1
|
Mehta H, Ambele MA, Mokgautsi N, Moela P. Probing the Effects of Retinoblastoma Binding Protein 6 (RBBP6) Knockdown on the Sensitivity of Cisplatin in Cervical Cancer Cells. Cells 2024; 13:700. [PMID: 38667315 PMCID: PMC11049397 DOI: 10.3390/cells13080700] [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: 01/03/2024] [Revised: 04/05/2024] [Accepted: 04/13/2024] [Indexed: 04/28/2024] Open
Abstract
Cervical cancer is a major cause of death in women despite the advancement of current treatment modalities. The conventional therapeutic agent, cisplatin (CCDP), is the standard treatment for CC; however, resistance often develops due to the cancer's heterogeneity. Therefore, a detailed elucidation of the specific molecular mechanisms driving CC is crucial for the development of targeted therapeutic strategies. Retinoblastoma binding protein 6 (RBBP6) is a potential biomarker associated with cell proliferation and is upregulated in cervical cancer sites, exhibiting apoptosis and dysregulated p53 expression. Furthermore, RBBP6 has been demonstrated to sensitize cancer cells to radiation and certain chemotherapeutic agents by regulating the Bcl-2 gene, thus suggesting a crosstalk among RBBP6/p53/BCL-2 oncogenic signatures. The present study, therefore, investigated the relationship between cisplatin and RBBP6 expression in CC cells. Herein, we first explored bioinformatics simulations and identified that the RBBP6/p53/BCL-2 signaling pathway is overexpressed and correlated with CC. For further analysis, we explored the Genomics of Drug Sensitivity in Cancer (GDSC) and found that most of the CC cell lines are sensitive to CCDP. To validate these findings, RBBP6 was silenced in HeLa and Vero cells using RNAi technology, followed by measurement of wild-type p53 and Bcl-2 at the mRNA level using qPCR. Cells co-treated with cisplatin and siRBBP6 were subsequently analyzed for apoptosis induction and real-time growth monitoring using flow cytometry and the xCELLigence system, respectively. Cancer cells in the co-treatment group showed a reduction in apoptosis compared to the cisplatin-treated group. Moreover, the real-time growth monitoring revealed a reduced growth rate in RBBP6 knockdown cells treated with cisplatin. Although wild-type p53 remained unchanged in the co-treatment group of cancer cells, Bcl-2 was completely repressed, suggesting that RBBP6 is necessary for sensitizing cervical cancer cells to cisplatin treatment by downregulating Bcl-2. The Vero cell population, which served as a non-cancerous control cell line in this study, remained viable following treatment with both siRBBP6 and cisplatin. Findings from this study suggest that RBBP6 expression promotes cisplatin sensitivity in HeLa cells through Bcl-2 downregulation. Knockdown of RBBP6 limits apoptosis induction and delays cell growth inhibition in response to cisplatin. The knowledge obtained here has the potential to help improve cisplatin efficacy through personalized administration based on the expression profile of RBBP6 among individual patients.
Collapse
Affiliation(s)
- Harshini Mehta
- Division of Genetics, Department of Biochemistry, Genetics and Microbiology, Faculty of Natural and Agricultural Sciences, University of Pretoria, Pretoria 0002, South Africa; (H.M.); (N.M.)
| | - Melvin Anyasi Ambele
- Institute for Cellular and Molecular Medicine, Department of Immunology and SAMRC Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa;
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa
| | - Ntlotlang Mokgautsi
- Division of Genetics, Department of Biochemistry, Genetics and Microbiology, Faculty of Natural and Agricultural Sciences, University of Pretoria, Pretoria 0002, South Africa; (H.M.); (N.M.)
| | - Pontsho Moela
- Division of Genetics, Department of Biochemistry, Genetics and Microbiology, Faculty of Natural and Agricultural Sciences, University of Pretoria, Pretoria 0002, South Africa; (H.M.); (N.M.)
