease and is associated with substantial morbidity and mortality. Combination of azoxymethane (AOM) and dextran sulfate sodium (DSS) has been extensively used for inflammation-mediated colon tumor development due to its reproducibility, potency, histological and molecular changes, and resemblance to human CACC. In the tumor microenvironment and extra-intestinal tissues, PARP-1, NLRP3 inflammasome, and autophagy's biological functions are complicated and encompass intricate interactions between these molecular components. The focus of the present investigation is to determine the colonic and extra-intestinal tissue damage induced by AOM-DSS and related molecular mechanisms. Azoxymethane (10 mg/kg, i.p.; single injection) followed by DSS (3 cycles, 7 days per cycle) over a period of 10 weeks induced colitis-associated colon cancer in male BALB/c mice. By initiating carcinogenesis with a single injection of azoxymethane (AOM) and then establishing inflammation with dextran sulfate sodium (DSS), a two-stage murine model for CACC was developed. Biochemical parameters, ELISA, histopathological and immunohistochemical analysis, and western blotting have been performed to evaluate the colonic, hepatic, testicular and pancreatic damage. In addition, the AOM/DSS-induced damage has been assessed by analyzing the expression of a variety of molecular targets, including proliferating cell nuclear antigen (PCNA), interleukin-10 (IL-10), AMP-activated protein kinase (AMPK), poly (ADP-ribose) polymerase-1 (PARP-1), cysteine-associated protein kinase-1 (caspase-1), NLR family pyrin domain containing 3 (NLRP3), beclin-1, and interleukin-1β (IL-1β). Present findings revealed that AOM/DSS developed tumors in colon tissue followed by extra-intestinal hepatic, testicular, and pancreatic damages.
Collapse
Affiliation(s)
- Shivani Singla
- Facility for Risk Assessment and Intervention Studies, Dept. of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S, Nagar, Punjab, 160062, India
| | - Gopabandhu Jena
- Facility for Risk Assessment and Intervention Studies, Dept. of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S, Nagar, Punjab, 160062, India.
| |
Collapse
|
5
|
Marcazzan S, Braz Carvalho MJ, Nguyen NT, Strangmann J, Slotta-Huspenina J, Tenditnaya A, Tschurtschenthaler M, Rieder J, Proaño-Vasco A, Ntziachristos V, Steiger K, Gorpas D, Quante M, Kossatz S. PARP1-targeted fluorescence molecular endoscopy as novel tool for early detection of esophageal dysplasia and adenocarcinoma. J Exp Clin Cancer Res 2024; 43:53. [PMID: 38383387 PMCID: PMC10880256 DOI: 10.1186/s13046-024-02963-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/22/2024] [Indexed: 02/23/2024] Open
Abstract
BACKGROUND Esophageal cancer is one of the 10 most common cancers worldwide and its incidence is dramatically increasing. Despite some improvements, the current surveillance protocol with white light endoscopy and random untargeted biopsies collection (Seattle protocol) fails to diagnose dysplastic and cancerous lesions in up to 50% of patients. Therefore, new endoscopic imaging technologies in combination with tumor-specific molecular probes are needed to improve early detection. Herein, we investigated the use of the fluorescent Poly (ADP-ribose) Polymerase 1 (PARP1)-inhibitor PARPi-FL for early detection of dysplastic lesions in patient-derived organoids and transgenic mouse models, which closely mimic the transformation from non-malignant Barrett's Esophagus (BE) to invasive esophageal adenocarcinoma (EAC). METHODS We determined PARP1 expression via immunohistochemistry (IHC) in human biospecimens and mouse tissues. We also assessed PARPi-FL uptake in patient- and mouse-derived organoids. Following intravenous injection of 75 nmol PARPi-FL/mouse in L2-IL1B (n = 4) and L2-IL1B/IL8Tg mice (n = 12), we conducted fluorescence molecular endoscopy (FME) and/or imaged whole excised stomachs to assess PARPi-FL accumulation in dysplastic lesions. L2-IL1B/IL8Tg mice (n = 3) and wild-type (WT) mice (n = 2) without PARPi-FL injection served as controls. The imaging results were validated by confocal microscopy and IHC of excised tissues. RESULTS IHC on patient and murine tissue revealed similar patterns of increasing PARP1 expression in presence of dysplasia and cancer. In human and murine organoids, PARPi-FL localized to PARP1-expressing epithelial cell nuclei after 10 min of incubation. Injection of PARPi-FL in transgenic mouse models of BE resulted in the successful detection of lesions via FME, with a mean target-to-background ratio > 2 independently from the disease stage. The localization of PARPi-FL in the lesions was confirmed by imaging of the excised stomachs and confocal microscopy. Without PARPi-FL injection, identification of lesions via FME in transgenic mice was not possible. CONCLUSION PARPi-FL imaging is a promising approach for clinically needed improved detection of dysplastic and malignant EAC lesions in patients with BE. Since PARPi-FL is currently evaluated in a phase 2 clinical trial for oral cancer detection after topical application, clinical translation for early detection of dysplasia and EAC in BE patients via FME screening appears feasible.
Collapse
Affiliation(s)
- Sabrina Marcazzan
- II. Medizinische Klinik, TUM School of Medicine and Health, Klinikum Rechts der Isar at Technical University of Munich, Munich, 81675, Germany
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, 85764 Neuherberg, Germany and Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), TUM School of Medicine and Health, Technical University of Munich, Munich, 81675, Germany
- Clinical Radiology, Medical School OWL, Bielefeld University, Bielefeld, 33615, Germany
| | - Marcos J Braz Carvalho
- II. Medizinische Klinik, TUM School of Medicine and Health, Klinikum Rechts der Isar at Technical University of Munich, Munich, 81675, Germany
| | - Nghia T Nguyen
- Department of Nuclear Medicine, TUM School of Medicine and Health, Klinikum Rechts der Isar at Technical University of Munich, Munich, 81675, Germany
- Central Institute for Translational Cancer Research (TranslaTUM), TUM School of Medicine and Health, Technical University of Munich, Munich, 81675, Germany
| | - Julia Strangmann
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, 79106, Germany
| | - Julia Slotta-Huspenina
- Institute of Pathology, TUM School of Medicine and Health, Technical University of Munich, Munich, 81675, Germany
| | - Anna Tenditnaya
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, 85764 Neuherberg, Germany and Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), TUM School of Medicine and Health, Technical University of Munich, Munich, 81675, Germany
| | - Markus Tschurtschenthaler
- Central Institute for Translational Cancer Research (TranslaTUM), TUM School of Medicine and Health, Technical University of Munich, Munich, 81675, Germany
- Division of Translational Cancer Research, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, 69120, Germany
- Chair of Translational Cancer Research and Institute of Experimental Cancer Therapy, TUM School of Medicine and Health, Klinikum rechts der Isar at Technical University of Munich, Munich, 81675, Germany
| | - Jonas Rieder
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, 79106, Germany
| | - Andrea Proaño-Vasco
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, 79106, Germany
| | - Vasilis Ntziachristos
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, 85764 Neuherberg, Germany and Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), TUM School of Medicine and Health, Technical University of Munich, Munich, 81675, Germany
| | - Katja Steiger
- Institute of Pathology, TUM School of Medicine and Health, Technical University of Munich, Munich, 81675, Germany
- Comparative Experimental Pathology (CEP) and IBioTUM tissue biobank, TUM School of Medicine and Health, Technical University of Munich, München, 81675, Germany
| | - Dimitris Gorpas
- Institute of Biological and Medical Imaging, Helmholtz Zentrum München, 85764 Neuherberg, Germany and Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), TUM School of Medicine and Health, Technical University of Munich, Munich, 81675, Germany
| | - Michael Quante
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, 79106, Germany.
| | - Susanne Kossatz
- Department of Nuclear Medicine, TUM School of Medicine and Health, Klinikum Rechts der Isar at Technical University of Munich, Munich, 81675, Germany.
- Central Institute for Translational Cancer Research (TranslaTUM), TUM School of Medicine and Health, Technical University of Munich, Munich, 81675, Germany.
- Department of Chemistry, TUM School of Natural Sciences, Technical University of Munich, Munich, 85748, Germany.
| |
Collapse
|
6
|
PARP-1 Expression Influences Cancer Stem Cell Phenotype in Colorectal Cancer Depending on p53. Int J Mol Sci 2023; 24:ijms24054787. [PMID: 36902215 PMCID: PMC10002521 DOI: 10.3390/ijms24054787] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/23/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
Poly(ADP-ribose) polymerase-1 (PARP-1) is a protein involved in multiple physiological processes. Elevated PARP-1 expression has been found in several tumours, being associated with stemness and tumorigenesis. In colorectal cancer (CRC), some controversy among studies has been described. In this study, we analysed the expression of PARP-1 and cancer stem cell (CSC) markers in CRC patients with different p53 status. In addition, we used an in vitro model to evaluate the influence of PARP-1 in CSC phenotype regarding p53. In CRC patients, PARP-1 expression correlated with the differentiation grade, but this association was only maintained for tumours harbouring wild-type p53. Additionally, in those tumours, PARP-1 and CSC markers were positively correlated. In mutated p53 tumours, no associations were found, but PARP-1 was an independent factor for survival. According to our in vitro model, PARP-1 regulates CSC phenotype depending on p53 status. PARP-1 overexpression in a wild type p53 context increases CSC markers and sphere forming ability. By contrast, those features were reduced in mutated p53 cells. These results could implicate that patients with elevated PARP-1 expression and wild type p53 could benefit from PARP-1 inhibition therapies, meanwhile it could have adverse effects for those carrying mutated p53 tumours.
Collapse
|
7
|
Haq MFU, Hussain MZ, Mahjabeen I, Akram Z, Saeed N, Shafique R, Abbasi SF, Kayani MA. Oncometabolic role of mitochondrial sirtuins in glioma patients. PLoS One 2023; 18:e0281840. [PMID: 36809279 PMCID: PMC9943017 DOI: 10.1371/journal.pone.0281840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 01/31/2023] [Indexed: 02/23/2023] Open
Abstract
Mitochondrial sirtuins have diverse role specifically in aging, metabolism and cancer. In cancer, these sirtuins play dichotomous role as tumor suppressor and promoter. Previous studies have reported the involvement of sirtuins in different cancers. However, till now no study has been published with respect to mitochondrial sirtuins and glioma risks. Present study was purposed to figure out the expression level of mitochondrial sirtuins (SIRT3, SIRT4, SIRT5) and related genes (GDH, OGG1-2α, SOD1, SOD2, HIF1α and PARP1) in 153 glioma tissue samples and 200 brain tissue samples from epilepsy patients (taken as controls). To understand the role of selected situins in gliomagenesis, DNA damage was measured using the comet assay and oncometabolic role (oxidative stress level, ATP level and NAD level) was measured using the ELISA and quantitative PCR. Results analysis showed significant down-regulation of SIRT4 (p = 0.0337), SIRT5 (p<0.0001), GDH (p = 0.0305), OGG1-2α (p = 0.0001), SOD1 (p<0.0001) and SOD2 (p<0.0001) in glioma patients compared to controls. In case of SIRT3 (p = 0.0322), HIF1α (p = 0.0385) and PARP1 (p = 0.0203), significant up-regulation was observed. ROC curve analysis and cox regression analysis showed the good diagnostic and prognostic value of mitochondrial sirtuins in glioma patients. Oncometabolic rate assessment analysis showed significant increased ATP level (p<0.0001), NAD+ level [(NMNAT1 (p<0.0001), NMNAT3 (p<0.0001) and NAMPT (p<0.04)] and glutathione level (p<0.0001) in glioma patients compared to controls. Significant increased level of damage ((p<0.04) and decrease level of antioxidant enzymes include superoxide dismutase (SOD, p<0.0001), catalase (CAT, p<0.0001) and glutathione peroxidase (GPx, p<0.0001) was observed in patients compared to controls. Present study data suggest that variation in expression pattern of mitochondrial sirtuins and increased metabolic rate may have diagnostic and prognostic significance in glioma patients.
