1
|
Nzitakera A, Surwumwe JB, Ndoricyimpaye EL, Uwamungu S, Uwamariya D, Manirakiza F, Ndayisaba MC, Ntakirutimana G, Seminega B, Dusabejambo V, Rutaganda E, Kamali P, Ngabonziza F, Ishikawa R, Rugwizangoga B, Iwashita Y, Yamada H, Yoshimura K, Sugimura H, Shinmura K. The spectrum of TP53 mutations in Rwandan patients with gastric cancer. Genes Environ 2024; 46:8. [PMID: 38459566 PMCID: PMC10921722 DOI: 10.1186/s41021-024-00302-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 02/18/2024] [Indexed: 03/10/2024] Open
Abstract
BACKGROUND Gastric cancer is the sixth most frequently diagnosed cancer and third in causing cancer-related death globally. The most frequently mutated gene in human cancers is TP53, which plays a pivotal role in cancer initiation and progression. In Africa, particularly in Rwanda, data on TP53 mutations are lacking. Therefore, this study intended to obtain TP53 mutation status in Rwandan patients with gastric cancer. RESULTS Formalin-fixed paraffin-embedded tissue blocks of 95 Rwandan patients with histopathologically proven gastric carcinoma were obtained from the University Teaching Hospital of Kigali. After DNA extraction, all coding regions of the TP53 gene and the exon-intron boundary region of TP53 were sequenced using the Sanger sequencing. Mutated TP53 were observed in 24 (25.3%) of the 95 cases, and a total of 29 mutations were identified. These TP53 mutations were distributed between exon 4 and 8 and most of them were missense mutations (19/29; 65.5%). Immunohistochemical analysis for TP53 revealed that most of the TP53 missense mutations were associated with TP53 protein accumulation. Among the 29 mutations, one was novel (c.459_477delCGGCACCCGCGTCCGCGCC). This 19-bp deletion mutation in exon 5 caused the production of truncated TP53 protein (p.G154Wfs*10). Regarding the spectrum of TP53 mutations, G:C > A:T at CpG sites was the most prevalent (10/29; 34.5%) and G:C > T:A was the second most prevalent (7/29; 24.1%). Interestingly, when the mutation spectrum of TP53 was compared to three previous TP53 mutational studies on non-Rwandan patients with gastric cancer, G:C > T:A mutations were significantly more frequent in this study than in our previous study (p = 0.013), the TCGA database (p = 0.017), and a previous study on patients from Hong Kong (p = 0.006). Even after correcting for false discovery, statistical significance was observed. CONCLUSIONS Our results suggested that TP53 G:C > T:A transversion mutation in Rwandan patients with gastric cancer is more frequent than in non-Rwandan patients with gastric cancer, indicating at an alternative etiological and carcinogenic progression of gastric cancer in Rwanda.
Collapse
Affiliation(s)
- Augustin Nzitakera
- Department of Tumor Pathology, Hamamatsu University School of Medicine (HUSM), 1-20-1 Handayama, Higashi-Ku, Hamamatsu, Shizuoka, 431-3192, Japan
- Department of Biomedical Laboratory Sciences, School of Health Sciences, College of Medicine and Health Sciences, University of Rwanda, P.O. Box 3286, Kigali, Rwanda
| | - Jean Bosco Surwumwe
- Department of Pathology, University Teaching Hospital of Kigali, P.O. Box 655, Kigali, Rwanda
| | - Ella Larissa Ndoricyimpaye
- Department of Biomedical Laboratory Sciences, School of Health Sciences, College of Medicine and Health Sciences, University of Rwanda, P.O. Box 3286, Kigali, Rwanda
- Université Catholique de Louvain, Médecine Expérimentale, Brussels, 1348, Belgium
| | - Schifra Uwamungu
- Department of Biomedical Laboratory Sciences, School of Health Sciences, College of Medicine and Health Sciences, University of Rwanda, P.O. Box 3286, Kigali, Rwanda
- Department of Pharmacology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, SE-40530, Sweden
| | - Delphine Uwamariya
- Department of Biomedical Laboratory Sciences, School of Health Sciences, College of Medicine and Health Sciences, University of Rwanda, P.O. Box 3286, Kigali, Rwanda
- Department of Pathology, University Teaching Hospital of Kigali, P.O. Box 655, Kigali, Rwanda
- Department of Pathology, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, P.O. Box 3286, Kigali, Rwanda
| | - Felix Manirakiza
- Department of Tumor Pathology, Hamamatsu University School of Medicine (HUSM), 1-20-1 Handayama, Higashi-Ku, Hamamatsu, Shizuoka, 431-3192, Japan
- Department of Pathology, University Teaching Hospital of Kigali, P.O. Box 655, Kigali, Rwanda
- Department of Pathology, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, P.O. Box 3286, Kigali, Rwanda
| | - Marie Claire Ndayisaba
- Department of Pathology, University Teaching Hospital of Kigali, P.O. Box 655, Kigali, Rwanda
- Department of Pathology, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, P.O. Box 3286, Kigali, Rwanda
| | - Gervais Ntakirutimana
- Department of Pathology, University Teaching Hospital of Kigali, P.O. Box 655, Kigali, Rwanda
- Department of Pathology, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, P.O. Box 3286, Kigali, Rwanda
| | - Benoit Seminega
- Department of Pathology, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, P.O. Box 3286, Kigali, Rwanda
- Department of Internal Medicine, University Teaching Hospital of Kigali, P.O. Box 655, Kigali, Rwanda
| | - Vincent Dusabejambo
- Department of Pathology, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, P.O. Box 3286, Kigali, Rwanda
- Department of Internal Medicine, University Teaching Hospital of Kigali, P.O. Box 655, Kigali, Rwanda
| | - Eric Rutaganda
- Department of Pathology, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, P.O. Box 3286, Kigali, Rwanda
- Department of Internal Medicine, University Teaching Hospital of Kigali, P.O. Box 655, Kigali, Rwanda
| | - Placide Kamali
- Department of Pathology, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, P.O. Box 3286, Kigali, Rwanda
- Department of Internal Medicine, University Teaching Hospital of Kigali, P.O. Box 655, Kigali, Rwanda
| | - François Ngabonziza
- Department of Pathology, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, P.O. Box 3286, Kigali, Rwanda
- Department of Internal Medicine, University Teaching Hospital of Kigali, P.O. Box 655, Kigali, Rwanda
| | - Rei Ishikawa
- Department of Tumor Pathology, Hamamatsu University School of Medicine (HUSM), 1-20-1 Handayama, Higashi-Ku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Belson Rugwizangoga
- Department of Pathology, University Teaching Hospital of Kigali, P.O. Box 655, Kigali, Rwanda
- Department of Pathology, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, P.O. Box 3286, Kigali, Rwanda
| | - Yuji Iwashita
- Department of Tumor Pathology, Hamamatsu University School of Medicine (HUSM), 1-20-1 Handayama, Higashi-Ku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Hidetaka Yamada
- Department of Tumor Pathology, Hamamatsu University School of Medicine (HUSM), 1-20-1 Handayama, Higashi-Ku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Kimio Yoshimura
- Department of Health Policy and Management, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Haruhiko Sugimura
- Department of Tumor Pathology, Hamamatsu University School of Medicine (HUSM), 1-20-1 Handayama, Higashi-Ku, Hamamatsu, Shizuoka, 431-3192, Japan.