| |
Collapse
|
2
|
Naqash AR, Floudas CS, Aber E, Maoz A, Nassar AH, Adib E, Choucair K, Xiu J, Baca Y, Ricciuti B, Alessi JV, Awad MM, Kim C, Judd J, Raez LE, Lopes G, Nieva JJ, Borghaei H, Takebe N, Ma PC, Halmos B, Kwiatkowski DJ, Liu SV, Mamdani H. Influence of TP53 Comutation on the Tumor Immune Microenvironment and Clinical Outcomes With Immune Checkpoint Inhibitors in STK11-Mutant Non-Small-Cell Lung Cancer. JCO Precis Oncol 2024; 8:e2300371. [PMID: 38330261 PMCID: PMC10860998 DOI: 10.1200/po.23.00371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 11/05/2023] [Accepted: 01/02/2024] [Indexed: 02/10/2024] Open
Abstract
PURPOSE Non-small-cell lung cancer (NSCLC) with STK11mut has inferior outcomes to immune checkpoint inhibitors (ICIs). Using multiomics, we evaluated whether a subtype of STK11mut NSCLC with a uniquely inflamed tumor immune microenvironment (TIME) harboring TP53 comutations could have favorable outcomes to ICIs. PATIENTS AND METHODS NSCLC tumors (N = 16,896) were analyzed by next-generation sequencing (DNA-Seq/592 genes). A subset (n = 5,034) underwent gene expression profiling (RNA-Seq/whole transcriptome). Exome-level neoantigen load for STK11mut NSCLC was obtained from published pan-immune analysis. Tumor immune cell content was obtained from transcriptome profiles using the microenvironment cell population (MCP) counter. ICI data from POPLAR/OAK (n = 34) and the study by Rizvi et al (n = 49) were used to model progression-free survival (PFS), and a separate ICI-treated cohort (n = 53) from Dana-Farber Cancer Institute (DFCI) was used to assess time to treatment failure (TTF) and tumor RECIST response for STK11mutTP53mut versus STK11mutTP53wt NSCLC. RESULTS Overall, 12.6% of NSCLC tumors had a STK11mut with the proportions of tumor mutational burden (TMB)-high (≥10 mut/Mb), PD-L1 ≥50%, and microsatellite instability-high being 38.3%, 11.8%, and 0.72%, respectively. Unsupervised hierarchical clustering of STK11mut (n = 463) for stimulator of interferon-gamma (STING) pathway genes identified a STING-high cluster, which was significantly enriched in TP53mut NSCLC (P < .01). Compared with STK11mutTP53wt, tumors with STK11mutTP53mut had higher CD8+T cells and natural killer cells (P < .01), higher TMB (P < .001) and neoantigen load (P < .001), and increased expression of MYC and HIF-1A (P < .01), along with higher expression (P < .01) of glycolysis/glutamine metabolism genes. Meta-analysis of data from OAK/POPLAR and the study by Rizvi et al showed a trend toward improved PFS in patients with STK11mutTP53mut. In the DFCI cohort, compared with the STK11mut TP53wt cohort, the STK11mutTP53mut tumors had higher objective response rates (42.9% v 16.7%; P = .04) and also had longer TTF (14.5 v 4.5 months, P adj = .054) with ICI. CONCLUSION STK11mut NSCLC with TP53 comutation is a distinct subgroup with an immunologically active TIME and metabolic reprogramming. These properties should be exploited to guide patient selection for novel ICI-based combination approaches.