Collapse
Affiliation(s)
- Maria Fazal Ul Haq
- Cancer Genetics and Epigenetics Research Group, Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | | | - Ishrat Mahjabeen
- Cancer Genetics and Epigenetics Research Group, Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
- * E-mail:
| | - Zertashia Akram
- Cancer Genetics and Epigenetics Research Group, Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Nadia Saeed
- Cancer Genetics and Epigenetics Research Group, Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Rabia Shafique
- Cancer Genetics and Epigenetics Research Group, Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Sumaira Fida Abbasi
- Cancer Genetics and Epigenetics Research Group, Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Mahmood Akhtar Kayani
- Cancer Genetics and Epigenetics Research Group, Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| |
Collapse
|
8
|
Abuzenadah AM, Al-Sayes F, Mahafujul Alam SS, Hoque M, Karim S, Hussain IMR, Tabrez S. Identification of Potential Poly (ADP-Ribose) Polymerase-1 Inhibitors Derived from Rauwolfia serpentina: Possible Implication in Cancer Therapy. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:3787162. [PMID: 35368755 PMCID: PMC8967534 DOI: 10.1155/2022/3787162] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 02/21/2022] [Indexed: 12/18/2022]
Abstract
Poly (ADP-ribose) polymerase-1 (PARP-1) has been recognized as a prospective target for the development of novel cancer therapeutics. Several PARP-1 inhibitors are currently being considered for anticancer drug development and clinical investigation. Lately, natural compounds seem to be excellent alternative drug candidates for cancer treatment. Rauwolfia serpentina is a medicinal plant traditionally used in Indian subcontinents to treat various diseases. This study has been designed to identify the bioactive compounds derived from R. serpentina for possible binding and inhibition of PARP-1 using the molecular docking approach. Thirteen compounds were found to interact with the target with a binding affinity greater than the value of -9.0 kcal/mol. After screening the physicochemical properties, only 5 ligands (ajmalicine, yohimbine, isorauhimbine, rauwolscine, and 1,2-dihydrovomilenine) were found to obey all the parameters of Lipinski's rule of five, showed maximum drug-likeness, and possess no significant toxicity. These ligands displayed strong interactions with target PARP-1 via several hydrogen bonds and hydrophobic interactions. Therefore, these identified compounds derived from R. serpentina can be considered for drug development against cancer-targeting PARP-1.
Collapse
Affiliation(s)
- Adel M. Abuzenadah
- Department of Medical Laboratory Science, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Fatin Al-Sayes
- Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Mehboob Hoque
- Applied Bio-Chemistry Lab, Department of Biological Sciences, Aliah University, Kolkata, India
| | - Sajjad Karim
- Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ibtessam M. R. Hussain
- Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Shams Tabrez
- Department of Medical Laboratory Science, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
9
|
Colombo G, Gelardi ELM, Balestrero FC, Moro M, Travelli C, Genazzani AA. Insight Into Nicotinamide Adenine Dinucleotide Homeostasis as a Targetable Metabolic Pathway in Colorectal Cancer. Front Pharmacol 2021; 12:758320. [PMID: 34880756 PMCID: PMC8645963 DOI: 10.3389/fphar.2021.758320] [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: 08/13/2021] [Accepted: 11/02/2021] [Indexed: 11/13/2022] Open
Abstract
Tumour cells modify their cellular metabolism with the aim to sustain uncontrolled proliferation. Cancer cells necessitate adequate amounts of NAD and NADPH to support several enzymes that are usually overexpressed and/or overactivated. Nicotinamide adenine dinucleotide (NAD) is an essential cofactor and substrate of several NAD-consuming enzymes, such as PARPs and sirtuins, while NADPH is important in the regulation of the redox status in cells. The present review explores the rationale for targeting the key enzymes that maintain the cellular NAD/NADPH pool in colorectal cancer and the enzymes that consume or use NADP(H).
Collapse
Affiliation(s)
- Giorgia Colombo
- Department of Pharmaceutical Sciences, Università Del Piemonte Orientale, Novara, Italy
| | | | | | - Marianna Moro
- Department of Pharmaceutical Sciences, Università Del Piemonte Orientale, Novara, Italy
| | - Cristina Travelli
- Department of Drug Sciences, Università Degli Studi di Pavia, Pavia, Italy
| | - Armando A. Genazzani
- Department of Pharmaceutical Sciences, Università Del Piemonte Orientale, Novara, Italy
| |
Collapse
|
10
|
Huang YH, Yin SJ, Gong YY, Li ZR, Yang Q, Fan YX, Zhou T, Meng R, Wang P, He GH. PARP1 as a prognostic biomarker for human cancers: a meta-analysis. Biomark Med 2021; 15:1563-1578. [PMID: 34651514 DOI: 10.2217/bmm-2020-0891] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Aim: A comprehensive meta-analysis was carried out to evaluate the association between high PARP1 expression and clinical outcomes in diverse types of cancers. Materials & methods: The electronic databases for all articles about PARP1 expression and cancers were searched. Additionally, bioinformatics analysis was utilized to validate the results of the meta-analysis. Results: Fifty-two studies with a total of 7140 patients were included in the current meta-analysis. High PARP1 expression was found to be significantly associated with poor overall survival and recurrence in various cancers, which were further strengthened and complemented by the results of bioinformatic analysis. Furthermore, increased PAPR1 expression was also related to clinicopathological features. Conclusion: Our findings confirmed that PARP1 might be a promising biomarker for prognosis in human cancers.
Collapse
Affiliation(s)
- Yan-Hua Huang
- Research Center of Clinical Pharmacology, Yunnan Provincial Hospital of Traditional Chinese Medicine, 120 Guanghua Rd, Kunming, 650032, China.,Department of Pharmacy, 920th Hospital of Joint Logistics Support Force, 212 Daguan Rd, Kunming, 650032, China
| | - Sun-Jun Yin
- Department of Pharmacy, 920th Hospital of Joint Logistics Support Force, 212 Daguan Rd, Kunming, 650032, China
| | - Yuan-Yuan Gong
- Department of Pharmacy, 920th Hospital of Joint Logistics Support Force, 212 Daguan Rd, Kunming, 650032, China
| | - Zhi-Ran Li
- Department of Pharmacy, 920th Hospital of Joint Logistics Support Force, 212 Daguan Rd, Kunming, 650032, China
| | - Qin Yang
- Department of Pharmacy, 920th Hospital of Joint Logistics Support Force, 212 Daguan Rd, Kunming, 650032, China
| | - Yu-Xin Fan
- Department of Pharmacy, 920th Hospital of Joint Logistics Support Force, 212 Daguan Rd, Kunming, 650032, China
| | - Tao Zhou
- Department of Pharmacy, 920th Hospital of Joint Logistics Support Force, 212 Daguan Rd, Kunming, 650032, China
| | - Rui Meng
- Department of Pharmacy, 920th Hospital of Joint Logistics Support Force, 212 Daguan Rd, Kunming, 650032, China
| | - Ping Wang
- Department of Pharmacy, 920th Hospital of Joint Logistics Support Force, 212 Daguan Rd, Kunming, 650032, China
| | - Gong-Hao He
- Research Center of Clinical Pharmacology, Yunnan Provincial Hospital of Traditional Chinese Medicine, 120 Guanghua Rd, Kunming, 650032, China.,Department of Pharmacy, 920th Hospital of Joint Logistics Support Force, 212 Daguan Rd, Kunming, 650032, China
| |
Collapse
|
11
|
Anticancer Activities of Biogenic Silver Nanoparticles Targeting Apoptosis and Inflammatory Pathways in Colon Cancer Cells. J CLUST SCI 2021. [DOI: 10.1007/s10876-021-02143-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
12
|
Therapeutic Potential of PARP Inhibitors in the Treatment of Gastrointestinal Cancers. Biomedicines 2021; 9:biomedicines9081024. [PMID: 34440228 PMCID: PMC8392860 DOI: 10.3390/biomedicines9081024] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/05/2021] [Accepted: 08/10/2021] [Indexed: 12/20/2022] Open
Abstract
Gastrointestinal (GI) malignancies are a major global health burden, with high mortality rates. The identification of novel therapeutic strategies is crucial to improve treatment and survival of patients. The poly (ADP-ribose) polymerase (PARP) enzymes involved in the DNA damage response (DDR) play major roles in the development, progression and treatment response of cancer, with PARP inhibitors (PARPi) currently used in the clinic for breast, ovarian, fallopian, primary peritoneal, pancreatic and prostate cancers with deficiencies in homologous recombination (HR) DNA repair. This article examines the current evidence for the role of the DDR PARP enzymes (PARP1, 2, 3 and 4) in the development, progression and treatment response of GI cancers. Furthermore, we discuss the role of HR status as a predictive biomarker of PARPi efficacy in GI cancer patients and examine the pre-clinical and clinical evidence for PARPi and cytotoxic therapy combination strategies in GI cancer. We also include an analysis of the genomic and transcriptomic landscape of the DDR PARP genes and key HR genes (BRCA1, BRCA2, ATM, RAD51, MRE11, PALB2) in GI patient tumours (n = 1744) using publicly available datasets to identify patients that may benefit from PARPi therapeutic approaches.
Collapse
|
13
|
Zhai L, Liang H, Du J, Sun M, Qiu W, Tang H, Luo H. PARP-1 via regulation of p53 and p16, is involved in the hydroquinone-induced malignant transformation of TK6 cells by decelerating the cell cycle. Toxicol In Vitro 2021; 74:105153. [PMID: 33771647 DOI: 10.1016/j.tiv.2021.105153] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 02/23/2021] [Accepted: 03/22/2021] [Indexed: 12/24/2022]
Abstract
Poly(ADP-ribose)polymerase-1 (PARP-1) plays a crucial role in DNA damage repair and could be viewed as both a tumor promoter and tumor-suppressor gene. However, the effects of PARP-1 in hydroquinone-induced malignant transformation of TK6 cells remain to be further elucidated. The present research evaluated the potential mechanism of PARP-1 in hydroquinone-induced malignant transformation of TK6 cells. The results indicated that high PARP-1 inhibited TK6 cells malignant transformation after chronic exposure to HQ. We further confirmed that PARP-1 overexpression blocked cell proliferation, and decelerated cell cycle progression in vitro and in vivo. The immunoblotting analysis indicated that PARP-1 regulated cell cycle progression via p16/Rb and p53. Therefore, we conclude that PARP-1 is involved in HQ-induced malignant transformation associated with increasing p16/Rb and p53 which resulting in decelerating the cell cycle progression.
Collapse
Affiliation(s)
- Lu Zhai
- Department of Environmental and Occupational Health, Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, China
| | - Hairong Liang
- Department of Environmental and Occupational Health, Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, China
| | - Jinlin Du
- Department of Environmental and Occupational Health, Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, China
| | - Mingwei Sun
- Department of Environmental and Occupational Health, Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, China
| | - Weifeng Qiu
- Department of Environmental and Occupational Health, Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, China
| | - Huanwen Tang
- Department of Environmental and Occupational Health, Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, China.
| | - Hao Luo
- Department of Environmental and Occupational Health, Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, China.
| |
Collapse
|
14
|
Jeong KY, Park M. Poly adenosine diphosphate-ribosylation, a promising target for colorectal cancer treatment. World J Gastrointest Oncol 2021. [PMID: 34163574 DOI: 10.4251/wjgo.v13.i6.574.] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The development of colorectal cancer (CRC) can result from changes in a variety of cellular systems within the tumor microenvironment. Particularly, it is primarily associated with genomic instability that is the gradual accumulation of genetic and epigenetic changes consisting of a characteristic set of mutations crucial for pathways in CRC progression. Based on this background, the potential to focus on poly [adenosine diphosphate (ADP)-ribose] polymerase (PARP)-1 and poly-ADP ribosylation (PARylation) as the main causes of malignant formation of CRC may be considered. One of the important functions of PARP-1 and PARylation is its deoxyribonucleic acid (DNA) repair function, which plays a pivotal role in the DNA damage response and prevention of DNA damage maintaining the redox homeostasis involved in the regulation of oxidation and superoxide. PARP-1 and PARylation can also alter epigenetic markers and chromatin structure involved in transcriptional regulation for the oncogenes or tumor suppressor genes by remodeling histone and chromatin enzymes. Given the high importance of these processes in CRC, it can be considered that PARP-1 and PARylation are at the forefront of the pathological changes required for CRC progression. Therefore, this review addresses the current molecular biological features for understanding the multifactorial function of PARP-1 and PARylation in CRC related to the aforementioned roles; furthermore, it presents a summary of recent approaches with PARP-1 inhibition in non-clinical and clinical studies targeting CRC. This understanding could help embrace the importance of targeting PARP-1 and PARylation in the treatment of CRC, which may present the potential to identify various research topics that can be challenged both non-clinically and clinically.