- Sasaki Institute Sasaki Foundation, 2-2 Kanda Surugadai, Chiyoda-Ku, Tokyo, 101-0062, Japan.
| | - Kazuya Shinmura
- Department of Tumor Pathology, Hamamatsu University School of Medicine (HUSM), 1-20-1 Handayama, Higashi-Ku, Hamamatsu, Shizuoka, 431-3192, Japan.
| |
Collapse
|
2
|
Resveratrol and Quercetin Potentiate the Cell Protection and Rescue Effects of NAD + Precursors in HEK293 Cells Challenged by DNA Damaging Agent, N-Methyl- N′-nitro- N-nitrosoguanidine. Nat Prod Commun 2021. [DOI: 10.1177/1934578x211045465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
DNA damage plays an essential role in the human ageing process. Recently, accumulating evidence has demonstrated that a decreased level of nicotinamide adenine dinucleotide (NAD+) is involved in human ageing and suggested that the natural supplements of NAD+ precursors and its homeostasis regulators might serve as a promising modality to slow down the human ageing process. In the present study, we analyzed the combinational effects and potential mechanism of NAD+ precursors, nicotinic acid (NA) and nicotinamide (NM), and the NAD+’ homeostasis regulators, resveratrol (R), and quercetin (Q) in the protection and rescue of HEK293 cells from N-methyl- N'-nitro- N nitrosoguanidine (MNNG)-induced DNA damage. The results indicate that resveratrol and quercetin can significantly potentiate the cell protection and rescue effects of NAD+ precursors in HEK293 cells attacked by the DNA damaging agent, MNNG. Intracellular NAD+ homeostasis and the PARP1 activation status are the key factors in determining the fate of the cells under DNA damaging stress.
Collapse
|
3
|
Sassa A, Fukuda T, Ukai A, Nakamura M, Sato R, Fujiwara S, Hirota K, Takeda S, Sugiyama KI, Honma M, Yasui M. Follow-up genotoxicity assessment of Ames-positive/equivocal chemicals using the improved thymidine kinase gene mutation assay in DNA repair-deficient human TK6 cells. Mutagenesis 2021; 36:331-338. [PMID: 34216473 DOI: 10.1093/mutage/geab025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 07/02/2021] [Indexed: 11/14/2022] Open
Abstract
Genotoxicity testing plays an important role in the safety assessment of pharmaceuticals, pesticides, and chemical substances. Among the guidelines for various genotoxicity tests, the in vitro genotoxicity test battery comprises the bacterial Ames test and mammalian cell assays. Several chemicals exhibit conflicting results for the bacterial Ames test and mammalian cell genotoxicity studies, which may stem from the differences in DNA repair capacity or metabolism, between different cell types or species. For better understanding the mechanistic implications regarding conflict outcomes between different assay systems, it is necessary to develop in vitro genotoxicity testing approaches with higher specificity towards DNA-damaging reagents. We have recently established an improved thymidine kinase (TK) gene mutation assay (TK assay) that is deficient in DNA excision repair system using human lymphoblastoid TK6 cells lacking XRCC1 and XPA (XRCC1 -/-/XPA -/-), the core factors of base excision repair and nucleotide excision repair, respectively. This DNA repair-deficient TK6 cell line is expected to specifically evaluate the genotoxic potential of chemical substances based on the DNA damage. We focused on four reagents, N-(1-naphthyl)ethylenediamine dihydrochloride (NEDA), p-phenylenediamine (PPD), auramine, and malachite green (MG) as the Ames test-positive chemicals. In our assay, assessment using XRCC1 -/-/XPA -/- cells revealed no statistically significant increase in the mutant frequencies after treatment with NEDA, PPD, and MG, suggesting the chemicals to be non-genotoxic in humans. The observations were consistent with that of the follow-up in vivo studies. In contrast, the mutant frequency was markedly increased in XRCC1 -/-/XPA -/- cells after treatment with auramine. The results suggest that auramine is the genotoxic reagent that preferentially induces DNA damages resolved by BER and/or NER in mammals. Taken together, BER/NER deficient cell-based genotoxicity testing will contribute to elucidate the mechanism of genotoxicity and therefore play a pivotal role in the accurate safety assessment of chemical substances.