Collapse
Affiliation(s)
- Abdul Rafeh Naqash
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | | | - Etan Aber
- Center for Immuno-Oncology, National Cancer Institute, NIH, Bethesda, MD
| | - Asaf Maoz
- Dana Farber Cancer Institute, Boston, MA
| | - Amin H. Nassar
- Department of Hematology/Oncology, Yale New Haven Hospital, New Haven, CT
| | - Elio Adib
- Dana Farber Cancer Institute, Boston, MA
| | - Khalil Choucair
- Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI
| | | | | | | | | | | | - Chul Kim
- Department of Hematology and Oncology, Georgetown University, Washington, DC
| | - Julia Judd
- Fox Chase Cancer Center, Philadelphia, PA
| | - Luis E. Raez
- Memorial Cancer Institute//Florida Atlantic University (FAU), Miami, FL
| | - Gilberto Lopes
- University of Miami Miller School of Medicine, Miami, FL
| | | | | | - Naoko Takebe
- Developmental Therapeutics Clinic, National Cancer Institute, Bethesda, MD
| | - Patrick C. Ma
- Department of Hematology/ Oncology, Penn State Cancer Institute, Hershey, PA
| | - Balazs Halmos
- Medical Oncology, Albert Einstein College of Medicine, NY
| | | | - Stephen V. Liu
- Department of Hematology and Oncology, Georgetown University, Washington, DC
| | - Hirva Mamdani
- Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI
| |
Collapse
|
3
|
Low temperature plasma suppresses proliferation, invasion, migration and survival of SK-BR-3 breast cancer cells. Mol Biol Rep 2023; 50:2025-2031. [PMID: 36538172 DOI: 10.1007/s11033-022-08026-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 10/10/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Low temperature plasma (LTP) is a developing field in recent years to play important roles of sterilization, material modification and wound healing. Breast cancer is a common gynecological malignant tumor. Recent studies have shown that LTP is a promising selective anti-cancer treatment. The effect of LTP on breast cancer is still unclear. In this study, We treated breast cancer cell lines with low temperature plasma for different periods of time and analyzed the relevant differences. METHODS AND RESULTS SK-BR-3 cell nutrient solution was firstly treated by ACP for 0, 10, 20, 40, 80 and 120 s, which was next used to cultivateSK-BR-3cells for overnight.we found that LTP was able to suppress cell vitality, proliferation, invasion and migration of SK-BR-3 cells. Also, SK-BR-3 apoptosis was induced by LTP in a time-dependent manner. CONCLUSION These evidences suggest the negative effect of LTP on malignant development of SK-BR-3 cells, and LTP has the potential clinical application for breast cancer treatment.
Collapse
|
4
|
Baumgartner C, Yadav AK, Chefetz I. AMPK-like proteins and their function in female reproduction and gynecologic cancer. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 134:245-270. [PMID: 36858738 DOI: 10.1016/bs.apcsb.2022.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Serine-threonine kinase (STK11), also known as liver kinase B1 (LKB1), is a regulator of cellular homeostasis through regulating the cellular ATP-to-ADP ratio. LKB1 is classified as a tumor suppressor and functions as the key activator of AMP-activated protein kinase (AMPK) and a family of serine-threonine kinases called AMPK-like proteins. These proteins include novel (nua) kinase family 1 (NUAK1 and 2), salt inducible kinase (SIK1), QIK (known as SIK2), QSK (known as SIK3 kinase), and maternal embryonic leuzine zipper kinase (MELK) on tightly controlled and specific residual sites. LKB1 also regulates brain selective kinases 1 and 2 (BRSK1 and 2), additional members of AMPK-like protein family, which functions are probably less studied. AMPK-like proteins play a role in variety of reproductive physiology functions such as follicular maturation, menopause, embryogenesis, oocyte maturation, and preimplantation development. In addition, dysfunctional activity of AMPK-like proteins contributes to apoptosis blockade in cancer cells and induction of the epithelial-mesenchymal transition required for metastasis. Dysregulation of these proteins occurs in ovarian, endometrial, and cervical cancers. AMPK-like proteins are still undergoing further classification and may represent novel targets for targeted gynecologic cancer therapies. In this chapter, we describe the AMPK-like family of proteins and their roles in reproductive physiology and gynecologic cancers.
Collapse
Affiliation(s)
| | - Anil Kumar Yadav
- The Hormel Institute, University of Minnesota, Austin, MN, United States
| | - Ilana Chefetz
- The Hormel Institute, University of Minnesota, Austin, MN, United States; Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States; Stem Cell Institute, University of Minnesota, Minneapolis, MN, United States; Department of Obstetrics, Gynecology and Women's Health, University of Minnesota, Minneapolis, MN, United States.