Collapse
Affiliation(s)
- Keun-Yeong Jeong
- Research and Development, Metimedi Pharmaceuticals, Incheon 22006, South Korea.
| | - Minhee Park
- Research and Development, Metimedi Pharmaceuticals, Incheon 22006, South Korea
| |
Collapse
|
15
|
Jeong KY, Park M. Poly adenosine diphosphate-ribosylation, a promising target for colorectal cancer treatment. World J Gastrointest Oncol 2021; 13:574-588. [PMID: 34163574 PMCID: PMC8204356 DOI: 10.4251/wjgo.v13.i6.574] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/22/2021] [Accepted: 05/08/2021] [Indexed: 02/06/2023] Open
Abstract
The development of colorectal cancer (CRC) can result from changes in a variety of cellular systems within the tumor microenvironment. Particularly, it is primarily associated with genomic instability that is the gradual accumulation of genetic and epigenetic changes consisting of a characteristic set of mutations crucial for pathways in CRC progression. Based on this background, the potential to focus on poly [adenosine diphosphate (ADP)-ribose] polymerase (PARP)-1 and poly-ADP ribosylation (PARylation) as the main causes of malignant formation of CRC may be considered. One of the important functions of PARP-1 and PARylation is its deoxyribonucleic acid (DNA) repair function, which plays a pivotal role in the DNA damage response and prevention of DNA damage maintaining the redox homeostasis involved in the regulation of oxidation and superoxide. PARP-1 and PARylation can also alter epigenetic markers and chromatin structure involved in transcriptional regulation for the oncogenes or tumor suppressor genes by remodeling histone and chromatin enzymes. Given the high importance of these processes in CRC, it can be considered that PARP-1 and PARylation are at the forefront of the pathological changes required for CRC progression. Therefore, this review addresses the current molecular biological features for understanding the multifactorial function of PARP-1 and PARylation in CRC related to the aforementioned roles; furthermore, it presents a summary of recent approaches with PARP-1 inhibition in non-clinical and clinical studies targeting CRC. This understanding could help embrace the importance of targeting PARP-1 and PARylation in the treatment of CRC, which may present the potential to identify various research topics that can be challenged both non-clinically and clinically.
Collapse
Affiliation(s)
- Keun-Yeong Jeong
- Research and Development, Metimedi Pharmaceuticals, Incheon 22006, South Korea
| | - Minhee Park
- Research and Development, Metimedi Pharmaceuticals, Incheon 22006, South Korea
| |
Collapse
|
16
|
Ergul M, Aktan F, Yildiz MT, Tutar Y. Perturbation of HSP Network in MCF-7 Breast Cancer Cell Line Triggers Inducible HSP70 Expression and Leads to Tumor Suppression. Anticancer Agents Med Chem 2021; 20:1051-1060. [PMID: 32053081 DOI: 10.2174/1871520620666200213102210] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 10/09/2019] [Accepted: 12/28/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Heat shock protein 70 (HSP70) is constitutively expressed in normal cells but aberrantly expressed in several types of tumor cells, helping their survival in extreme conditions. Thus, specific inhibition of HSP70 in tumor cells is a promising strategy in the treatment of cancer. HSP70 has a variety of isoforms in the cellular organelles and form different functions by coordinating and cooperating with cochaperones. Cancer cells overexpress HSPs during cell growth and proliferation and HSP network provides resistance against apoptosis. The present study aimed to evaluate quantitative changes in HSPs- and cancerassociated gene expressions and their interactions in the presence of 2-phenylethyenesulfonamide (PES) in MCF-7 cells. METHODS Antiproliferative activity of PES was evaluated using the XTT assay. Inducible HSP70 (HSP70i) levels in the PES-treated cells were determined using the ELISA kit. PCR Array was performed to assess the HSPs- and cancer-pathway focused gene expression profiling. Gene network analysis was performed using the X2K, yEd (V.3.18.1) programs, and web-based gene list enrichment analysis tool Enrichr. RESULTS The results demonstrated that PES exposure increased the amount of both HSP70i gene and protein expression surprisingly. However, the expression of HSP70 isoforms as well as other co-chaperones, and 17 cancer-associated genes decreased remarkably as expected. Additionally, interaction network analysis revealed a different mechanism; PES induction of HSP70i employs a cell cycle negative regulator, RB1, which is a tumor suppressor gene. CONCLUSION PES treatment inhibited MCF-7 cell proliferation and changed several HSPs- and cancer-related gene expressions along with their interactions through a unique mechanism although it causes an interesting increase at HSP70i gene and protein expressions. RB1 gene expression may play an important role in this effect as revealed by the interaction network analysis.
Collapse
Affiliation(s)
- Mustafa Ergul
- Department of Biochemistry, Faculty of Pharmacy, Sivas Cumhuriyet University, Sivas, Turkey
| | - Fugen Aktan
- Department of Biochemistry, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - Mehmet T Yildiz
- Division of Molecular Medicine, Hamidiye Institute of Health Sciences, University of Health Sciences, Istanbul, Turkey
| | - Yusuf Tutar
- Division of Molecular Medicine, Hamidiye Institute of Health Sciences, University of Health Sciences, Istanbul, Turkey
| |
Collapse
|
17
|
|
18
|
The Prognostic Values of PARP-1 Expression in Uveal Melanoma. Cells 2021; 10:cells10020285. [PMID: 33572586 PMCID: PMC7911114 DOI: 10.3390/cells10020285] [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: 12/31/2020] [Revised: 01/25/2021] [Accepted: 01/27/2021] [Indexed: 12/19/2022] Open
Abstract
Background: Uveal melanoma is the most common primary intraocular malignancy in adults. In advanced cases, the prognosis is very poor. Thus far, no effective methods of pharmacotherapy of this cancer have been found. The aim of the study was to evaluate the expression of PARP-1, the best-known member of the family of poly(ADP-ribose) polymerases, in uveal melanoma and its associations with clinicopathological parameters, overall survival, and disease-free survival. Methods: The study included 91 patients who underwent enucleation due to uveal melanoma. PARP-1 expression was assessed by immunohistochemistry. Results: High PARP-1 expression was associated with more frequent chromosome 3 loss, higher histopathological grade, bigger tumor size, and absence of intrascleral extension. High PARP-1 expression was associated with shorter overall survival time and disease-free survival time. Conclusions: The above findings indicate that high expression of PARP-1 can be considered as an unfavorable prognostic factor in uveal melanoma.
Collapse
|
19
|
Kossatz S, Pirovano G, Demétrio De Souza França P, Strome AL, Sunny SP, Zanoni DK, Mauguen A, Carney B, Brand C, Shah V, Ramanajinappa RD, Hedne N, Birur P, Sihag S, Ghossein RA, Gönen M, Strome M, Suresh A, Molena D, Ganly I, Kuriakose MA, Patel SG, Reiner T. Validation of the use of a fluorescent PARP1 inhibitor for the detection of oral, oropharyngeal and oesophageal epithelial cancers. Nat Biomed Eng 2020; 4:272-285. [PMID: 32165735 PMCID: PMC7136849 DOI: 10.1038/s41551-020-0526-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 02/06/2020] [Indexed: 11/09/2022]
Abstract
For oral, oropharyngeal and oesophageal cancer, the early detection of tumours and of residual tumour after surgery are prognostic factors of recurrence rates and patient survival. Here, we report the validation, in animal models and a human, of the use of a previously described fluorescently labelled small-molecule inhibitor of the DNA repair enzyme poly(ADP-ribose) polymerase 1 (PARP1) for the detection of cancers of the oral cavity, pharynx and oesophagus. We show that the fluorescent contrast agent can be used to quantify the expression levels of PARP1 and to detect oral, oropharyngeal and oesophageal tumours in mice, pigs and fresh human biospecimens when delivered topically or intravenously. The fluorescent PARP1 inhibitor can also detect oral carcinoma in a patient when applied as a mouthwash, and discriminate between fresh biopsied samples of the oral tumour and the surgical resection margin with more than 95% sensitivity and specificity. The PARP1 inhibitor could serve as the basis of a rapid and sensitive assay for the early detection and for the surgical-margin assessment of epithelial cancers of the upper intestinal tract.
Collapse
Affiliation(s)
- Susanne Kossatz
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Nuclear Medicine, University Hospital Klinikum Rechts der Isar, Technical University Munich, Munich, Germany
| | - Giacomo Pirovano
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Arianna L Strome
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sumsum P Sunny
- Integrated Head and Neck Oncology Research Program, Mazumdar Shaw Medical Foundation, Narayana Health, Bengaluru, India
- Head and Neck Oncology, Mazumdar Shaw Medical Center, Narayana Health, Bengaluru, India
| | | | - Audrey Mauguen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Brandon Carney
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Christian Brand
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Summit Biomedical Imaging, New York, NY, USA
| | - Veer Shah
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ravindra D Ramanajinappa
- Integrated Head and Neck Oncology Research Program, Mazumdar Shaw Medical Foundation, Narayana Health, Bengaluru, India
| | - Naveen Hedne
- Head and Neck Oncology, Mazumdar Shaw Medical Center, Narayana Health, Bengaluru, India
| | - Praveen Birur
- Integrated Head and Neck Oncology Research Program, Mazumdar Shaw Medical Foundation, Narayana Health, Bengaluru, India
- Department of Oral Medicine and Radiology, KLES Institute of Dental Sciences, Bangalore, India
| | - Smita Sihag
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ronald A Ghossein
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mithat Gönen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Amritha Suresh
- Integrated Head and Neck Oncology Research Program, Mazumdar Shaw Medical Foundation, Narayana Health, Bengaluru, India
| | - Daniela Molena
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ian Ganly
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Moni A Kuriakose
- Integrated Head and Neck Oncology Research Program, Mazumdar Shaw Medical Foundation, Narayana Health, Bengaluru, India
- Cochin Cancer Research Center, Kochi, India
| | - Snehal G Patel
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Radiology, Weill Cornell Medical College, New York, NY, USA.
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
20
|
Tudek A, Czerwińska J, Kosicki K, Zdżalik-Bielecka D, Shahmoradi Ghahe S, Bażlekowa-Karaban M, Borsuk EM, Speina E. DNA damage, repair and the improvement of cancer therapy - A tribute to the life and research of Barbara Tudek. Mutat Res 2020; 852:503160. [PMID: 32265045 DOI: 10.1016/j.mrgentox.2020.503160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/10/2020] [Accepted: 02/13/2020] [Indexed: 10/25/2022]
Abstract
Professor Barbara Tudek received the Frits Sobels Award in 2019 from the European Environmental Mutagenesis and Genomics Society (EEMGS). This article presents her outstanding character and most important lines of research. The focus of her studies covered alkylative and oxidative damage to DNA bases, in particular mutagenic and carcinogenic properties of purines with an open imidazole ring and 8-oxo-7,8-dihydroguanine (8-oxoGua). They also included analysis of mutagenic properties and pathways for the repair of DNA adducts of lipid peroxidation (LPO) products arising in large quantities during inflammation. Professor Tudek did all of this in the hope of deciphering the mechanisms of DNA damage removal, in particular by the base excision repair (BER) pathway. Some lines of research aimed at discovering factors that can modulate the activity of DNA damage repair in hope to enhance existing anti-cancer therapies. The group's ongoing research aims at deciphering the resistance mechanisms of cancer cell lines acquired following prolonged exposure to photodynamic therapy (PDT) and the possibility of re-sensitizing cells to PDT in order to increase the application of this minimally invasive therapeutic method.
Collapse
Affiliation(s)
- Agnieszka Tudek
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106 Warsaw, Poland
| | - Jolanta Czerwińska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106 Warsaw, Poland
| | - Konrad Kosicki
- Faculty of Biology, Institute of Genetics and Biotechnology, University of Warsaw, Pawińskiego 5a, 02-106 Warsaw, Poland
| | - Daria Zdżalik-Bielecka
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, Księcia Trojdena 4, 02-109 Warsaw, Poland
| | - Somayeh Shahmoradi Ghahe
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106 Warsaw, Poland
| | - Milena Bażlekowa-Karaban
- Faculty of Biology, Institute of Genetics and Biotechnology, University of Warsaw, Pawińskiego 5a, 02-106 Warsaw, Poland; UMR 8200 C.N.R.S., Université Paris-Saclay, Gustave Roussy Cancer Campus, F-94805 Villejuif Cedex, France
| | - Ewelina M Borsuk
- Laboratory of Structural Biology, International Institute of Molecular and Cell Biology, Księcia Trojdena 4, 02-109 Warsaw, Poland
| | - Elżbieta Speina
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106 Warsaw, Poland.
| |
Collapse
|
21
|
Abstract
In this review, Slade provides an overview of the molecular mechanisms and cellular consequences of PARP and PARG inhibition. The author also highlights the clinical performance of four PARP inhibitors used in cancer therapy (olaparib, rucaparib, niraparib, and talazoparib) and discusses the predictive biomarkers of inhibitor sensitivity and mechanisms of resistance as well as the means of overcoming them through combination therapy. Oxidative and replication stress underlie genomic instability of cancer cells. Amplifying genomic instability through radiotherapy and chemotherapy has been a powerful but nonselective means of killing cancer cells. Precision medicine has revolutionized cancer therapy by putting forth the concept of selective targeting of cancer cells. Poly(ADP-ribose) polymerase (PARP) inhibitors represent a successful example of precision medicine as the first drugs targeting DNA damage response to have entered the clinic. PARP inhibitors act through synthetic lethality with mutations in DNA repair genes and were approved for the treatment of BRCA mutated ovarian and breast cancer. PARP inhibitors destabilize replication forks through PARP DNA entrapment and induce cell death through replication stress-induced mitotic catastrophe. Inhibitors of poly(ADP-ribose) glycohydrolase (PARG) exploit and exacerbate replication deficiencies of cancer cells and may complement PARP inhibitors in targeting a broad range of cancer types with different sources of genomic instability. Here I provide an overview of the molecular mechanisms and cellular consequences of PARP and PARG inhibition. I highlight clinical performance of four PARP inhibitors used in cancer therapy (olaparib, rucaparib, niraparib, and talazoparib) and discuss the predictive biomarkers of inhibitor sensitivity, mechanisms of resistance as well as the means of overcoming them through combination therapy.