Collapse
Affiliation(s)
- Akira Sassa
- Department of Biology, Graduate School of Science, Chiba University, Chiba, Japan
| | - Takayuki Fukuda
- Tokyo Laboratory, BoZo Research Center Inc., Hanegi, Setagaya-ku, Tokyo, Japan
| | - Akiko Ukai
- Division of Genetics and Mutagenesis, National Institute of Health Sciences, Tonomachi, Kawasaki-ku, Kawasaki Japan
| | - Maki Nakamura
- Tokyo Laboratory, BoZo Research Center Inc., Hanegi, Setagaya-ku, Tokyo, Japan
| | - Ryosuke Sato
- Tokyo Laboratory, BoZo Research Center Inc., Hanegi, Setagaya-ku, Tokyo, Japan
| | - Sho Fujiwara
- Tokyo Laboratory, BoZo Research Center Inc., Hanegi, Setagaya-ku, Tokyo, Japan
| | - Kouji Hirota
- Department of Chemistry, Graduate School of Science, Tokyo Metropolitan University, Hachioji-shi, Tokyo, Japan
| | - Shunichi Takeda
- Department of Radiation Genetics, Graduate School of Medicine, Kyoto University, Yoshida Konoe, Sakyo-ku, Kyoto, Japan
| | - Kei-Ichi Sugiyama
- Division of Genetics and Mutagenesis, National Institute of Health Sciences, Tonomachi, Kawasaki-ku, Kawasaki Japan
| | - Masamitsu Honma
- Division of Genetics and Mutagenesis, National Institute of Health Sciences, Tonomachi, Kawasaki-ku, Kawasaki Japan
| | - Manabu Yasui
- Division of Genetics and Mutagenesis, National Institute of Health Sciences, Tonomachi, Kawasaki-ku, Kawasaki Japan
| |
Collapse
|
4
|
Tokumaru Y, Katsuta E, Oshi M, Sporn JC, Yan L, Le L, Matsuhashi N, Futamura M, Akao Y, Yoshida K, Takabe K. High Expression of miR-34a Associated with Less Aggressive Cancer Biology but Not with Survival in Breast Cancer. Int J Mol Sci 2020; 21:ijms21093045. [PMID: 32357442 PMCID: PMC7246662 DOI: 10.3390/ijms21093045] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 01/06/2023] Open
Abstract
Most breast cancer (BC) patients succumb to metastatic disease. MiR-34a is a well-known tumor suppressive microRNA which exerts its anti-cancer functions by playing a role in p53, apoptosis induction, and epithelial-mesenchymal transition (EMT) suppression. Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) and The Cancer Genome Atlas (TCGA) cohorts were used to test our hypothesis that miR-34a high BCs translate to less aggressive cancer biology and better survival in large cohorts. There was no association between miR-34a expression levels and clinicopathological features of BC patients except for HER2 positivity. MiR-34a high expressing tumors were associated with lower Nottingham pathological grades and lower MKI67 expression. In agreement, high miR-34a tumors demonstrated lower GSVA scores of cell cycle and cell proliferation-related gene sets. High miR-34a tumors enriched the p53 pathway and apoptosis gene sets. Unexpectedly, high miR-34a tumors also associated with elevated EMT pathway score and ZEB1 and two expressions. MiR-34a expression did not associate with any distant metastasis. Further, high miR-34a tumors did not associate with better survival compared with miR-34a low tumors. In conclusion, the clinical relevance of miR-34a high expressing tumors was associated with suppressed cell proliferation, enhanced p53 pathway and apoptosis, but enhanced EMT and these findings did not reflect better survival outcomes in large BC patient cohorts.
Collapse
Affiliation(s)
- Yoshihisa Tokumaru
- Breast Surgery, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (Y.T.); (E.K.); (M.O.); (J.C.S.); (L.L.)
- Department of Surgical Oncology, Graduate School of Medicine, Gifu University, Gifu 501-1194, Japan; (N.M.); (M.F.); (K.Y.)
| | - Eriko Katsuta
- Breast Surgery, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (Y.T.); (E.K.); (M.O.); (J.C.S.); (L.L.)
| | - Masanori Oshi
- Breast Surgery, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (Y.T.); (E.K.); (M.O.); (J.C.S.); (L.L.)
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama 236-004, Japan
| | - Judith C. Sporn
- Breast Surgery, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (Y.T.); (E.K.); (M.O.); (J.C.S.); (L.L.)
| | - Li Yan
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
| | - Lan Le
- Breast Surgery, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (Y.T.); (E.K.); (M.O.); (J.C.S.); (L.L.)
- Department of Surgery, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, The State University of New York, Buffalo, NY 14203, USA
| | - Nobuhisa Matsuhashi
- Department of Surgical Oncology, Graduate School of Medicine, Gifu University, Gifu 501-1194, Japan; (N.M.); (M.F.); (K.Y.)
| | - Manabu Futamura
- Department of Surgical Oncology, Graduate School of Medicine, Gifu University, Gifu 501-1194, Japan; (N.M.); (M.F.); (K.Y.)
| | - Yukihiro Akao
- United Graduate School of Drug and Medical Information Sciences, Gifu University, Gifu 501-1194, Japan;
| | - Kazuhiro Yoshida
- Department of Surgical Oncology, Graduate School of Medicine, Gifu University, Gifu 501-1194, Japan; (N.M.); (M.F.); (K.Y.)
| | - Kazuaki Takabe
- Breast Surgery, Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; (Y.T.); (E.K.); (M.O.); (J.C.S.); (L.L.)
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama 236-004, Japan
- Department of Surgery, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, The State University of New York, Buffalo, NY 14203, USA
- Department of Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo 160-8402, Japan
- Department of Breast Surgery, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
- Correspondence:
| |
Collapse
|
5
|
Geraniol Inhibits Endometrial Carcinoma via Downregulating Oncogenes and Upregulating Tumour Suppressor Genes. Indian J Clin Biochem 2016; 32:214-219. [PMID: 28428697 DOI: 10.1007/s12291-016-0601-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 07/29/2016] [Indexed: 12/15/2022]
Abstract
Endometrial carcinoma is the fourth most abundant cancer worldwide in women. Female Wistar rats were segregated into five groups: group I-control, group II-MNNG (N-methyl-N'-nitro-N-nitrosoguanidine-150 mg/kg) administered through intravaginal detention of cotton absorbent, group III-geraniol (GOH) only, group IV-GOH-pretreated (7 days before the start of MNNG administration); and group V-Co-administration of geraniol with MNNG. In this study, reverse transcriptase- PCR of K-ras, MAPK, PI3K, Wnt/β-catenin genes, TGF-β and expressions of PCNA, PTEN, progesterone receptor and E-cadherin by Western blotting were performed from endometrial cancer tissue and control tissues. The mRNA expressions of K-ras, MAPK, PI3K, Wnt/β-catenin and TGF-β were amplified in MNNG induced group. Oral administration of GOH (both pre and co-administration) reversed the mRNA expression towards normal. The reversibility is more predominant in pretreatment groups (p < 0.05). The expression of PCNA was upregulated and downregulation of PTEN, progesterone receptor and E-cadherin was noticed in MNNG induced rats. Pre and co-administration of GOH significantly reversed the expression pattern of proteins. GOH treatment is more effective in pretreatment groups (p < 0.05). These results provide powerful evidences that GOH could influence modulation of MAPK pathways and Wnt signalling pathways in the prevention of endometrial carcinoma in rats.