| |
Collapse
|
5
|
Liu Y, Gu W. The complexity of p53-mediated metabolic regulation in tumor suppression. Semin Cancer Biol 2022; 85:4-32. [PMID: 33785447 PMCID: PMC8473587 DOI: 10.1016/j.semcancer.2021.03.010] [Citation(s) in RCA: 99] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/12/2021] [Accepted: 03/15/2021] [Indexed: 02/07/2023]
Abstract
Although the classic activities of p53 including induction of cell-cycle arrest, senescence, and apoptosis are well accepted as critical barriers to cancer development, accumulating evidence suggests that loss of these classic activities is not sufficient to abrogate the tumor suppression activity of p53. Numerous studies suggest that metabolic regulation contributes to tumor suppression, but the mechanisms by which it does so are not completely understood. Cancer cells rewire cellular metabolism to meet the energetic and substrate demands of tumor development. It is well established that p53 suppresses glycolysis and promotes mitochondrial oxidative phosphorylation through a number of downstream targets against the Warburg effect. The role of p53-mediated metabolic regulation in tumor suppression is complexed by its function to promote both cell survival and cell death under different physiological settings. Indeed, p53 can regulate both pro-oxidant and antioxidant target genes for complete opposite effects. In this review, we will summarize the roles of p53 in the regulation of glucose, lipid, amino acid, nucleotide, iron metabolism, and ROS production. We will highlight the mechanisms underlying p53-mediated ferroptosis, AKT/mTOR signaling as well as autophagy and discuss the complexity of p53-metabolic regulation in tumor development.
Collapse
Affiliation(s)
- Yanqing Liu
- Institute for Cancer Genetics, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY, 10032, USA
| | - Wei Gu
- Institute for Cancer Genetics, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY, 10032, USA; Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, 1130 Nicholas Ave, New York, NY, 10032, USA.
| |
Collapse
|
6
|
Melnik BC, Schmitz G. Milk Exosomal microRNAs: Postnatal Promoters of β Cell Proliferation but Potential Inducers of β Cell De-Differentiation in Adult Life. Int J Mol Sci 2022; 23:ijms231911503. [PMID: 36232796 PMCID: PMC9569743 DOI: 10.3390/ijms231911503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Pancreatic β cell expansion and functional maturation during the birth-to-weaning period is driven by epigenetic programs primarily triggered by growth factors, hormones, and nutrients provided by human milk. As shown recently, exosomes derived from various origins interact with β cells. This review elucidates the potential role of milk-derived exosomes (MEX) and their microRNAs (miRs) on pancreatic β cell programming during the postnatal period of lactation as well as during continuous cow milk exposure of adult humans to bovine MEX. Mechanistic evidence suggests that MEX miRs stimulate mTORC1/c-MYC-dependent postnatal β cell proliferation and glycolysis, but attenuate β cell differentiation, mitochondrial function, and insulin synthesis and secretion. MEX miR content is negatively affected by maternal obesity, gestational diabetes, psychological stress, caesarean delivery, and is completely absent in infant formula. Weaning-related disappearance of MEX miRs may be the critical event switching β cells from proliferation to TGF-β/AMPK-mediated cell differentiation, whereas continued exposure of adult humans to bovine MEX miRs via intake of pasteurized cow milk may reverse β cell differentiation, promoting β cell de-differentiation. Whereas MEX miR signaling supports postnatal β cell proliferation (diabetes prevention), persistent bovine MEX exposure after the lactation period may de-differentiate β cells back to the postnatal phenotype (diabetes induction).
Collapse
Affiliation(s)
- Bodo C. Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, D-49076 Osnabrück, Germany
- Correspondence: ; Tel.: +49-52-4198-8060
| | - Gerd Schmitz
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital of Regensburg, University of Regensburg, D-93053 Regensburg, Germany
| |
Collapse
|
7
|
Kulinczak M, Sromek M, Panek G, Zakrzewska K, Lotocka R, Szafron LM, Chechlinska M, Siwicki JK. Endometrial Cancer-Adjacent Tissues Express Higher Levels of Cancer-Promoting Genes than the Matched Tumors. Genes (Basel) 2022; 13:genes13091611. [PMID: 36140779 PMCID: PMC9527013 DOI: 10.3390/genes13091611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/02/2022] [Accepted: 09/06/2022] [Indexed: 11/16/2022] Open
Abstract
Molecular alterations in tumor-adjacent tissues have recently been recognized in some types of cancer. This phenomenon has not been studied in endometrial cancer. We aimed to analyze the expression of genes associated with cancer progression and metabolism in primary endometrial cancer samples and the matched tumor-adjacent tissues and in the samples of endometria from cancer-free patients with uterine leiomyomas. Paired samples of tumor-adjacent tissues and primary tumors from 49 patients with endometrial cancer (EC), samples of endometrium from 25 patients with leiomyomas of the uterus, and 4 endometrial cancer cell lines were examined by the RT-qPCR, for MYC, NR5A2, CXCR2, HMGA2, LIN28A, OCT4A, OCT4B, OCT4B1, TWIST1, STK11, SNAI1, and miR-205-5p expression. The expression levels of MYC, NR5A2, SNAI1, TWIST1, and STK11 were significantly higher in tumor-adjacent tissues than in the matched EC samples, and this difference was not influenced by the content of cancer cells in cancer-adjacent tissues. The expression of MYC, NR5A2, and SNAI1 was also higher in EC-adjacent tissues than in samples from cancer-free patients. In addition, the expression of MYC and CXCR2 in the tumor related to non-endometrioid adenocarcinoma and reduced the risk of recurrence, respectively, and higher NR5A2 expression in tumor-adjacent tissue increased the risk of death. In conclusion, tissues proximal to EC present higher levels of some cancer-promoting genes than the matched tumors. Malignant tumor-adjacent tissues carry a diagnostic potential and emerge as new promising target of anticancer therapy.