Collapse
Affiliation(s)
- Dea Slade
- Department of Biochemistry, Max Perutz Labs, Vienna Biocenter (VBC), University of Vienna, 1030 Vienna, Austria
| |
Collapse
|
22
|
Pazzaglia S, Pioli C. Multifaceted Role of PARP-1 in DNA Repair and Inflammation: Pathological and Therapeutic Implications in Cancer and Non-Cancer Diseases. Cells 2019; 9:cells9010041. [PMID: 31877876 PMCID: PMC7017201 DOI: 10.3390/cells9010041] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 12/18/2019] [Accepted: 12/20/2019] [Indexed: 12/12/2022] Open
Abstract
PARP-1 (poly(ADP-ribose)-polymerase 1), mainly known for its protective role in DNA repair, also regulates inflammatory processes. Notably, defects in DNA repair and chronic inflammation may both predispose to cancer development. On the other hand, inhibition of DNA repair and inflammatory responses can be beneficial in cancer therapy and PARP inhibitors are currently used for their lethal effects on tumor cells. Furthermore, excess of PARP-1 activity has been associated with many tumors and inflammation-related clinical conditions, including asthma, sepsis, arthritis, atherosclerosis, and neurodegenerative diseases, to name a few. Activation and inhibition of PARP represent, therefore, a double-edged sword that can be exploited for therapeutic purposes. In our review, we will discuss recent findings highlighting the composite multifaceted role of PARP-1 in cancer and inflammation-related diseases.
Collapse
Affiliation(s)
- Simonetta Pazzaglia
- Correspondence: (S.P.); (C.P.); Tel.: +39-06-3048-6535 (S.P.); +39-06-3048-3398 (C.P.)
| | - Claudio Pioli
- Correspondence: (S.P.); (C.P.); Tel.: +39-06-3048-6535 (S.P.); +39-06-3048-3398 (C.P.)
| |
Collapse
|
23
|
Ni F, Tang H, Wang C, Wang Z, Yu F, Chen B, Sun L. Berzosertib (VE-822) inhibits gastric cancer cell proliferation via base excision repair system. Cancer Manag Res 2019; 11:8391-8405. [PMID: 31571995 PMCID: PMC6750847 DOI: 10.2147/cmar.s217375] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 09/03/2019] [Indexed: 12/15/2022] Open
Abstract
Background Current investigations suggest that the Base Excision Repair (BER) system may change DNA repair capacity and affect clinical gastric cancer progression such as overall survival. However, the prognostic value of BER system members in gastric cancer remains unclear. Methods We explored the prognostic correlation between 7 individual BER genes, including uracil-DNA glycosylase (UNG), Single-strand-selective monofunctional uracil-DNA glycosylase 1 (SMUG1), Methyl-CpG binding domain 4 (MBD4), thymine DNA glycosylase (TDG), 8-oxoguanine DNA glycosylase (OGG1), MutY DNA glycosylase (MUTYH) and Nei like DNA glycosylase 1 (NEIL1), expression and overall survival (OS) in different clinical data, such as Lauren classification, pathological stages, human epidermal growth factor receptor-2 (HER2) expression status, treatment strategy, gender and differentiation degree in gastric cancer patients, using Kaplan-Meier plotter (KM plotter) online database. Based on the bioinformatics analysis, we utilized Berzosertib (VE-822) to inhibit DNA damage repair in cancer cells compared to solvent control group via real-time cellular analysis (RTCA), flow cytometry, colony formation and migration assay. Finally, we utilized reverse transcription-polymerase chain reaction (RT-PCR) to confirm the expression of BER members between normal and two gastric cancer cells or solvent and VE-822 treated groups. Results Our work revealed that high UNG mRNA expression was correlated with high overall survival probability; however, high SMUG1, MBD4, TDG, OGG1, MUTYH and NEIL1 mRNA expression showed relatively low overall survival probability in all GC patients. Additionally, UNG was associated with high overall survival probability in intestinal and diffuse types, but SMUG1 and NEIL1 showed opposite results. Further, VE-822 pharmacological experiment suggested that inhibition of DNA damage repair suppressed gastric cancer cells’ proliferation and migration ability via inducing apoptosis. Further, real-time polymerase chain reaction results proposed the inhibition of gastric cancer cells by VE-822 may be through UNG, MUTYH and OGG-1 of BER system. Conclusion We comprehensively analyze the prognostic value of the BER system (UNG, SMUG1, MBD4, TDG, OGG1, MUTYH and NEIL1) based on bioinformatics analysis and experimental confirmation. BER members are associated with distinctive prognostic significance and maybe new valuable prognostic indicators in gastric cancer.
Collapse
Affiliation(s)
- Fubiao Ni
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, Zhejiang Provincial Top Key Discipline in Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Hengjie Tang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, Zhejiang Provincial Top Key Discipline in Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Cheng Wang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, Zhejiang Provincial Top Key Discipline in Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Zixiang Wang
- First College of Clinical Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Fangyi Yu
- First College of Clinical Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Bicheng Chen
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, Zhejiang Provincial Top Key Discipline in Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| | - Linxiao Sun
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, Zhejiang Provincial Top Key Discipline in Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China
| |
Collapse
|
24
|
Abstract
Mitosis ensures accurate segregation of duplicated DNA through tight regulation of chromosome condensation, bipolar spindle assembly, chromosome alignment in the metaphase plate, chromosome segregation and cytokinesis. Poly(ADP-ribose) polymerases (PARPs), in particular PARP1, PARP2, PARP3, PARP5a (TNKS1), as well as poly(ADP-ribose) glycohydrolase (PARG), regulate different mitotic functions, including centrosome function, mitotic spindle assembly, mitotic checkpoints, telomere length and telomere cohesion. PARP depletion or inhibition give rise to various mitotic defects such as centrosome amplification, multipolar spindles, chromosome misalignment, premature loss of cohesion, metaphase arrest, anaphase DNA bridges, lagging chromosomes, and micronuclei. As the mechanisms of PARP1/2 inhibitor-mediated cell death are being progressively elucidated, it is becoming clear that mitotic defects caused by PARP1/2 inhibition arise due to replication stress and DNA damage in S phase. As it stands, entrapment of inactive PARP1/2 on DNA phenocopies replication stress through accumulation of unresolved replication intermediates, double-stranded DNA breaks (DSBs) and incorrectly repaired DSBs, which can be transmitted from S phase to mitosis and instigate various mitotic defects, giving rise to both numerical and structural chromosomal aberrations. Cancer cells have increased levels of replication stress, which makes them particularly susceptible to a combination of agents that compromise replication fork stability. Indeed, combining PARP1/2 inhibitors with genetic deficiencies in DNA repair pathways, DNA-damaging agents, ATR and other cell cycle checkpoint inhibitors has yielded synergistic effects in killing cancer cells. Here I provide a comprehensive overview of the mitotic functions of PARPs and PARG, mitotic phenotypes induced by their depletion or inhibition, as well as the therapeutic relevance of targeting mitotic cells by directly interfering with mitotic functions or indirectly through replication stress.
Collapse
Affiliation(s)
- Dea Slade
- Department of Biochemistry, Max F. Perutz Laboratories, University of Vienna, Vienna Biocenter (VBC), Dr. Bohr-gasse 9, 1030 Vienna, Austria.
| |
Collapse
|
25
|
PARP1: A potential biomarker for gastric cancer. Pathol Res Pract 2019; 215:152472. [DOI: 10.1016/j.prp.2019.152472] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 05/13/2019] [Accepted: 05/23/2019] [Indexed: 12/14/2022]
|
26
|
Schlimpert M, Lagies S, Müller B, Budnyk V, Blanz KD, Walz G, Kammerer B. Metabolic perturbations caused by depletion of nephronophthisis factor Anks6 in mIMCD3 cells. Metabolomics 2019; 15:71. [PMID: 31041607 DOI: 10.1007/s11306-019-1535-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 04/24/2019] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Nephronophthisis (NPH) is an inherited form of cystic kidney disease with various extrarenal manifestations accounting for the largest amount of endstage renal disease in childhood. Patient mutations of Anks6 have also been found to cause NPH like phenotypes in animal models. However, little is known about functionality of Anks6. OBJECTIVES/METHODS We investigated the impact of Anks6 depletion on cellular metabolism of inner medullary collecting duct cells by GC-MS profiling and targeted LC-MS/MS analysis using two different shRNA cell lines for tetracycline-inducible Anks6 downregulation, namely mIMCD3 krab shANKS6 i52 and mIMCD3 krab shANKS6 i12. RESULTS In combination, we could successfully identify 158 metabolites of which 20 compounds showed similar alterations in both knockdown systems. Especially, large neutral amino acids, such as phenylalanine, where found to be significantly downregulated indicating disturbances in amino acid metabolism. Arginine, lysine and spermidine, which play an important role in cell survival and proliferation, were found to be downregulated. Accordingly, cell growth was diminished in tet treated mIMCD3 krab shANKS6 i52 knockdown cells. Deoxynucleosides were significantly downregulated in both knockdown systems. Hence, PARP1 levels were increased in tet treated mIMCD3 krab shANKS6 i52 cells, but not in tet treated mIMCD3 krab shANKS6 i12 cells. However, yH2AX was found to be increased in the latter. CONCLUSION In combination, we hypothesise that Anks6 affects DNA damage responses and proliferation and plays a crucial role in physiological amino acid and purine/pyrimidine metabolism.
Collapse
Affiliation(s)
- Manuel Schlimpert
- Center for Biological Systems Analysis, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
- Faculty of Biology, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Simon Lagies
- Center for Biological Systems Analysis, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
- Faculty of Biology, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
| | - Barbara Müller
- Renal Division, Department of Medicine, Albert-Ludwigs-University of Freiburg, Medical Center, Freiburg, Germany
| | - Vadym Budnyk
- Renal Division, Department of Medicine, Albert-Ludwigs-University of Freiburg, Medical Center, Freiburg, Germany
| | - Kelly Daryll Blanz
- Spemann Graduate School of Biology and Medicine, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
- Faculty of Biology, Albert-Ludwigs-University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Albert-Ludwigs-University of Freiburg, Medical Center, Freiburg, Germany
| | - Gerd Walz
- Renal Division, Department of Medicine, Albert-Ludwigs-University of Freiburg, Medical Center, Freiburg, Germany
| | - Bernd Kammerer
- Center for Biological Systems Analysis, Albert-Ludwigs-University of Freiburg, Freiburg, Germany.
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
27
|
Yun EJ, Lin CJ, Dang A, Hernandez E, Guo J, Chen WM, Allison J, Kim N, Kapur P, Brugarolas J, Wu K, He D, Lai CH, Lin H, Saha D, Baek ST, Chen BPC, Hsieh JT. Downregulation of Human DAB2IP Gene Expression in Renal Cell Carcinoma Results in Resistance to Ionizing Radiation. Clin Cancer Res 2019; 25:4542-4551. [PMID: 31000589 DOI: 10.1158/1078-0432.ccr-18-3004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 01/21/2019] [Accepted: 04/15/2019] [Indexed: 12/24/2022]
Abstract
PURPOSE Renal cell carcinoma (RCC) is known to be highly radioresistant but the mechanisms associated with radioresistance have remained elusive. We found DOC-2/DAB2 interactive protein (DAB2IP) frequently downregulated in RCC, is associated with radioresistance. In this study, we investigated the underlying mechanism regulating radioresistance by DAB2IP and developed appropriate treatment. EXPERIMENTAL DESIGN Several RCC lines with or without DAB2IP expression were irradiated with ionizing radiation (IR) for determining their radiosensitivities based on colony formation assay. To investigate the underlying regulatory mechanism of DAB2IP, immunoprecipitation-mass spectrometry was performed to identify DAB2IP-interactive proteins. PARP-1 expression and enzymatic activity were determined using qRT-PCR, Western blot analysis, and ELISA. In vivo ubiquitination assay was used to test PARP-1 degradation. Furthermore, in vivo mice xenograft model and patient-derived xenograft (PDX) model were used to determine the effect of combination therapy to sensitizing tumors to IR. RESULTS We notice that DAB2IP-deficient RCC cells acquire IR-resistance. Mechanistically, DAB2IP can form a complex with PARP-1 and E3 ligases that is responsible for degrading PARP-1. Indeed, elevated PARP-1 levels are associated with the IR resistance in RCC cells. Furthermore, PARP-1 inhibitor can enhance the IR response of either RCC xenograft model or PDX model. CONCLUSIONS In this study, we unveil that loss of DAB2IP resulted in elevated PARP-1 protein is associated with IR-resistance in RCC. These results provide a new targeting strategy to improve the efficacy of radiotherapy of RCC.