Collapse
|
6
|
Ah-Koon L, Lesage D, Lemadre E, Souissi I, Fagard R, Varin-Blank N, Fabre EE, Schischmanoff O. Cellular response to alkylating agent MNNG is impaired in STAT1-deficients cells. J Cell Mol Med 2016; 20:1956-65. [PMID: 27464833 PMCID: PMC5020624 DOI: 10.1111/jcmm.12887] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 04/27/2016] [Indexed: 11/30/2022] Open
Abstract
The SN 1 alkylating agents activate the mismatch repair system leading to delayed G2 /M cell cycle arrest and DNA repair with subsequent survival or cell death. STAT1, an anti-proliferative and pro-apoptotic transcription factor is known to potentiate p53 and to affect DNA-damage cellular response. We studied whether STAT1 may modulate cell fate following activation of the mismatch repair system upon exposure to the alkylating agent N-methyl-N'-nitro-N-nitrosoguanidine (MNNG). Using STAT1-proficient or -deficient cell lines, we found that STAT1 is required for: (i) reduction in the extent of DNA lesions, (ii) rapid phosphorylation of T68-CHK2 and of S15-p53, (iii) progression through the G2 /M checkpoint and (iv) long-term survival following treatment with MNNG. Presence of STAT1 is critical for the formation of a p53-DNA complex comprising: STAT1, c-Abl and MLH1 following exposure to MNNG. Importantly, presence of STAT1 allows recruitment of c-Abl to p53-DNA complex and links c-Abl tyrosine kinase activity to MNNG-toxicity. Thus, our data highlight the important modulatory role of STAT1 in the signalling pathway activated by the mismatch repair system. This ability of STAT1 to favour resistance to MNNG indicates the targeting of STAT1 pathway as a therapeutic option for enhancing the efficacy of SN1 alkylating agent-based chemotherapy.
Collapse
Affiliation(s)
- Laurent Ah-Koon
- INSERM, U978, Bobigny, France.,Université Paris 13, UFR SMBH, Sorbonne Paris Cité, Laboratoire d'excellence INFLAMEX, Bobigny, France
| | - Denis Lesage
- INSERM, U978, Bobigny, France.,Université Paris 13, UFR SMBH, Sorbonne Paris Cité, Laboratoire d'excellence INFLAMEX, Bobigny, France
| | - Elodie Lemadre
- INSERM, U978, Bobigny, France.,Université Paris 13, UFR SMBH, Sorbonne Paris Cité, Laboratoire d'excellence INFLAMEX, Bobigny, France
| | - Inès Souissi
- INSERM, U978, Bobigny, France.,Université Paris 13, UFR SMBH, Sorbonne Paris Cité, Laboratoire d'excellence INFLAMEX, Bobigny, France
| | - Remi Fagard
- INSERM, U978, Bobigny, France.,Université Paris 13, UFR SMBH, Sorbonne Paris Cité, Laboratoire d'excellence INFLAMEX, Bobigny, France.,AP-HP, GHU-PSSD, Hôpital Avicenne, Service de Biochimie, Bobigny, France
| | - Nadine Varin-Blank
- INSERM, U978, Bobigny, France. .,Université Paris 13, UFR SMBH, Sorbonne Paris Cité, Laboratoire d'excellence INFLAMEX, Bobigny, France.
| | - Emmanuelle E Fabre
- INSERM, U978, Bobigny, France.,Université Paris 13, UFR SMBH, Sorbonne Paris Cité, Laboratoire d'excellence INFLAMEX, Bobigny, France.,AP-HP, GHU-PSSD, Hôpital Avicenne, Service de Biochimie, Bobigny, France
| | - Olivier Schischmanoff
- INSERM, U978, Bobigny, France. .,Université Paris 13, UFR SMBH, Sorbonne Paris Cité, Laboratoire d'excellence INFLAMEX, Bobigny, France. .,AP-HP, GHU-PSSD, Hôpital Avicenne, Service de Biochimie, Bobigny, France.
| |
Collapse
|
7
|
Fatokun AA, Dawson VL, Dawson TM. Parthanatos: mitochondrial-linked mechanisms and therapeutic opportunities. Br J Pharmacol 2014; 171:2000-16. [PMID: 24684389 PMCID: PMC3976618 DOI: 10.1111/bph.12416] [Citation(s) in RCA: 414] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Revised: 08/27/2013] [Accepted: 09/02/2013] [Indexed: 12/12/2022] Open
Abstract
Cells die by a variety of mechanisms. Terminally differentiated cells such as neurones die in a variety of disorders, in part, via parthanatos, a process dependent on the activity of poly (ADP-ribose)-polymerase (PARP). Parthanatos does not require the mediation of caspases for its execution, but is clearly mechanistically dependent on the nuclear translocation of the mitochondrial-associated apoptosis-inducing factor (AIF). The nuclear translocation of this otherwise beneficial mitochondrial protein, occasioned by poly (ADP-ribose) (PAR) produced through PARP overactivation, causes large-scale DNA fragmentation and chromatin condensation, leading to cell death. This review describes the multistep course of parthanatos and its dependence on PAR signalling and nuclear AIF translocation. The review also discusses potential targets in the parthanatos cascade as promising avenues for the development of novel, disease-modifying, therapeutic agents.
Collapse
Affiliation(s)
- Amos A Fatokun
- Institute of Cell Signalling, School of Biomedical Sciences, University of NottinghamNottingham, UK
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of MedicineBaltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of MedicineBaltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimore, MD, USA
- Department of Physiology, Johns Hopkins University School of MedicineBaltimore, MD, USA
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of MedicineBaltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of MedicineBaltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimore, MD, USA
| |
Collapse
|
8
|
da Silva CG, Minussi DC, Ferran C, Bredel M. A20 expressing tumors and anticancer drug resistance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 809:65-81. [PMID: 25302366 DOI: 10.1007/978-1-4939-0398-6_5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Resistance to anticancer drugs is a major impediment to treating patients with cancer. The molecular mechanisms deciding whether a tumor cell commits to cell death or survives under chemotherapy are complex. Mounting evidence indicates a critical role of cell death and survival pathways in determining the response of human cancers to chemotherapy. Nuclear factor-kappaB (NF-kappaB) is a eukaryotic transcription factor on the crossroad of a cell's decision to live or die. Under physiological conditions, NF-kappaB is regulated by a complex network of endogenous pathway modulators. Tumor necrosis factor alpha induced protein 3 (tnfaip3), a gene encoding the A20 protein, is one of the cell's own inhibitory molecule, which regulates canonical NF-kappaB activation by interacting with upstream signaling pathway components. Interestingly, A20 is also itself a NF-kappaB dependent gene, that has been shown to also exert cell-type specific anti- or pro-apoptotic functions. Recent reports suggest that A20 expression is increased in a number of solid human tumors. This likely contributes to both carcinogenesis and response to chemotherapy. These data uncover the complexities of the mechanisms involved in A20's impact on tumor development and response to treatment, highlighting tumor and drug-type specific outcomes. While A20-targeted therapies may certainly add to the chemotherapeutic armamentarium, better understanding of A20 regulation, molecular targets and function(s) in every single tumor and in response to any given drug is required prior to any clinical implementation. Current renewed appreciation of the unique molecular signature of each tumor holds promise for personalized chemotherapeutic regimen hopefully comprising specific A20-targeting agents i.e., both inhibitors and enhancers.