Collapse
Affiliation(s)
- Mariusz Kulinczak
- Department of Cancer Biology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Maria Sromek
- Department of Cancer Biology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Grzegorz Panek
- Department of Gynecologic Oncology and Obstetrics, Centre of Postgraduate Medical Education, 00-416 Warsaw, Poland
| | - Klara Zakrzewska
- Department of Pathology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Renata Lotocka
- Cancer Molecular and Genetic Diagnostics Laboratory, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Lukasz Michal Szafron
- Department of Cancer Biology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Magdalena Chechlinska
- Department of Cancer Biology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Jan Konrad Siwicki
- Department of Cancer Biology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
- Correspondence: ; Tel.: +48-22-546-2787
| |
Collapse
|
8
|
Cui D, Qu R, Liu D, Xiong X, Liang T, Zhao Y. The Cross Talk Between p53 and mTOR Pathways in Response to Physiological and Genotoxic Stresses. Front Cell Dev Biol 2021; 9:775507. [PMID: 34869377 PMCID: PMC8638743 DOI: 10.3389/fcell.2021.775507] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 11/03/2021] [Indexed: 12/25/2022] Open
Abstract
The tumor suppressor p53 is activated upon multiple cellular stresses, including DNA damage, oncogene activation, ribosomal stress, and hypoxia, to induce cell cycle arrest, apoptosis, and senescence. Mammalian target of rapamycin (mTOR), an evolutionarily conserved serine/threonine protein kinase, serves as a central regulator of cell growth, proliferation, and survival by coordinating nutrients, energy, growth factors, and oxygen levels. p53 dysfunction and mTOR pathway hyperactivation are hallmarks of human cancer. The balance between response to stresses or commitment to cell proliferation and survival is governed by various regulatory loops between the p53 and mTOR pathways. In this review, we first briefly introduce the tumor suppressor p53 and then describe the upstream regulators and downstream effectors of the mTOR pathway. Next, we discuss the role of p53 in regulating the mTOR pathway through its transcriptional and non-transcriptional effects. We further describe the complicated role of the mTOR pathway in modulating p53 activity. Finally, we discuss the current knowledge and future perspectives on the coordinated regulation of the p53 and mTOR pathways.