Collapse
Affiliation(s)
- Eun-Jin Yun
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas
- Division of Integrative Bioscience and Biotechnology, POSTECH, Pohang, Republic of Korea
| | - Chun-Jung Lin
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Andrew Dang
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Elizabeth Hernandez
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jiaming Guo
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Wei-Min Chen
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Joyce Allison
- Department of Internal Medicine and Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Nathan Kim
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Payal Kapur
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - James Brugarolas
- Department of Internal Medicine and Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Kaijie Wu
- Department of Urology, The First Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, China
| | - Dalin He
- Department of Urology, The First Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, China
| | - Chih-Ho Lai
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ho Lin
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Debabrata Saha
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Seung Tae Baek
- Division of Integrative Bioscience and Biotechnology, POSTECH, Pohang, Republic of Korea
| | - Benjamin P C Chen
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas.
| | - Jer-Tsong Hsieh
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas.
- Graduate Institute of Cancer Biology, China Medical University Hospital, Taichung, Taiwan, Republic of China
| |
Collapse
|
28
|
Shirai F, Tsumura T, Yashiroda Y, Yuki H, Niwa H, Sato S, Chikada T, Koda Y, Washizuka K, Yoshimoto N, Abe M, Onuki T, Mazaki Y, Hirama C, Fukami T, Watanabe H, Honma T, Umehara T, Shirouzu M, Okue M, Kano Y, Watanabe T, Kitamura K, Shitara E, Muramatsu Y, Yoshida H, Mizutani A, Seimiya H, Yoshida M, Koyama H. Discovery of Novel Spiroindoline Derivatives as Selective Tankyrase Inhibitors. J Med Chem 2019; 62:3407-3427. [DOI: 10.1021/acs.jmedchem.8b01888] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Takehiro Fukami
- RIKEN Program for Drug Discovery and Medical Technology Platforms, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | | | | | | | | | | | | | | | | | | | - Yukiko Muramatsu
- Division of Molecular Biotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, 3-8-31 Ariake,
Koto-ku, Tokyo 135-8850, Japan
| | - Haruka Yoshida
- Division of Molecular Biotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, 3-8-31 Ariake,
Koto-ku, Tokyo 135-8850, Japan
| | - Anna Mizutani
- Division of Molecular Biotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, 3-8-31 Ariake,
Koto-ku, Tokyo 135-8850, Japan
| | - Hiroyuki Seimiya
- Division of Molecular Biotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, 3-8-31 Ariake,
Koto-ku, Tokyo 135-8850, Japan
| | - Minoru Yoshida
- Department of Biotechnology, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | | |
Collapse
|
29
|
Strome A, Kossatz S, Zanoni DK, Rajadhyaksha M, Patel S, Reiner T. Current Practice and Emerging Molecular Imaging Technologies in Oral Cancer Screening. Mol Imaging 2018; 17:1536012118808644. [PMID: 32852263 PMCID: PMC6287312 DOI: 10.1177/1536012118808644] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Oral cancer is one of the most common cancers globally. Survival rates for patients are directly correlated with stage of diagnosis; despite this knowledge, 60% of individuals are presenting with late-stage disease. Currently, the initial evaluation of a questionable lesion is performed by a conventional visual examination with white light. If a lesion is deemed suspicious, a biopsy is taken for diagnosis. However, not all lesions present suspicious under visual white light examination, and there is limited specificity in differentiating between benign and malignant transformations. Several vital dyes, light-based detection systems, and cytology evaluation methods have been formulated to aid in the visualization process, but their lack of specific biomarkers resulted in high false-positive rates and thus limits their reliability as screening and guidance tools. In this review, we will analyze the current methodologies and demonstrate the need for specific intraoral imaging agents to aid in screening and diagnosis to identify patients earlier. Several novel molecular imaging agents will be presented as, by result of their molecular targeting, they aim to have high specificity for tumor pathways and can support in identifying dysplastic/cancerous lesions and guiding visualization of biopsy sites. Imaging agents that are easy to use, inexpensive, noninvasive, and specific can be utilized to increase the number of patients who are screened and monitored in a variety of different environments, with the ultimate goal of increasing early detection.
Collapse
Affiliation(s)
- Arianna Strome
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Susanne Kossatz
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Milind Rajadhyaksha
- Dermatology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Snehal Patel
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Radiology, Weill-Cornell Medical College, New York, NY, USA
| |
Collapse
|
30
|
Schiewer MJ, Mandigo AC, Gordon N, Huang F, Gaur S, de Leeuw R, Zhao SG, Evans J, Han S, Parsons T, Birbe R, McCue P, McNair C, Chand SN, Cendon-Florez Y, Gallagher P, McCann JJ, Poudel Neupane N, Shafi AA, Dylgjeri E, Brand LJ, Visakorpi T, Raj GV, Lallas CD, Trabulsi EJ, Gomella LG, Dicker AP, Kelly WK, Leiby BE, Knudsen B, Feng FY, Knudsen KE. PARP-1 regulates DNA repair factor availability. EMBO Mol Med 2018; 10:e8816. [PMID: 30467127 PMCID: PMC6284389 DOI: 10.15252/emmm.201708816] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 10/10/2018] [Accepted: 10/25/2018] [Indexed: 12/22/2022] Open
Abstract
PARP-1 holds major functions on chromatin, DNA damage repair and transcriptional regulation, both of which are relevant in the context of cancer. Here, unbiased transcriptional profiling revealed the downstream transcriptional profile of PARP-1 enzymatic activity. Further investigation of the PARP-1-regulated transcriptome and secondary strategies for assessing PARP-1 activity in patient tissues revealed that PARP-1 activity was unexpectedly enriched as a function of disease progression and was associated with poor outcome independent of DNA double-strand breaks, suggesting that enhanced PARP-1 activity may promote aggressive phenotypes. Mechanistic investigation revealed that active PARP-1 served to enhance E2F1 transcription factor activity, and specifically promoted E2F1-mediated induction of DNA repair factors involved in homologous recombination (HR). Conversely, PARP-1 inhibition reduced HR factor availability and thus acted to induce or enhance "BRCA-ness". These observations bring new understanding of PARP-1 function in cancer and have significant ramifications on predicting PARP-1 inhibitor function in the clinical setting.
Collapse
Affiliation(s)
- Matthew J Schiewer
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Cancer Center Thomas Jefferson University, Philadelphia, PA, USA
| | - Amy C Mandigo
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Cancer Center Thomas Jefferson University, Philadelphia, PA, USA
| | - Nicolas Gordon
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Cancer Center Thomas Jefferson University, Philadelphia, PA, USA
| | | | | | - Renée de Leeuw
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Cancer Center Thomas Jefferson University, Philadelphia, PA, USA
| | - Shuang G Zhao
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Joseph Evans
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Sumin Han
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Theodore Parsons
- Sidney Kimmel Cancer Center Thomas Jefferson University, Philadelphia, PA, USA
- Department of Pathology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ruth Birbe
- Cooper University Health, Camden, NJ, USA
| | - Peter McCue
- Sidney Kimmel Cancer Center Thomas Jefferson University, Philadelphia, PA, USA
- Department of Pathology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Christopher McNair
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Cancer Center Thomas Jefferson University, Philadelphia, PA, USA
| | - Saswati N Chand
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Cancer Center Thomas Jefferson University, Philadelphia, PA, USA
| | - Ylenia Cendon-Florez
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Cancer Center Thomas Jefferson University, Philadelphia, PA, USA
| | - Peter Gallagher
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Cancer Center Thomas Jefferson University, Philadelphia, PA, USA
| | - Jennifer J McCann
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Cancer Center Thomas Jefferson University, Philadelphia, PA, USA
| | - Neermala Poudel Neupane
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Cancer Center Thomas Jefferson University, Philadelphia, PA, USA
| | - Ayesha A Shafi
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Cancer Center Thomas Jefferson University, Philadelphia, PA, USA
| | - Emanuela Dylgjeri
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Cancer Center Thomas Jefferson University, Philadelphia, PA, USA
| | - Lucas J Brand
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Cancer Center Thomas Jefferson University, Philadelphia, PA, USA
| | | | | | - Costas D Lallas
- Sidney Kimmel Cancer Center Thomas Jefferson University, Philadelphia, PA, USA
- Department of Urology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Edouard J Trabulsi
- Sidney Kimmel Cancer Center Thomas Jefferson University, Philadelphia, PA, USA
- Department of Urology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Leonard G Gomella
- Sidney Kimmel Cancer Center Thomas Jefferson University, Philadelphia, PA, USA
- Department of Urology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Adam P Dicker
- Sidney Kimmel Cancer Center Thomas Jefferson University, Philadelphia, PA, USA
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Wm Kevin Kelly
- Sidney Kimmel Cancer Center Thomas Jefferson University, Philadelphia, PA, USA
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Benjamin E Leiby
- Sidney Kimmel Cancer Center Thomas Jefferson University, Philadelphia, PA, USA
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Felix Y Feng
- Departments of Radiation Oncology, Urology, and Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Karen E Knudsen
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Cancer Center Thomas Jefferson University, Philadelphia, PA, USA
- Department of Urology, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
31
|
Li X, Li C, Jin J, Wang J, Huang J, Ma Z, Huang X, He X, Zhou Y, Xu Y, Yu M, Huang S, Yan X, Li F, Pan J, Wang Y, Yu Y, Jin J. High PARP-1 expression predicts poor survival in acute myeloid leukemia and PARP-1 inhibitor and SAHA-bendamustine hybrid inhibitor combination treatment synergistically enhances anti-tumor effects. EBioMedicine 2018; 38:47-56. [PMID: 30472087 PMCID: PMC6306376 DOI: 10.1016/j.ebiom.2018.11.025] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 11/05/2018] [Accepted: 11/13/2018] [Indexed: 02/05/2023] Open
Abstract
Background PARP-1 plays a critical role in DNA damage repair and contributes to progression of cancer. To explore the role of PARP-1 in acute myeloid leukemia (AML), we analyzed the expression of PARP-1 in AML and its relation to the clinical prognosis. Then, we investigated the efficacy and mechanism of PARP inhibitor BMN673 (Talazoparib) combined with NL101, a novel SAHA-bendamustine hybrid in vitro and in vivo. Methods The expression of PARP-1 in 339 cytogenetically normal AML (CN-AML) cases was evaluated using RT-PCR. According to the expression of PARP-1, the clinical characteristics and prognosis of the patients were grouped and compared. The combination effects of BMN673 and NL101 were studied in AML cells and B-NSG mice xenograft model of MV4-11. Findings We found patients in high PARP-1 expression group had higher levels of blast cells in bone marrow (P = .003) and white blood cells (WBC) in peripheral blood (P = .008), and were associated with a more frequent FLT3-ITD mutation (28.2% vs 17.3%, P = .031). The overall survival (OS) and event free survival (EFS) of the high expression group were significantly shorter than those in the low expression group (OS, P = .005 and EFS, P = .004). BMN673 combined with NL101 had a strong synergistic effect in treating AML. The combination significantly induced cell apoptosis and arrested cell cycle in G2/M phase. Mechanistically, BMN673 and NL101 combinatorial treatment promoted DNA damage. In vivo, the combination effectively delayed the development of AML and prolonged survival. Interpretation High PARP-1 expression predicts poor survival in CN-AML patients. The synergistic effects of PARP inhibitor BMN673 in combination with SAHA-bendamustine hybrid, NL101, provide a new therapeutic strategy against AML. Fund National Natural Science Foundation of China and Zhejiang Provincial Key Innovation Team.