Collapse
|
9
|
Suppression of alkylating agent induced cell transformation and gastric ulceration by low-dose alkylating agent pretreatment. Biochem Biophys Res Commun 2013; 435:714-9. [PMID: 23702486 DOI: 10.1016/j.bbrc.2013.05.049] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Accepted: 05/11/2013] [Indexed: 11/22/2022]
Abstract
Exposure to mild stress by chemicals and radiation causes DNA damage and leads to acquired stress resistance. Although the linear no-threshold (LNT) model of safety assessment assumes risk from any dose, evidence from radiological research demonstrates a conflicting hormetic phenomenon known as the hormesis effect. However, the mechanisms underlying radiation hormesis have not yet been clarified, and little is known about the effects of low doses of chemical carcinogens. We analyzed the efficacy of pretreatment with low doses of the alkylating agent N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) on the subsequent induction of cell transformation and gastric ulceration by high-dose MNNG. We used an in vitro Balb/3T3 A31-1-1 cell transformation test and monitored the formation of gastric ulcers in 5-week-old male ICR mice that were administered MNNG in drinking water. The treatment concentrations of MNNG were determined by the cell survival rate and past reports. For low-dose in vitro and in vivo experiments, MNNG was used at 0.028 μM, and 2.8 μg/mL, respectively. The frequency of cell transformation induced by 10 μm MNNG was decreased by low-dose MNNG pretreatment to levels similar to that of spontaneous transformation. In addition, reactive oxygen species (ROS) and mutation frequencies induced by 10 μm MNNG were decreased by low-dose MNNG pretreatment. Importantly, low-dose MNNG pretreatment had no effect on cell proliferation. In vivo studies showed that the number of gastric ulcers induced by 1 mg/mL MNNG decreased after low-dose MNNG pretreatment. These data indicate that low-dose pretreatment with carcinogens may play a beneficial role in the prevention of chemical toxicity under specified conditions.
Collapse
|
10
|
Topham CH, Billinton N, Walmsley RM. Nongenotoxic Apoptosis Inducers Do Not Produce Misleading Positive Results in the TK6 Cell-Based GADD45a-GFP Genotoxicity Assay. Toxicol Sci 2012; 128:79-91. [DOI: 10.1093/toxsci/kfs132] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
11
|
Chen YC, Lu PH, Hsu JL, Yu CC, Guh JH. ICAM-1 and AMPK regulate cell detachment and apoptosis by N-methyl-N'-nitro-N-nitrosoguanidine, a widely spread environmental chemical, in human hormone-refractory prostate cancers. Toxicol Appl Pharmacol 2011; 257:412-9. [PMID: 22001597 DOI: 10.1016/j.taap.2011.09.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Revised: 09/12/2011] [Accepted: 09/29/2011] [Indexed: 01/29/2023]
Abstract
Poly(ADP-ribose) polymerase-1 (PARP-1), a sensor of DNA damage, plays a crucial role in the regulation of DNA repair. PARP-1 hyperactivation causes DNA damage and cell death. The underlying mechanism is complicated and is through diverse pathways. The understanding of responsible signaling pathways may offer implications for effective therapies. After concentration-response determination of N-Methyl-N'-Nitro-N-Nitrosoguanidine (MNNG, a PARP-1 activating agent and an environmental mutagen) in human hormone-refractory prostate cancers, the data showed that concentrations below 5μM did not change cell survival but cause a time-dependent up-regulation of intracellular adhesion molecule-1 (ICAM-1) in mRNA, total protein and cell surface levels. Detection of phosphorylation and degradation of IκB-α and nuclear translocation of NF-κB showed that MNNG induced the activation of NF-κB that was responsible for the ICAM-1 up-regulation since PDTC (a NF-κB inhibitor) significantly abolished this effect. However, higher concentrations (e.g., 10μM) of MNNG induced a 61% detachment of the cells which were apoptosis associated with the activation of AMP-activated protein kinase (AMPK), c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK). Further identification showed that both AMPK and JNK other than p38 MAPK functionally contributed to cell death. The remaining 39% attached cells were survival associated with high ICAM-1 expression. In conclusion, the data suggest that NF-κB-dependent up-regulation of ICAM-1 plays a key role on cell attachment and survival; whereas, activation of AMPK and JNK participates in cytotoxic signaling pathways in detached cells caused by PARP-1 activation.
Collapse
Affiliation(s)
- Yi-Cheng Chen
- School of Pharmacy, College of Medicine, National Taiwan University, No. 1, Sect. 1, Jen-Ai Rd, Taipei 100, Taiwan
| | | | | | | | | |
Collapse
|
12
|
Miri H, Bathaie SZ, Mohagheghi MA, Mokhtari-Dizaji M, Shahbazfar AA. A noninvasive method for early detection of MNNG-induced gastric cancer of male Wistar rat: ultrasonic study. ULTRASOUND IN MEDICINE & BIOLOGY 2011; 37:780-787. [PMID: 21458142 DOI: 10.1016/j.ultrasmedbio.2010.11.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2010] [Revised: 11/06/2010] [Accepted: 11/23/2010] [Indexed: 05/30/2023]
Abstract
Gastric cancer is often diagnosed at advanced stages and there is no accurate method for its screening and diagnosis, especially in small animals. Here, we explain the application of B-mode ultrasound imaging (BMUI) for screening of gastric changes in the rat. Thus, male Albino Wistar rats, weighing 100-120 grams were randomly divided into two groups. The control group rats (n=10) were given water as routine; the remaining (n=90), were given N-methyl-N-nitro-N-nitrosoguanidine (MNNG, 100 μg/mL) in drinking water ad libitum for 40 weeks. Fifteen rats were killed at different time intervals and the others were sacrificed after 55 weeks. The BMUI of the stomach of animals after MNNG administration show some changes compared with the normal groups. Pathologic investigations of the stomach indicate cancer induction at different levels. The sensitivity and specificity of BMUI is 96.6% and 78.78%, respectively. Thus, it is a useful method of diagnosis of gastric cancer in rats.