Collapse
Affiliation(s)
- Danrui Cui
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Cancer Center, Zhejiang University, Hangzhou, China
| | - Ruirui Qu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Dian Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiufang Xiong
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.,Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Cancer Center, Zhejiang University, Hangzhou, China
| | - Yongchao Zhao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Cancer Center, Zhejiang University, Hangzhou, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
9
|
Cobb LP, Siamakpour-Reihani S, Zhang D, Qin X, Owzar K, Zhou C, Conrads TP, Maxwell GL, Darcy KM, Bateman NW, Litzi T, Bae-Jump V, Secord AA. Obesity and altered angiogenic-related gene expression in endometrial cancer. Gynecol Oncol 2021; 163:320-326. [PMID: 34538531 PMCID: PMC11018267 DOI: 10.1016/j.ygyno.2021.08.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 08/12/2021] [Accepted: 08/14/2021] [Indexed: 11/22/2022]
Abstract
OBJECTIVES Evaluate association between obesity and angiogenic-related gene expression in endometrial cancer (EC). Evaluate interaction between diet and metformin on angiogenic-related gene expression. METHODS We evaluated the association between 168 human angiogenic-related genes and body mass index (BMI) in the TCGA Uterine Corpus Endometrial Carcinoma cohort (endometrioid endometrial cancer (EEC) cohort n = 290, and copy number high cohort n = 55), an independent validation cohort from Gynecologic Cancer Center of Excellence (GYN-COE) (n = 62) and corresponding 185 homologous mouse genes in an LKB1fl/flp53fl/fl mouse model of EC (n = 20). Mice received 60% of calories from fat in a high-fat diet (HFD), mimicking diet-induced obesity, versus 10% of calories from fat in a low-fat diet (LFD). After tumor growth, HFD (n = 5) and LFD (n = 5) mice were treated with metformin (200 mg/kg/day) or control. Whole transcriptome analysis of mouse tumors was performed using RNA-Seq. RESULTS At a false-discovery rate of 10%, twenty-one angiogenic-related genes were differentially expressed with respect to BMI when adjusting for grade in the TCGA EEC cohort. Evaluation of these genes in the mouse model control group revealed association between increased Edil3 expression in HFD versus LFD mice (2.5-fold change (FC); unadjusted p = 0.03). An interaction was observed for expression of Edil3 between diet and metformin treatment (unadjusted p = 0.009). Association between BMI and increased expression of EDIL3 was validated in one of four EDIL3 probesets in the GYN-COE cohort (p = 0.0011, adjusted p = 0.0342). CONCLUSIONS Obesity may promote tumor progression via differential modulation of angiogenic pathways in EEC. Our exploratory findings demonstrated that EDIL3 may be a candidate gene of interest.
Collapse
Affiliation(s)
- Lauren Patterson Cobb
- Department of Gynecologic Oncology and Reproductive Medicine, MD Anderson Cancer Center, Houston, TX, USA; Division of Gynecology Oncology, Department of Obstetrics and Gynecology, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA.
| | - Sharareh Siamakpour-Reihani
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University Medical Center, USA
| | - Dadong Zhang
- Bioinformatics Shared Resource, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Xiaodi Qin
- Bioinformatics Shared Resource, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Kouros Owzar
- Bioinformatics Shared Resource, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA; Duke Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC, USA
| | - Chunxiao Zhou
- Division of Gynecologic Oncology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Thomas P Conrads
- Gynecologic Cancer Center of Excellence, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, Bethesda, MD, USA; Women's Health Integrated Research Center, Women's Service Line, Inova Health System, Falls Church, VA, USA
| | - G Larry Maxwell
- Gynecologic Cancer Center of Excellence, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, Bethesda, MD, USA; Women's Health Integrated Research Center, Women's Service Line, Inova Health System, Falls Church, VA, USA; Inova Schar Cancer Institute, Inova Center for Personalized Health, Falls Church, VA, USA
| | - Kathleen M Darcy
- Gynecologic Cancer Center of Excellence, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, Bethesda, MD, USA; The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD, USA
| | - Nicholas W Bateman
- Gynecologic Cancer Center of Excellence, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, Bethesda, MD, USA; The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD, USA
| | - Tracy Litzi
- Gynecologic Cancer Center of Excellence, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, Bethesda, MD, USA; The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD, USA
| | - Victoria Bae-Jump
- Division of Gynecologic Oncology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Angeles Alvarez Secord
- Division of Gynecology Oncology, Department of Obstetrics and Gynecology, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
10
|
Jin LY, Zhao K, Xu LJ, Zhao RX, Werle KD, Wang Y, Liu XL, Chen Q, Wu ZJ, Zhang K, Zhao Y, Jiang GQ, Cui FM, Xu ZX. LKB1 inactivation leads to centromere defects and genome instability via p53-dependent upregulation of survivin. Aging (Albany NY) 2020; 12:14341-14354. [PMID: 32668413 PMCID: PMC7425461 DOI: 10.18632/aging.103473] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 04/17/2020] [Indexed: 01/25/2023]
Abstract
Inactivating mutations in the liver kinase B1 (LKB1) tumor suppressor gene underlie Peutz-Jeghers syndrome (PJS) and occur frequently in various human cancers. We previously showed that LKB1 regulates centrosome duplication via PLK1. Here, we report that LKB1 further helps to maintain genomic stability through negative regulation of survivin, a member of the chromosomal passenger complex (CPC) that mediates CPC targeting to the centromere. We found that loss of LKB1 led to accumulation of misaligned and lagging chromosomes at metaphase and anaphase and increased the appearance of multi- and micro-nucleated cells. Ectopic LKB1 expression reduced these features and improved mitotic fidelity in LKB1-deficient cells. Through pharmacological and genetic manipulations, we showed that LKB1-mediated repression of survivin is independent of AMPK, but requires p53. Consistent with the key influence of LKB1 on survivin expression, immunohistochemical analysis indicated that survivin is highly expressed in intestinal polyps from a PJS patient. Lastly, we reaffirm a potential therapeutic avenue to treat LKB1-mutated tumors by demonstrating the increased sensitivity to survivin inhibitors of LKB1-deficient cells.