Collapse
Affiliation(s)
- Xia Li
- Department of Hematology, The First Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, PR China; Key Laboratory of Hematopoietic Malignancies, Diagnosis and Treatment, Zhejiang Province, PR China
| | - Chenying Li
- Department of Hematology, The First Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, PR China; Key Laboratory of Hematopoietic Malignancies, Diagnosis and Treatment, Zhejiang Province, PR China
| | - Jingrui Jin
- Department of Hematology, The First Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, PR China; Key Laboratory of Hematopoietic Malignancies, Diagnosis and Treatment, Zhejiang Province, PR China
| | - Jinghan Wang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, PR China
| | - Jiansong Huang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, PR China; Key Laboratory of Hematopoietic Malignancies, Diagnosis and Treatment, Zhejiang Province, PR China
| | - Zhixin Ma
- Department of Hematology, The First Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, PR China; Key Laboratory of Hematopoietic Malignancies, Diagnosis and Treatment, Zhejiang Province, PR China
| | - Xin Huang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, PR China; Key Laboratory of Hematopoietic Malignancies, Diagnosis and Treatment, Zhejiang Province, PR China
| | - Xiao He
- Department of Hematology, The First Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, PR China; Key Laboratory of Hematopoietic Malignancies, Diagnosis and Treatment, Zhejiang Province, PR China
| | - Yile Zhou
- Department of Hematology, The First Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, PR China; Key Laboratory of Hematopoietic Malignancies, Diagnosis and Treatment, Zhejiang Province, PR China
| | - Yu Xu
- Department of Hematology, The First Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, PR China
| | - Mengxia Yu
- Department of Hematology, Hangzhou First People's Hospital, Hangzhou, PR China
| | - Shujuan Huang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, PR China; Key Laboratory of Hematopoietic Malignancies, Diagnosis and Treatment, Zhejiang Province, PR China
| | - Xiao Yan
- Department of Hematology, The First Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, PR China; Key Laboratory of Hematopoietic Malignancies, Diagnosis and Treatment, Zhejiang Province, PR China
| | - Fenglin Li
- Department of Hematology, The First Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, PR China; Key Laboratory of Hematopoietic Malignancies, Diagnosis and Treatment, Zhejiang Province, PR China
| | - Jiajia Pan
- Department of Hematology, The First Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, PR China; Key Laboratory of Hematopoietic Malignancies, Diagnosis and Treatment, Zhejiang Province, PR China
| | - Yungui Wang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, PR China; Key Laboratory of Hematopoietic Malignancies, Diagnosis and Treatment, Zhejiang Province, PR China
| | - Yongping Yu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, PR China
| | - Jie Jin
- Department of Hematology, The First Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, PR China; Key Laboratory of Hematopoietic Malignancies, Diagnosis and Treatment, Zhejiang Province, PR China.
| |
Collapse
|
32
|
Vlahopoulos S, Adamaki M, Khoury N, Zoumpourlis V, Boldogh I. Roles of DNA repair enzyme OGG1 in innate immunity and its significance for lung cancer. Pharmacol Ther 2018; 194:59-72. [PMID: 30240635 DOI: 10.1016/j.pharmthera.2018.09.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cytokines are pivotal mediators of the immune response, and their coordinated expression protects host tissue from excessive damage and oxidant stress. Nevertheless, the development of lung pathology, including asthma, chronic obstructive pulmonary disease, and ozone-induced lung injury, is associated with oxidant stress; as evidence, there is a significant increase in levels of the modified guanine base 7,8-dihydro-8-oxoguanine (8-oxoG) in the genome. 8-OxoG is primarily recognized by 8-oxoguanine glycosylase 1 (OGG1), which catalyzes the first step in the DNA base excision repair pathway. However, oxidant stress in the cell transiently halts enzymatic activity of substrate-bound OGG1. The stalled OGG1 facilitates DNA binding of transactivators, including NF-κB, to their cognate sites to enable expression of cytokines and chemokines, with ensuing recruitments of inflammatory cells. Hence, defective OGG1 will modulate the coordination between innate and adaptive immunity through excessive oxidant stress and cytokine dysregulation. Both oxidant stress and cytokine dysregulation constitute key elements of oncogenesis by KRAS, which is mechanistically coupled to OGG1. Thus, analysis of the mechanism by which OGG1 modulates gene expression helps discern between beneficial and detrimental effects of oxidant stress, exposes a missing functional link as a marker, and yields a novel target for lung cancer.
Collapse
Affiliation(s)
- Spiros Vlahopoulos
- Ηoremeio Research Laboratory, First Department of Paediatrics, National and Kapodistrian University of Athens, 11527 Athens, Greece.
| | - Maria Adamaki
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 48 Vassileos Constantinou Avenue, 11635 Athens, Greece
| | - Nikolas Khoury
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 48 Vassileos Constantinou Avenue, 11635 Athens, Greece
| | - Vassilis Zoumpourlis
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 48 Vassileos Constantinou Avenue, 11635 Athens, Greece
| | - Istvan Boldogh
- Departments of Microbiology and Immunology and the Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, TX 77555, United States
| |
Collapse
|
33
|
Guffanti F, Fratelli M, Ganzinelli M, Bolis M, Ricci F, Bizzaro F, Chilà R, Sina FP, Fruscio R, Lupia M, Cavallaro U, Cappelletti MR, Generali D, Giavazzi R, Damia G. Platinum sensitivity and DNA repair in a recently established panel of patient-derived ovarian carcinoma xenografts. Oncotarget 2018; 9:24707-24717. [PMID: 29872499 PMCID: PMC5973859 DOI: 10.18632/oncotarget.25185] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 04/05/2018] [Indexed: 01/22/2023] Open
Abstract
A xenobank of patient-derived (PDX) ovarian tumor samples has been established consisting of tumors with different sensitivity to cisplatin (DDP), from very responsive to resistant. As the DNA repair pathway is an important driver in tumor response to DDP, we analyzed the mRNA expression of 20 genes involved in the nucleotide excision repair, fanconi anemia, homologous recombination, base excision repair, mismatch repair and translesion repair pathways and the methylation patterns of some of these genes. We also investigated the correlation with the response to platinum-based therapy. The mRNA levels of the selected genes were evaluated by Real Time-PCR (RT-PCR) with ad hoc validated primers and gene promoter methylation by pyrosequencing. All the DNA repair genes were variably expressed in all 42 PDX samples analyzed, with no particular histotype-specific pattern of expression. In high-grade serous/endometrioid PDXs, the CDK12 mRNA expression levels positively correlated with the expression of TP53BP1, PALB2, XPF and POLB. High-grade serous/endometrioid PDXs with TP53 mutations had significantly higher levels of POLQ, FANCD2, RAD51 and POLB than high-grade TP53 wild type PDXs. The mRNA levels of CDK12, PALB2 and XPF inversely associated with the in vivo DDP antitumor activity; higher CDK12 mRNA levels were associated with a higher recurrence rate in ovarian patients with low residual tumor. These data support the important role of CDK12 in the response to a platinum based therapy in ovarian patients.
Collapse
Affiliation(s)
- Federica Guffanti
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Maddalena Fratelli
- Department of Biochemistry, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Monica Ganzinelli
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Marco Bolis
- Department of Biochemistry, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Francesca Ricci
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Francesca Bizzaro
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Rosaria Chilà
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Federica Paola Sina
- Clinic of Obstetrics and Gynecology, San Gerardo Hospital, University of Milan-Bicocca, Department of Medicine and Surgery, Milan, Italy
| | - Robert Fruscio
- Clinic of Obstetrics and Gynecology, San Gerardo Hospital, University of Milan-Bicocca, Department of Medicine and Surgery, Milan, Italy
| | - Michela Lupia
- Unit of Gynecological Oncology Research, European Institute of Oncology, Milan, Italy
| | - Ugo Cavallaro
- Unit of Gynecological Oncology Research, European Institute of Oncology, Milan, Italy
| | | | - Daniele Generali
- Breast Cancer Unit and Translational Research Unit, ASST Cremona, Cremona, Italy.,Department of Medical, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Raffaella Giavazzi
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Giovanna Damia
- Department of Oncology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| |
Collapse
|
34
|
Laporte GA, Leguisamo NM, Kalil AN, Saffi J. Clinical importance of DNA repair in sporadic colorectal cancer. Crit Rev Oncol Hematol 2018; 126:168-185. [PMID: 29759559 DOI: 10.1016/j.critrevonc.2018.03.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 03/05/2018] [Accepted: 03/22/2018] [Indexed: 12/18/2022] Open
Abstract
Colorectal cancer (CRC) is the third major cause of cancer-related deaths worldwide. However, despite the scientific efforts to provide a molecular classification to improve CRC clinical practice management, prognosis and therapeutic decision are still strongly dependent on the TNM staging system. Mismatch repair system deficiencies can occur in many organs, but it is mainly a hallmark of CRC influencing clinical outcomes and response to therapy. This review will discuss the effect of the modulation of other DNA repair pathways (direct, excision and double strand break repairs) in the clinical and pathological aspects of colorectal cancer and its potential as prognostic and predictive biomarkers.
Collapse
Affiliation(s)
- Gustavo A Laporte
- Surgical Oncology Service, Santa Casa de Misericórdia de Porto Alegre (ISCMPA), Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Natalia M Leguisamo
- Institute of Cardiology/University Foundation of Cardiology, Porto Alegre, Rio Grande do Sul, Brazil; Laboratory of Genetic Toxicology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Antonio N Kalil
- Surgical Oncology Service, Santa Casa de Misericórdia de Porto Alegre (ISCMPA), Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Jenifer Saffi
- Laboratory of Genetic Toxicology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil.
| |
Collapse
|
35
|
Chen K, Li Y, Xu H, Zhang C, Li Z, Wang W, Wang B. WITHDRAWN: An analysis of the gene interaction networks identifying the role of PARP1 in metastasis of non-small cell lung cancer. Gene 2017:S0378-1119(17)30648-0. [PMID: 28843522 DOI: 10.1016/j.gene.2017.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 08/20/2017] [Indexed: 10/19/2022]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.
Collapse
Affiliation(s)
- Kai Chen
- Department of Respiratory Medicine, Baoji Central Hospital, Baoji 721008, Shaanxi, China
| | - Yajie Li
- Department of Cardiology, Baoji Central Hospital, Baoji 721008, Shaanxi, China.
| | - Hui Xu
- Department of Respiratory Medicine, Baoji Central Hospital, Baoji 721008, Shaanxi, China
| | - Chunfeng Zhang
- Department of Respiratory Medicine, Baoji Central Hospital, Baoji 721008, Shaanxi, China
| | - Zhiqiang Li
- Department of Respiratory Medicine, Baoji Central Hospital, Baoji 721008, Shaanxi, China
| | - Wei Wang
- Department of Respiratory Medicine, Baoji Central Hospital, Baoji 721008, Shaanxi, China
| | - Baofeng Wang
- Department of Respiratory Medicine, Baoji Central Hospital, Baoji 721008, Shaanxi, China
| |
Collapse
|
36
|
Chen K, Li Y, Xu H, Zhang C, Li Z, Wang W, Wang B. An analysis of the gene interaction networks identifying the role of PARP1 in metastasis of non-small cell lung cancer. Oncotarget 2017; 8:87263-87275. [PMID: 29152079 PMCID: PMC5675631 DOI: 10.18632/oncotarget.20256] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 07/12/2017] [Indexed: 01/09/2023] Open
Abstract
Background and Objective Though there were many researches about the effects of cancer cells on non-small cell lung cancer (NSCLC) currently, it has been rarely reported completed oncogene and its mechanism in tumors by far. Here, we used biological methods with known oncogene of NSCLC to find new oncogene and explore its functionary mechanism in NSCLC. Methods The study firstly built NSCLC genetic interaction network based on bioinformatics methods and then combined shortest path algorithm with significance test to confirmed core genes that were closely involved with given genes; real-time qPCR was conducted to detect expression levels between patients with NSCLC and normal people; additionally, detection of PARP1's role in migration and invasion was performed by trans-well assays and wound-healing. Results Through gene interaction network, it was found that, core genes like PARP1, EGFR and ALK had a direct interaction. TCGA database showed that PARP1 presented strong expression in NSCLC and the expression level of metastatic NSCLC was significantly higher than that of non-metastatic NSCLC. Cell migration of NSCLC in accordance to the scratch test was suppressed by PARP1 silence but stimulated noticeably by PARP1 overexpression. According to Kaplan-meier survival curve, the higher PARP1 expression, the poorer patient survival rate and prognosis. Thus, PARP1 expression had a negative correction with patient survival rate and prognosis. Conclusion New oncogene PARP1 was found from known NSCLC oncogene in terms of gene interaction network, demonstrating PARP1's impact on NSCLC cell migration.