Collapse
Affiliation(s)
- Hamidreza Miri
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | | | | | | |
Collapse
|
13
|
Zaïr ZM, Jenkins GJ, Doak SH, Singh R, Brown K, Johnson GE. N-Methylpurine DNA Glycosylase Plays a Pivotal Role in the Threshold Response of Ethyl Methanesulfonate–Induced Chromosome Damage. Toxicol Sci 2010; 119:346-58. [DOI: 10.1093/toxsci/kfq341] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
14
|
Sun G, Jin S, Baskaran R. MMR/c-Abl-dependent activation of ING2/p73alpha signaling regulates the cell death response to N-methyl-N'-nitro-N-nitrosoguanidine. Exp Cell Res 2009; 315:3163-75. [PMID: 19766113 DOI: 10.1016/j.yexcr.2009.09.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2009] [Revised: 09/10/2009] [Accepted: 09/10/2009] [Indexed: 01/09/2023]
Abstract
Agents inducing O(6)-methylguanine (O(6)MeG) in DNA such as N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) are cytotoxic and a deficiency in mismatch repair (MMR) results in lack of sensitivity to this genotoxin (termed alkylation tolerance). Here, we show that ING2, a member of the inhibitor of growth family, is required for cell death induced by MNNG. We further observe that MNNG treatment increases cellular protein levels of ING2 that is dependent on intact MMR function and that MNNG-induced ING2 localizes and associates with p73alpha in the nucleus. Suppression of ING2 by short hairpin RNA (shRNA) in MMR-proficient colorectal cancer cells decreased its sensitivity to MNNG and, in addition, abrogated MNNG-induced stabilization and acetylation of p73alpha. Interestingly, suppression of p73alpha had a greater impact on MNNG-induced cell death than ING2 leading us to conclude that ING2 regulates the cell death response, in part, through p73alpha. Inhibition of c-Abl by STI571 or suppression of c-Abl expression by shRNA blocked ING2 induction and p73alpha acetylation induced by this alkylator. Similarly, suppression of MMR (MLH1) by shRNA abrogated ING2 induction/p73alpha acetylation. Taken together, these results demonstrate that MLH1/c-Abl-dependent activation of ING2>p73alpha signaling regulates cell death triggered by MNNG and further suggests that dysregulation of this event may, in part, be responsible for alkylation tolerance observed in MMR compromised cells.
Collapse
Affiliation(s)
- Guoming Sun
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, E1205 Biomedical Science Tower, Pittsburgh, PA 15261, USA
| | | | | |
Collapse
|
15
|
Li LS, Morales JC, Hwang A, Wagner MW, Boothman DA. DNA mismatch repair-dependent activation of c-Abl/p73alpha/GADD45alpha-mediated apoptosis. J Biol Chem 2008; 283:21394-403. [PMID: 18480060 DOI: 10.1074/jbc.m709954200] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Cells with functional DNA mismatch repair (MMR) stimulate G(2) cell cycle checkpoint arrest and apoptosis in response to N-methyl-N'-nitro-N-nitrosoguanidine (MNNG). MMR-deficient cells fail to detect MNNG-induced DNA damage, resulting in the survival of "mutator" cells. The retrograde (nucleus-to-cytoplasm) signaling that initiates MMR-dependent G(2) arrest and cell death remains undefined. Since MMR-dependent phosphorylation and stabilization of p53 were noted, we investigated its role(s) in G(2) arrest and apoptosis. Loss of p53 function by E6 expression, dominant-negative p53, or stable p53 knockdown failed to prevent MMR-dependent G(2) arrest, apoptosis, or lethality. MMR-dependent c-Abl-mediated p73alpha and GADD45alpha protein up-regulation after MNNG exposure prompted us to examine c-Abl/p73alpha/GADD45alpha signaling in cell death responses. STI571 (Gleevec, a c-Abl tyrosine kinase inhibitor) and stable c-Abl, p73alpha, and GADD45alpha knockdown prevented MMR-dependent apoptosis. Interestingly, stable p73alpha knockdown blocked MMR-dependent apoptosis, but not G(2) arrest, thereby uncoupling G(2) arrest from lethality. Thus, MMR-dependent intrinsic apoptosis is p53-independent, but stimulated by hMLH1/c-Abl/p73alpha/GADD45alpha retrograde signaling.
Collapse
Affiliation(s)
- Long Shan Li
- Laboratory of Molecular Stress Responses, Department of Oncology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center at Dallas, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | | | | | | | | |
Collapse
|
16
|
Ai L, Kim WJ, Demircan B, Dyer LM, Bray KJ, Skehan RR, Massoll NA, Brown KD. The transglutaminase 2 gene (TGM2), a potential molecular marker for chemotherapeutic drug sensitivity, is epigenetically silenced in breast cancer. Carcinogenesis 2008; 29:510-8. [PMID: 18174247 DOI: 10.1093/carcin/bgm280] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Tissue transglutaminase (TG2) is a ubiquitously expressed enzyme capable of catalyzing protein cross-links. TG2-dependent cross-links are important in extracellular matrix integrity and it has been proposed that this TG2 activity establishes a barrier to tumor spread. Furthermore, TG2 controls sensitivity to the chemotherapeutic drug doxorubicin. Both doxorubicin sensitivity and TG2 expression are highly variable in cultured human breast cancer cell lines and inspection of the human gene (termed TGM2) determined that a canonical CpG island exists within its 5' flank. These features, when combined with its potential tumor suppressor activity, make TG2 an attractive candidate for epigenetic silencing. Consistent with this, we observed that culturing breast tumor cells with the DNA demethylating agent 5-aza-2'-deoxycytidine (5-azadC) resulted in a robust increase in TG2 expression. Analysis of DNA harvested from cultured lines and primary breast tumor samples indicated that TGM2 often displays aberrant hypermethylation and that there is a statistically significant correlation between gene methylation and reduced expression. Finally, we observed that doxorubicin-resistant MCF-7/ADR cells do not show TGM2 silencing but that doxorubicin-sensitive MCF-7 cells do and that culturing MCF-7 cells on 5-azadC and subsequently restoring TG2 expression reduced sensitivity to doxorubicin. This work indicates that the TGM2 gene is a target for epigenetic silencing in breast cancer and suggests that this aberrant molecular event is a potential marker for chemotherapeutic drug sensitivity.