Collapse
Affiliation(s)
- Li-Yan Jin
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China.,Department of General Surgery, The Second Affiliated Hospital, Soochow University, Suzhou 215004, China
| | - Kui Zhao
- Department of General Surgery, The Second Affiliated Hospital, Soochow University, Suzhou 215004, China
| | - Long-Jiang Xu
- Department of Pathology, The Second Affiliated Hospital, Soochow University, Suzhou 215004, China
| | - Rui-Xun Zhao
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Kaitlin D Werle
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Yong Wang
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Xiao-Long Liu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China.,Department of Urology, The Second Affiliated Hospital, Soochow University, Suzhou 215004, China
| | - Qiu Chen
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
| | - Zhuo-Jun Wu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
| | - Ke Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
| | - Ying Zhao
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
| | - Guo-Qin Jiang
- Department of General Surgery, The Second Affiliated Hospital, Soochow University, Suzhou 215004, China
| | - Feng-Mei Cui
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China.,Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Zhi-Xiang Xu
- School of Life Sciences, Henan University, Kaifeng, Henan Province 475004, China.,Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
11
|
Lacroix M, Riscal R, Arena G, Linares LK, Le Cam L. Metabolic functions of the tumor suppressor p53: Implications in normal physiology, metabolic disorders, and cancer. Mol Metab 2020; 33:2-22. [PMID: 31685430 PMCID: PMC7056927 DOI: 10.1016/j.molmet.2019.10.002] [Citation(s) in RCA: 197] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 09/24/2019] [Accepted: 10/05/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The TP53 gene is one of the most commonly inactivated tumor suppressors in human cancers. p53 functions during cancer progression have been linked to a variety of transcriptional and non-transcriptional activities that lead to the tight control of cell proliferation, senescence, DNA repair, and cell death. However, converging evidence indicates that p53 also plays a major role in metabolism in both normal and cancer cells. SCOPE OF REVIEW We provide an overview of the current knowledge on the metabolic activities of wild type (WT) p53 and highlight some of the mechanisms by which p53 contributes to whole body energy homeostasis. We will also pinpoint some evidences suggesting that deregulation of p53-associated metabolic activities leads to human pathologies beyond cancer, including obesity, diabetes, liver, and cardiovascular diseases. MAJOR CONCLUSIONS p53 is activated when cells are metabolically challenged but the origin, duration, and intensity of these stresses will dictate the outcome of the p53 response. p53 plays pivotal roles both upstream and downstream of several key metabolic regulators and is involved in multiple feedback-loops that ensure proper cellular homeostasis. The physiological roles of p53 in metabolism involve complex mechanisms of regulation implicating both cell autonomous effects as well as autocrine loops. However, the mechanisms by which p53 coordinates metabolism at the organismal level remain poorly understood. Perturbations of p53-regulated metabolic activities contribute to various metabolic disorders and are pivotal during cancer progression.