Collapse
Affiliation(s)
- Kai Chen
- Department of Respiratory Medicine, Baoji Central Hospital, Baoji 721008, Shaanxi, China
| | - Yajie Li
- Department of Cardiology, Baoji Central Hospital, Baoji 721008, Shaanxi, China
| | - Hui Xu
- Department of Respiratory Medicine, Baoji Central Hospital, Baoji 721008, Shaanxi, China
| | - Chunfeng Zhang
- Department of Respiratory Medicine, Baoji Central Hospital, Baoji 721008, Shaanxi, China
| | - Zhiqiang Li
- Department of Respiratory Medicine, Baoji Central Hospital, Baoji 721008, Shaanxi, China
| | - Wei Wang
- Department of Respiratory Medicine, Baoji Central Hospital, Baoji 721008, Shaanxi, China
| | - Baofeng Wang
- Department of Respiratory Medicine, Baoji Central Hospital, Baoji 721008, Shaanxi, China
| |
Collapse
|
37
|
Hong MY, Hoh E, Kang B, DeHamer R, Kim JY, Lumibao J. Fish Oil Contaminated with Persistent Organic Pollutants Induces Colonic Aberrant Crypt Foci Formation and Reduces Antioxidant Enzyme Gene Expression in Rats. J Nutr 2017; 147:1524-1530. [PMID: 28659405 PMCID: PMC5525110 DOI: 10.3945/jn.117.251082] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 03/28/2017] [Accepted: 06/05/2017] [Indexed: 12/16/2022] Open
Abstract
Background: Epidemiologic, clinical, and experimental studies have suggested that fish oil (FO), a rich source of n-3 (ω-3) polyunsaturated fatty acids, protects against colon cancer. However, this message is confounded by the FDA's warning that the consumption of certain types of fish should be restricted because of contamination with persistent organic pollutants (POPs), such as polychlorinated biphenyls (PCBs) and organochlorine pesticides.Objective: We examined FO contaminated with POPs (PCBs, dichlorodiphenyltrichloroethane, and chlordane) compared with unmodified FO on the risk factors of colon cancer development.Methods: Male Sprague-Dawley rats aged 28 d (n = 30) were allocated into 3 groups and fed 15% corn oil (CO), FO, or POP-contaminated FO for 9 wk with a subcutaneous injection of colon carcinogen azoxymethane at weeks 3 and 4. Colonic aberrant crypt foci (ACF) and cell proliferation were enumerated, and the gene expression of inflammation, antioxidant enzymes, and repair enzymes were determined with the use of real-time quantitative polymerase chain reaction analysis.Results: FO-fed rats had a lower number of ACF (mean ± SE: 29 ± 4.0 for FO compared with 53 ± 8.4 for CO and 44 ± 4.6 for POP FO) and higher-multiplicity ACF than the CO and POP FO groups (4.7 ± 0.9 for FO compared with 11 ± 1.5 for CO and 9.6 ± 1.8 for POP FO) (P < 0.05). FO feeding lowered the proliferation index compared with the CO and POP FO feeding groups (18% ± 1.1% for FO compared with 25% ± 1.6% for CO and 23% ± 0.7% for POP FO) (P = 0.009). Superoxide dismutase [2.4 ± 0.6 relative quantification (RQ) for FO compared with 1.2 ± 0.2 RQ for CO and 1.3 ± 0.3 RQ for POP FO] and catalase gene expression (10 ± 2.0 RQ for FO compared with 5.4 ± 1.1 RQ for CO and 6.6 ± 1.5 RQ for POP FO) were higher in the FO group than in the CO and POP FO groups (P < 0.05). There were no differences between CO and POP FO on the variables.Conclusion: These results indicate that POPs in FO reduce the preventive effects of FO on colon carcinogenesis by increasing preneoplastic lesion formation through the downregulation of antioxidant enzyme expression and increasing cell proliferation in rats.
Collapse
Affiliation(s)
| | - Eunha Hoh
- Graduate School of Public Health, San Diego State University, San Diego, CA; and
| | - Brian Kang
- School of Exercise and Nutritional Sciences and
| | | | - Jin Young Kim
- Department of Food Science and Engineering, Ewha Womans University, Seoul, South Korea
| | - Jan Lumibao
- School of Exercise and Nutritional Sciences and
| |
Collapse
|
38
|
Rajawat J, Shukla N, Mishra DP. Therapeutic Targeting of Poly(ADP-Ribose) Polymerase-1 (PARP1) in Cancer: Current Developments, Therapeutic Strategies, and Future Opportunities. Med Res Rev 2017; 37:1461-1491. [PMID: 28510338 DOI: 10.1002/med.21442] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 01/31/2017] [Accepted: 02/16/2017] [Indexed: 12/16/2022]
Abstract
Poly(ADP-ribose) polymerase-1 (PARP-1) plays a central role in numerous cellular processes including DNA repair, replication, and transcription. PARP interacts directly, indirectly or via PARylation with various oncogenic proteins and regulates several transcription factors thereby modulating carcinogenesis. Therapeutic inhibition of PARP is therefore perceived as a promising anticancer strategy and a number of PARP inhibitors (PARPi) are currently under development and clinical evaluation. PARPi inhibit the DNA repair pathway and thus form the concept of synthetic lethality in cancer therapeutics. Preclinical and clinical studies have shown the potential of PARPi as chemopotentiator, radiosensitizer, or as adjuvant therapeutic agents. Recent studies have shown that PARP-1 could be either oncogenic or tumor suppressive in different cancers. PARP inhibitor resistance is also a growing concern in the clinical setting. Recently, changes in the levels of PARP-1 activity or expression in cancer patients have provided the basis for consideration of PARP-1 regulatory proteins as potential biomarkers. This review focuses on the current developments related to the role of PARP in cancer progression, therapeutic strategies targeting PARP-associated oncogenic signaling, and future opportunities in use of PARPi in anticancer therapeutics.
Collapse
Affiliation(s)
- Jyotika Rajawat
- Cell Death Research Laboratory, Endocrinology Division, CSIR-Central Drug Research Institute, B.S. 10/1, Sector-10, Jankipuram Extension, Lucknow, Uttar Pradesh, 226031, India
| | - Nidhi Shukla
- Cell Death Research Laboratory, Endocrinology Division, CSIR-Central Drug Research Institute, B.S. 10/1, Sector-10, Jankipuram Extension, Lucknow, Uttar Pradesh, 226031, India
| | - Durga Prasad Mishra
- Cell Death Research Laboratory, Endocrinology Division, CSIR-Central Drug Research Institute, B.S. 10/1, Sector-10, Jankipuram Extension, Lucknow, Uttar Pradesh, 226031, India
| |
Collapse
|
39
|
Zhang S, Li J, Li Y, Liu Y, Guo H, Xu X. Nitric Oxide Synthase Activity Correlates with OGG1 in Ozone-Induced Lung Injury Animal Models. Front Physiol 2017; 8:249. [PMID: 28496412 PMCID: PMC5406453 DOI: 10.3389/fphys.2017.00249] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 04/07/2017] [Indexed: 12/12/2022] Open
Abstract
Background: NO is an important cellular signaling molecule which is derived from L-arginine by nitric oxide synthase (NOS) and the effects of NOS signaling in lung injury is conflicting. The present study was designed to observe the effect of NOS and Arginase signaling in the occurrence and development of lung injury and its mechanism. Methods: An ozone-stressed lung injury animal model was established by exposure to 2.0 ppm O3 for 30 min every day for consecutive 12 day with or without the administration of NO precursor L-arginine or non-selective NOS inhibitor N-nitro-L-arginine methyl ester (L-NAME). Then, the lung histopathology, the releases of inflammatory mediators and the production of ROS were assayed by immunohistochemistry, ELISA and flow cytometry respectively. The activities and expression of NOS and Arginase were assayed by biochemical methods and western blot. Correspondingly, the release of 8-oxoguanine glycosylase 1(8-OxoG) and 8-oxoguanine glycosylase 1 (OGG1) were assayed by ELISA and western blot. The correlation between NOS/Arginase signaling with 8-OxoG/ OGG1 was also analyzed by Pearson correlation coefficients and immunofluorescence in NOS deficient bronchial epithelial cells. Results: In ozone-induced rat lung injury models, lung inflammation as well as lung architecture was disrupted in a time dependent manner. Ozone treatment with L-arginine showed a substantial attenuation of adverse lung histopathological changes and treatment with L-NAME promoted the inflammation and remodeling. Importantly, the expression of NOS was promoted by L-arginine and inhibited by L-NAME and the expression of Arginase was promoted by L-NAME treatment. Further, we observed significantly higher levels of 8-OxoG and lower levels of OGG1 in ozone group which was reversed by L-arginine and promoted by L-NAME. The expression of NOS is closely related with 8-OxoG /OCG1. Conclusion: These findings give further evidence that the NOS signaling is related with base excise repair.
Collapse
Affiliation(s)
- Suqin Zhang
- Department of Pediatrics, First Affiliated Hospital of Zhengzhou UniversityZhengzhou, China
| | - Jianhua Li
- Department of General Surgery, First Affiliated Hospital of Zhengzhou UniversityZhengzhou, China
| | - Yuqin Li
- Department of Pediatrics, First Affiliated Hospital of Zhengzhou UniversityZhengzhou, China
| | - Yufeng Liu
- Department of Pediatrics, First Affiliated Hospital of Zhengzhou UniversityZhengzhou, China
| | - Hongxiang Guo
- Department of Pediatrics, First Affiliated Hospital of Zhengzhou UniversityZhengzhou, China
| | - Xiaoli Xu
- Department of Pediatrics, First Affiliated Hospital of Zhengzhou UniversityZhengzhou, China
| |
Collapse
|
40
|
Wiśniewska-Chudy E, Szylberg Ł, Dworacki G, Mizera-Nyczak E, Marszałek A. pSTAT5 and ERK exhibit different expression in myeloproliferative neoplasms. Oncol Rep 2017; 37:2295-2307. [DOI: 10.3892/or.2017.5476] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 12/02/2016] [Indexed: 11/06/2022] Open
|
41
|
Li Z, Lv T, Liu Y, Huang X, Qiu Z, Li J. PARP1 is a novel independent prognostic factor for the poor prognosis of chordoma. Cancer Biomark 2017; 16:633-9. [PMID: 27002766 DOI: 10.3233/cbm-160605] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVE To evaluate the expression of PARP1 in chordoma and analyzed its association with clinical factors and patients' prognosis. METHODS The expression of Poly (ADP-ribose) polymerase 1 (PARP1) in chordoma specimens from 74 chordoma patients (50 primary and 24 recurrent tumors of 50 patients)and 20 distant normal tissue specimens was measured by immunohistochemical staining. The association of PARP1with the clinical factors and patients' prognosis was also analyzed. RESULTS Of all the chordoma samples, 78% showed high expression of PARP1, whereas, only 10% of distant normal tissues expressed a high level of PARP1 (p< 0.01). Chi-square analysis revealed that high expression of PARP1 was significantly correlated with tumor recurrence (p< 0.01) and invasion into surrounding muscle (p< 0.01), while the data did not indicate any association with patients' gender, age, tumor location and size (p> 0.05). Kaplan-Meier survival curve and log-rank test showed that continuous disease free survival time (CDFS) was significantly shorter in the PARP1-positive group than in the PARP1-negative group (P= 0.019). CONCLUSION High expression of PARP1 is significantly associated with chordoma invasion and recurrence. PARP1 may become a potential biomarker for chordoma in predicting its recurrence and patients' prognosis.
Collapse
|
42
|
Szebeni A, Szebeni K, DiPeri TP, Johnson LA, Stockmeier CA, Crawford JD, Chandley MJ, Hernandez LJ, Burgess KC, Brown RW, Ordway GA. Elevated DNA Oxidation and DNA Repair Enzyme Expression in Brain White Matter in Major Depressive Disorder. Int J Neuropsychopharmacol 2016; 20:363-373. [PMID: 28034960 PMCID: PMC5412018 DOI: 10.1093/ijnp/pyw114] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 12/20/2016] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Pathology of white matter in brains of patients with major depressive disorder (MDD) is well-documented, but the cellular and molecular basis of this pathology are poorly understood. METHODS Levels of DNA oxidation and gene expression of DNA damage repair enzymes were measured in Brodmann area 10 (BA10) and/or amygdala (uncinate fasciculus) white matter tissue from brains of MDD (n=10) and psychiatrically normal control donors (n=13). DNA oxidation was also measured in BA10 white matter of schizophrenia donors (n=10) and in prefrontal cortical white matter from control rats (n=8) and rats with repeated stress-induced anhedonia (n=8). RESULTS DNA oxidation in BA10 white matter was robustly elevated in MDD as compared to control donors, with a smaller elevation occurring in schizophrenia donors. DNA oxidation levels in psychiatrically affected donors that died by suicide did not significantly differ from DNA oxidation levels in psychiatrically affected donors dying by other causes (non-suicide). Gene expression levels of two base excision repair enzymes, PARP1 and OGG1, were robustly elevated in oligodendrocytes laser captured from BA10 and amygdala white matter of MDD donors, with smaller but significant elevations of these gene expressions in astrocytes. In rats, repeated stress-induced anhedonia, as measured by a reduction in sucrose preference, was associated with increased DNA oxidation in white, but not gray, matter. CONCLUSIONS Cellular residents of brain white matter demonstrate markers of oxidative damage in MDD. Medications that interfere with oxidative damage or pathways activated by oxidative damage have potential to improve treatment for MDD.