Collapse
Affiliation(s)
- Lingbao Ai
- Department of Biochemistry and Molecular Biology and University of Florida Shands Cancer Center Program in Cancer Genetics, Epigenetics and Tumor Virology, University of Florida College of Medicine, Box 100245, Gainesville, FL 32610, USA
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Lee MW, Kim WJ, Beardsley DI, Brown KD. N-methyl-N'-nitro-N-nitrosoguanidine activates multiple cell death mechanisms in human fibroblasts. DNA Cell Biol 2007; 26:683-94. [PMID: 17678437 DOI: 10.1089/dna.2007.0594] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Response to genotoxic stress may trigger the activation of distinct mechanisms that serve to promote cell death, including apoptosis and necrosis. In this study we examined the response of human fibroblasts, either proficient or deficient for the damage-activated protein kinase ataxia telangiectasia-mutated (ATM), to the alkylating agent N-methyl-N'-nitro-N-nitrosoguanidine (MNNG). Analysis of both long- and short-term viability shows that both ATM-proficient YZ-5 and ATM-deficient EBS-7 fibroblasts display a cytotoxic response to MNNG. Consistent with activation of apoptosis in response to MNNG, we observed increased caspase-3 cleavage and activity, appearance of fragmented nuclei, and increased staining with annexin V in both ATM-proficient and -deficient fibroblasts. Flow cytometry demonstrated that these cell lines also display a nonapoptotic cell death in response to MNNG. This form of cell death is associated with activation of poly-ADP ribose polymerase (PARP), and analysis of PARP activity indicated increased protein poly(ADP-ribosylation) in YZ-5 when compared to EBS-7. This PARP activity was accompanied by apoptosis-inducing factor release and translocation from the mitochondria to the nucleus. Finally, the PARP inhibitor 3,4-dihydro-5-[4-(1-piperidinyl)butoxy]-1(2H)-isoquinolinone (DPQ) or the caspase-3 inhibitor benzyloxycarbonyl-VAD-fluoromethyl ketone dramatically diminished the cytotoxic response to MNNG, reinforcing the roles for apoptotic and nonapoptotic cell death in human fibroblasts treated with MNNG. From these findings, we conclude that MNNG induces a heterogeneous death response in human fibroblasts.
Collapse
Affiliation(s)
- Michael W Lee
- Department of Biochemistry and Molecular Biology and the UF-Shands Cancer Center Program in Cancer Genetics, Epigenetics and Tumor Virology, University of Florida College of Medicine, Gainesville, Florida 32610, USA
| | | | | | | |
Collapse
|
18
|
Kim WJ, Rajasekaran B, Brown KD. MLH1- and ATM-dependent MAPK signaling is activated through c-Abl in response to the alkylator N-methyl-N'-nitro-N'-nitrosoguanidine. J Biol Chem 2007; 282:32021-31. [PMID: 17804421 DOI: 10.1074/jbc.m701451200] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
N-Methyl-N'-nitro-N'-nitrosoguanidine (MNNG) is a DNA-methylating agent, and deficiency in mismatch repair (MMR) results in lack of sensitivity to this genotoxin (termed alkylation tolerance). A number of DNA damage response pathways are activated in a MMR-dependent manner following MNNG, and several also require ATM kinase activity. Here we show that activation of the transcription factor c-Jun is dependent upon both the MMR component MLH1 and ATM, but not ATR, in response to MNNG. In addition to c-Jun, the upstream MAPKs JNK and MKK4 are also activated in a MLH1- and ATM-dependent manner. We document that c-Jun activation is dependent on the MAPK kinase kinase MEKK1. Additionally, the tyrosine kinase c-Abl is required to activate this signaling cascade and forms a complex with MEKK1 and MLH1. This study indicates that an arm of DNA damage-activated MAPK signaling is activated in an MLH1- and ATM-dependent manner in response to MNNG and perhaps suggests that dysregulation of this signaling is responsible, in part, for alkylation tolerance.
Collapse
Affiliation(s)
- Wan-Ju Kim
- Department of Biochemistry and Molecular Biology and the University of Florida Shands Cancer Center, University of Florida College of Medicine, Gainesville, Florida 32610, USA
| | | | | |
Collapse
|
19
|
Skourou V, Keramitsoglou T, Koussoulakou D, Mitashov V, Koussoulakos S. Immunostimulation exacerbates the biological effects of chemical carcinogens. BIOL BULL+ 2007. [DOI: 10.1134/s1062359007040048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
20
|
Raver-Shapira N, Marciano E, Meiri E, Spector Y, Rosenfeld N, Moskovits N, Bentwich Z, Oren M. Transcriptional activation of miR-34a contributes to p53-mediated apoptosis. Mol Cell 2007; 26:731-43. [PMID: 17540598 DOI: 10.1016/j.molcel.2007.05.017] [Citation(s) in RCA: 1019] [Impact Index Per Article: 56.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2007] [Revised: 05/05/2007] [Accepted: 05/17/2007] [Indexed: 02/07/2023]
Abstract
p53 is a potent tumor suppressor, whose biological effects are largely due to its function as a transcriptional regulator. Here we report that, in addition to regulating the expression of hundreds of protein-coding genes, p53 also modulates the levels of microRNAs (miRNAs). Specifically, p53 can induce expression of microRNA-34a (miR-34a) in cultured cells as well as in irradiated mice, by binding to a perfect p53 binding site located within the gene that gives rise to miR-34a. Processing of the primary transcript into mature miR-34a involves the excision of a 30 kb intron. Notably, inactivation of miR-34a strongly attenuates p53-mediated apoptosis in cells exposed to genotoxic stress, whereas overexpression of miR-34a mildly increases apoptosis. Hence, miR-34a is a direct proapoptotic transcriptional target of p53 that can mediate some of p53's biological effects. Perturbation of miR-34a expression, as occurs in some human cancers, may thus contribute to tumorigenesis by attenuating p53-dependent apoptosis.
Collapse
Affiliation(s)
- Nina Raver-Shapira
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot 76100, Israel
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Moubarak RS, Yuste VJ, Artus C, Bouharrour A, Greer PA, Menissier-de Murcia J, Susin SA. Sequential activation of poly(ADP-ribose) polymerase 1, calpains, and Bax is essential in apoptosis-inducing factor-mediated programmed necrosis. Mol Cell Biol 2007; 27:4844-62. [PMID: 17470554 PMCID: PMC1951482 DOI: 10.1128/mcb.02141-06] [Citation(s) in RCA: 238] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Alkylating DNA damage induces a necrotic type of programmed cell death through the poly(ADP-ribose) polymerases (PARP) and apoptosis-inducing factor (AIF). Following PARP activation, AIF is released from mitochondria and translocates to the nucleus, where it causes chromatin condensation and DNA fragmentation. By employing a large panel of gene knockout cells, we identified and describe here two essential molecular links between PARP and AIF: calpains and Bax. Alkylating DNA damage initiated a p53-independent form of death involving PARP-1 but not PARP-2. Once activated, PARP-1 mediated mitochondrial AIF release and necrosis through a mechanism requiring calpains but not cathepsins or caspases. Importantly, single ablation of the proapoptotic Bcl-2 family member Bax, but not Bak, prevented both AIF release and alkylating DNA damage-induced death. Thus, Bax is indispensable for this type of necrosis. Our data also revealed that Bcl-2 regulates N-methyl-N'-nitro-N'-nitrosoguanidine-induced necrosis. Finally, we established the molecular ordering of PARP-1, calpains, Bax, and AIF activation, and we showed that AIF downregulation confers resistance to alkylating DNA damage-induced necrosis. Our data shed new light on the mechanisms regulating AIF-dependent necrosis and support the notion that, like apoptosis, necrosis could be a highly regulated cell death program.