Collapse
Affiliation(s)
- Attila Szebeni
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee (Drs A. Szebeni and K. Szebeni, Mr DiPeri, Mr Johnson, Dr Crawford, Ms Hernandez, Dr Brown, and Ms Burgess); Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, Mississippi (Dr Stockmeier); Department of Psychiatry, Case Western Reserve University, Cleveland, Ohio (Dr Stockmeier); Departments of Health Sciences & Biomedical Sciences (Dr Chandley), and Departments of Biomedical Sciences & Psychiatry and Behavioral Sciences (Dr Ordway), East Tennessee State University, Johnson City, Tennessee
| | - Katalin Szebeni
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee (Drs A. Szebeni and K. Szebeni, Mr DiPeri, Mr Johnson, Dr Crawford, Ms Hernandez, Dr Brown, and Ms Burgess); Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, Mississippi (Dr Stockmeier); Department of Psychiatry, Case Western Reserve University, Cleveland, Ohio (Dr Stockmeier); Departments of Health Sciences & Biomedical Sciences (Dr Chandley), and Departments of Biomedical Sciences & Psychiatry and Behavioral Sciences (Dr Ordway), East Tennessee State University, Johnson City, Tennessee
| | - Timothy P. DiPeri
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee (Drs A. Szebeni and K. Szebeni, Mr DiPeri, Mr Johnson, Dr Crawford, Ms Hernandez, Dr Brown, and Ms Burgess); Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, Mississippi (Dr Stockmeier); Department of Psychiatry, Case Western Reserve University, Cleveland, Ohio (Dr Stockmeier); Departments of Health Sciences & Biomedical Sciences (Dr Chandley), and Departments of Biomedical Sciences & Psychiatry and Behavioral Sciences (Dr Ordway), East Tennessee State University, Johnson City, Tennessee
| | - Luke A. Johnson
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee (Drs A. Szebeni and K. Szebeni, Mr DiPeri, Mr Johnson, Dr Crawford, Ms Hernandez, Dr Brown, and Ms Burgess); Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, Mississippi (Dr Stockmeier); Department of Psychiatry, Case Western Reserve University, Cleveland, Ohio (Dr Stockmeier); Departments of Health Sciences & Biomedical Sciences (Dr Chandley), and Departments of Biomedical Sciences & Psychiatry and Behavioral Sciences (Dr Ordway), East Tennessee State University, Johnson City, Tennessee
| | - Craig A. Stockmeier
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee (Drs A. Szebeni and K. Szebeni, Mr DiPeri, Mr Johnson, Dr Crawford, Ms Hernandez, Dr Brown, and Ms Burgess); Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, Mississippi (Dr Stockmeier); Department of Psychiatry, Case Western Reserve University, Cleveland, Ohio (Dr Stockmeier); Departments of Health Sciences & Biomedical Sciences (Dr Chandley), and Departments of Biomedical Sciences & Psychiatry and Behavioral Sciences (Dr Ordway), East Tennessee State University, Johnson City, Tennessee
| | - Jessica D. Crawford
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee (Drs A. Szebeni and K. Szebeni, Mr DiPeri, Mr Johnson, Dr Crawford, Ms Hernandez, Dr Brown, and Ms Burgess); Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, Mississippi (Dr Stockmeier); Department of Psychiatry, Case Western Reserve University, Cleveland, Ohio (Dr Stockmeier); Departments of Health Sciences & Biomedical Sciences (Dr Chandley), and Departments of Biomedical Sciences & Psychiatry and Behavioral Sciences (Dr Ordway), East Tennessee State University, Johnson City, Tennessee
| | - Michelle J. Chandley
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee (Drs A. Szebeni and K. Szebeni, Mr DiPeri, Mr Johnson, Dr Crawford, Ms Hernandez, Dr Brown, and Ms Burgess); Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, Mississippi (Dr Stockmeier); Department of Psychiatry, Case Western Reserve University, Cleveland, Ohio (Dr Stockmeier); Departments of Health Sciences & Biomedical Sciences (Dr Chandley), and Departments of Biomedical Sciences & Psychiatry and Behavioral Sciences (Dr Ordway), East Tennessee State University, Johnson City, Tennessee
| | - Liza J. Hernandez
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee (Drs A. Szebeni and K. Szebeni, Mr DiPeri, Mr Johnson, Dr Crawford, Ms Hernandez, Dr Brown, and Ms Burgess); Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, Mississippi (Dr Stockmeier); Department of Psychiatry, Case Western Reserve University, Cleveland, Ohio (Dr Stockmeier); Departments of Health Sciences & Biomedical Sciences (Dr Chandley), and Departments of Biomedical Sciences & Psychiatry and Behavioral Sciences (Dr Ordway), East Tennessee State University, Johnson City, Tennessee
| | - Katherine C. Burgess
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee (Drs A. Szebeni and K. Szebeni, Mr DiPeri, Mr Johnson, Dr Crawford, Ms Hernandez, Dr Brown, and Ms Burgess); Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, Mississippi (Dr Stockmeier); Department of Psychiatry, Case Western Reserve University, Cleveland, Ohio (Dr Stockmeier); Departments of Health Sciences & Biomedical Sciences (Dr Chandley), and Departments of Biomedical Sciences & Psychiatry and Behavioral Sciences (Dr Ordway), East Tennessee State University, Johnson City, Tennessee
| | - Russell W. Brown
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee (Drs A. Szebeni and K. Szebeni, Mr DiPeri, Mr Johnson, Dr Crawford, Ms Hernandez, Dr Brown, and Ms Burgess); Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, Mississippi (Dr Stockmeier); Department of Psychiatry, Case Western Reserve University, Cleveland, Ohio (Dr Stockmeier); Departments of Health Sciences & Biomedical Sciences (Dr Chandley), and Departments of Biomedical Sciences & Psychiatry and Behavioral Sciences (Dr Ordway), East Tennessee State University, Johnson City, Tennessee
| | - Gregory A. Ordway
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee (Drs A. Szebeni and K. Szebeni, Mr DiPeri, Mr Johnson, Dr Crawford, Ms Hernandez, Dr Brown, and Ms Burgess); Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, Mississippi (Dr Stockmeier); Department of Psychiatry, Case Western Reserve University, Cleveland, Ohio (Dr Stockmeier); Departments of Health Sciences & Biomedical Sciences (Dr Chandley), and Departments of Biomedical Sciences & Psychiatry and Behavioral Sciences (Dr Ordway), East Tennessee State University, Johnson City, Tennessee
| |
Collapse
|
43
|
Kossatz S, Brand C, Gutiontov S, Liu JTC, Lee NY, Gönen M, Weber WA, Reiner T. Detection and delineation of oral cancer with a PARP1 targeted optical imaging agent. Sci Rep 2016; 6:21371. [PMID: 26900125 PMCID: PMC4761964 DOI: 10.1038/srep21371] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 01/20/2016] [Indexed: 12/01/2022] Open
Abstract
Earlier and more accurate detection of oral squamous cell carcinoma (OSCC) is essential to improve the prognosis of patients and to reduce the morbidity of surgical therapy. Here, we demonstrate that the nuclear enzyme Poly(ADP-ribose)Polymerase 1 (PARP1) is a promising target for optical imaging of OSCC with the fluorescent dye PARPi-FL. In patient-derived OSCC specimens, PARP1 expression was increased 7.8 ± 2.6-fold when compared to normal tissue. Intravenous injection of PARPi-FL allowed for high contrast in vivo imaging of human OSCC models in mice with a surgical fluorescence stereoscope and high-resolution imaging systems. The emitted signal was specific for PARP1 expression and, most importantly, PARPi-FL can be used as a topical imaging agent, spatially resolving the orthotopic tongue tumors in vivo. Collectively, our results suggest that PARP1 imaging with PARPi-FL can enhance the detection of oral cancer, serve as a screening tool and help to guide surgical resections.
Collapse
Affiliation(s)
- Susanne Kossatz
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Christian Brand
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Stanley Gutiontov
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jonathan T C Liu
- Department of Mechanical Engineering, University of Washington, Seattle, WA 98195, USA
| | - Nancy Y Lee
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Mithat Gönen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Wolfgang A Weber
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Department of Radiology, Weill Cornell Medical College, New York, NY 10065, USA.,Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Department of Radiology, Weill Cornell Medical College, New York, NY 10065, USA
| |
Collapse
|
44
|
Kossatz S, Weber WA, Reiner T. Optical Imaging of PARP1 in Response to Radiation in Oral Squamous Cell Carcinoma. PLoS One 2016; 11:e0147752. [PMID: 26808835 PMCID: PMC4726809 DOI: 10.1371/journal.pone.0147752] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 01/07/2016] [Indexed: 12/15/2022] Open
Abstract
Targeting and inhibiting DNA repair pathways is a powerful strategy of controlling malignant growth. One such strategy includes the inhibition of PARP1, a central element in the intracellular DNA damage response. To determine and visualize the expression and intercellular distribution of PARP1 in vivo, and to monitor the pharmacokinetics of PARP1 targeted therapeutics, fluorescent small probes were developed. To date, however, it is unclear how these probes behave in a more realistic clinical setting, where DNA damage has been induced through one or more prior lines of therapy. Here, we use one such imaging agent, PARPi-FL, in tissues both with and without prior DNA damage, and investigate its value as a probe for PARP1 imaging. We show that PARP1 expression in oral cancer is high, and that the uptake of PARPi-FL is selective, irrespective of whether cells were exposed to irradiation or not. We also show that PARPi-FL uptake increases in response to DNA damage, and that this increase is reflected in higher enzyme expression. Our findings provide a framework for measuring exposure of cells to external beam radiation, and could help to elucidate the effects of such treatments non-invasively in mouse models of cancer.
Collapse
Affiliation(s)
- Susanne Kossatz
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, 10065, United States of America
| | - Wolfgang A. Weber
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, 10065, United States of America
- Department of Radiology, Weill Cornell Medical College, New York, New York, 10065, United States of America
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, 10065, United States of America
- Department of Radiology, Weill Cornell Medical College, New York, New York, 10065, United States of America
| |
Collapse
|
45
|
Donley N, Jaruga P, Coskun E, Dizdaroglu M, McCullough AK, Lloyd RS. Small Molecule Inhibitors of 8-Oxoguanine DNA Glycosylase-1 (OGG1). ACS Chem Biol 2015. [PMID: 26218629 DOI: 10.1021/acschembio.5b00452] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The DNA base excision repair (BER) pathway, which utilizes DNA glycosylases to initiate repair of specific DNA lesions, is the major pathway for the repair of DNA damage induced by oxidation, alkylation, and deamination. Early results from clinical trials suggest that inhibiting certain enzymes in the BER pathway can be a useful anticancer strategy when combined with certain DNA-damaging agents or tumor-specific genetic deficiencies. Despite this general validation of BER enzymes as drug targets, there are many enzymes that function in the BER pathway that have few, if any, specific inhibitors. There is a growing body of evidence that suggests inhibition of 8-oxoguanine DNA glycosylase-1 (OGG1) could be useful as a monotherapy or in combination therapy to treat certain types of cancer. To identify inhibitors of OGG1, a fluorescence-based screen was developed to analyze OGG1 activity in a high-throughput manner. From a primary screen of ∼50,000 molecules, 13 inhibitors were identified, 12 of which were hydrazides or acyl hydrazones. Five inhibitors with an IC50 value of less than 1 μM were chosen for further experimentation and verified using two additional biochemical assays. None of the five OGG1 inhibitors reduced DNA binding of OGG1 to a 7,8-dihydro-8-oxoguanine (8-oxo-Gua)-containing substrate, but all five inhibited Schiff base formation during OGG1-mediated catalysis. All of these inhibitors displayed a >100-fold selectivity for OGG1 relative to several other DNA glycosylases involved in repair of oxidatively damaged bases. These inhibitors represent the most potent and selective OGG1 inhibitors identified to date.
Collapse
Affiliation(s)
- Nathan Donley
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, Oregon 97239, United States
| | - Pawel Jaruga
- Biomolecular
Measurement Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
| | - Erdem Coskun
- Biomolecular
Measurement Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
| | - Miral Dizdaroglu
- Biomolecular
Measurement Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
| | - Amanda K. McCullough
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, Oregon 97239, United States
| | - R. Stephen Lloyd
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, Oregon 97239, United States
| |
Collapse
|