Collapse
Affiliation(s)
- Rana S Moubarak
- Apoptose et Système Immunitaire, CNRS-URA 1961, Institut Pasteur, 25 Rue du Dr. Roux, 75015 Paris, France
| | | | | | | | | | | | | |
Collapse
|
22
|
Yang L, Li D, Chen J, Yang J, Xue L, Hu S, Wu K. Microarray expression analysis of the early N-methy-N-nitrosourea-induced retinal degeneration in rat. Neurosci Lett 2007; 418:38-43. [PMID: 17412507 DOI: 10.1016/j.neulet.2007.02.084] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2006] [Revised: 01/23/2007] [Accepted: 02/25/2007] [Indexed: 11/24/2022]
Abstract
The study was undertaken to investigate the gene expressions in N-methy-N-nitrosourea (MNU)-induced rat retinal degeneration (RD) by performing microarray analysis of retinal RNA at 12h. All rats were randomly divided into a normal group, a 12h model group and a 24h model group. Rats in the two model groups received a single intraperitoneal injection of 40 mg/kg body weight of MNU, while those in the normal group were injected with equivalent volume of physiological saline. After 12h and 24h of the injection, rats in each respective group were sacrificed, respectively. One eye of each animal was used for hematoxylin and erosin (H&E) staining, and fresh retinas of the other eye of each animal in the both normal group and 12h model group were used to extract total RNA, which was analyzed by microarray and real time RT-PCR. Retinal histological alteration was found in the 24h model group. There were 75 genes differently expressed (ratio > or =2.0), including 64 genes up-regulated and 11 genes down-regulated. Seven genes were assayed by real time RT-PCR and demonstrated the same alteration tendency as in microarray analysis. These genes that expressed differently mainly involved signal transduction, development, immune and defense, and apoptosis, etc. The major pathways were MAP-kinase signaling pathways, Toll-like receptor signaling pathway and apoptosis pathway involved. The results suggest that there are significant changes of gene expression in the early stage of MNU-induced RD. These microarray results provide clues to understand the molecular pathways underlying photoreceptor degeneration and indicate directions for future studies.
Collapse
Affiliation(s)
- Liu Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, PR China
| | | | | | | | | | | | | |
Collapse
|
23
|
Amaravadi RK, Yu D, Lum JJ, Bui T, Christophorou MA, Evan GI, Thomas-Tikhonenko A, Thompson CB. Autophagy inhibition enhances therapy-induced apoptosis in a Myc-induced model of lymphoma. J Clin Invest 2007; 117:326-36. [PMID: 17235397 PMCID: PMC1765515 DOI: 10.1172/jci28833] [Citation(s) in RCA: 912] [Impact Index Per Article: 50.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2006] [Accepted: 11/28/2006] [Indexed: 12/13/2022] Open
Abstract
Autophagy is a lysosome-dependent degradative pathway frequently activated in tumor cells treated with chemotherapy or radiation. Whether autophagy observed in treated cancer cells represents a mechanism that allows tumor cells to survive therapy or a mechanism for initiating a nonapoptotic form of programmed cell death remains controversial. To address this issue, the role of autophagy in a Myc-induced model of lymphoma generated from cells derived from p53ER(TAM)/p53ER(TAM) mice (with ER denoting estrogen receptor) was examined. Such tumors are resistant to apoptosis due to a lack of nuclear p53. Systemic administration of tamoxifen led to p53 activation and tumor regression followed by tumor recurrence. Activation of p53 was associated with the rapid appearance of apoptotic cells and the induction of autophagy in surviving cells. Inhibition of autophagy with either chloroquine or ATG5 short hairpin RNA (shRNA) enhanced the ability of either p53 activation or alkylating drug therapy to induce tumor cell death. These studies provide evidence that autophagy serves as a survival pathway in tumor cells treated with apoptosis activators and a rationale for the use of autophagy inhibitors such as chloroquine in combination with therapies designed to induce apoptosis in human cancers.
Collapse
Affiliation(s)
- Ravi K. Amaravadi
- Division of Hematology/Oncology, Department of Medicine,
Department of Cancer Biology, Abramson Family Cancer Research Institute, and
Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Cancer Research Institute, University of California San Francisco Comprehensive Cancer Center, San Francisco, California, USA
| | - Duonan Yu
- Division of Hematology/Oncology, Department of Medicine,
Department of Cancer Biology, Abramson Family Cancer Research Institute, and
Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Cancer Research Institute, University of California San Francisco Comprehensive Cancer Center, San Francisco, California, USA
| | - Julian J. Lum
- Division of Hematology/Oncology, Department of Medicine,
Department of Cancer Biology, Abramson Family Cancer Research Institute, and
Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Cancer Research Institute, University of California San Francisco Comprehensive Cancer Center, San Francisco, California, USA
| | - Thi Bui
- Division of Hematology/Oncology, Department of Medicine,
Department of Cancer Biology, Abramson Family Cancer Research Institute, and
Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Cancer Research Institute, University of California San Francisco Comprehensive Cancer Center, San Francisco, California, USA
| | - Maria A. Christophorou
- Division of Hematology/Oncology, Department of Medicine,
Department of Cancer Biology, Abramson Family Cancer Research Institute, and
Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Cancer Research Institute, University of California San Francisco Comprehensive Cancer Center, San Francisco, California, USA
| | - Gerard I. Evan
- Division of Hematology/Oncology, Department of Medicine,
Department of Cancer Biology, Abramson Family Cancer Research Institute, and
Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Cancer Research Institute, University of California San Francisco Comprehensive Cancer Center, San Francisco, California, USA
| | - Andrei Thomas-Tikhonenko
- Division of Hematology/Oncology, Department of Medicine,
Department of Cancer Biology, Abramson Family Cancer Research Institute, and
Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Cancer Research Institute, University of California San Francisco Comprehensive Cancer Center, San Francisco, California, USA
| | - Craig B. Thompson
- Division of Hematology/Oncology, Department of Medicine,
Department of Cancer Biology, Abramson Family Cancer Research Institute, and
Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
Cancer Research Institute, University of California San Francisco Comprehensive Cancer Center, San Francisco, California, USA
| |
Collapse
|