Collapse
Affiliation(s)
- Matthieu Lacroix
- Institut de Recherche en Cancérologie de Montpellier, INSERM, Université de Montpellier, Institut Régional du Cancer de Montpellier, Montpellier, France; Equipe labélisée Ligue Contre le Cancer, France
| | - Romain Riscal
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Giuseppe Arena
- Gustave Roussy Cancer Campus, INSERM U1030, Villejuif, France
| | - Laetitia Karine Linares
- Institut de Recherche en Cancérologie de Montpellier, INSERM, Université de Montpellier, Institut Régional du Cancer de Montpellier, Montpellier, France; Equipe labélisée Ligue Contre le Cancer, France
| | - Laurent Le Cam
- Institut de Recherche en Cancérologie de Montpellier, INSERM, Université de Montpellier, Institut Régional du Cancer de Montpellier, Montpellier, France; Equipe labélisée Ligue Contre le Cancer, France.
| |
Collapse
|
12
|
Guo H, Kong W, Zhang L, Han J, Clark LH, Yin Y, Fang Z, Sun W, Wang J, Gilliam TP, Lee D, Makowski L, Zhou C, Bae-Jump VL. Reversal of obesity-driven aggressiveness of endometrial cancer by metformin. Am J Cancer Res 2019; 9:2170-2193. [PMID: 31720081 PMCID: PMC6834476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 09/20/2019] [Indexed: 06/10/2023] Open
Abstract
BACKGROUND Obesity and diabetes are associated with increased risk and worse outcomes for endometrial cancer. Metformin is a widely prescribed generic drug for the treatment of type II diabetes and metabolic syndrome and may also have anti-tumorigenic effects. Thus, we assessed the metabolic anti-tumorigenic effects of metformin in (1) human endometrial cancer cell lines under varying glucose concentrations, and (2) a novel genetically engineered mouse model of endometrioid endometrial cancer under obese and lean conditions. METHODS The effects of metformin on cytotoxicity, apoptosis, cell cycle progression, and the AMPK/mTOR/S6 and MAPK pathways were assessed in ECC-1 and Ishikawa cells under low, normal and high glucose conditions. The impact of metformin treatment on tumor growth under obese and lean conditions was evaluated using a novel LKB1fl/fl p53fl/fl mouse model of endometrial cancer. Global, untargeted metabolomics was used to identify (1) obesity-associated differences between endometrial tumors and (2) the obesity-dependent effects of metformin in the endometrial tumors. RESULTS Hypoglycemic conditions significantly enhanced the sensitivity of the cells to metformin in regards to its anti-proliferative and apoptotic effects, as compared to hyperglycemic and normal glucose conditions. Metformin inhibited tumor growth in both the obese and lean mice, which metformin-induced inhibition of tumor progression in obese mice was significantly greater than in lean mice. Metabolomic profiling in endometrial cancer tissues revealed significant differences between obese- and lean-mice. Enhanced energy metabolism was seen in obese- versus lean-mice as evidenced by increases in glycolytic and oxidative phosphorylation intermediates. In addition, dramatic increases in lipid biosynthesis and lipid peroxidation were found in the obese- versus lean-mice, whereas metformin obviously reversed the obesity-driven upregulation of lipid and protein biosynthesis in the obese mice. CONCLUSIONS The obese state promoted tumor aggressiveness in the LKB1fl/fl p53fl/fl mouse model, accompanied by increases in energy metabolism, lipid biosynthesis, and markers of lipid peroxidation. Metformin had increased efficacy against endometrial cancer in obese versus lean mice and reversed the detrimental metabolic effects of obesity in the endometrial tumors. Taken together, it is likely that the unique metabolic milieu underlies metformin's improved efficacy in treating endometrial cancer which develop in an obese host environment.
Collapse
Affiliation(s)
- Hui Guo
- Department of Gynecologic Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinan, Shandong, China
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC, USA
- School of Medicine and Life Sciences, University of Jinan, Shandong Academy of Medical SciencesJinan, Shandong, China
| | - Weimin Kong
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical UniversityBeijing, China
| | - Lu Zhang
- Department of Gynecologic Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinan, Shandong, China
| | - Jianjun Han
- Department of Surgical Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinan, Shandong, China
| | - Leslie H Clark
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC, USA
| | - Yajie Yin
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC, USA
| | - Ziwei Fang
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical UniversityBeijing, China
| | - Wenchuan Sun
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC, USA
| | - Jiandong Wang
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical UniversityBeijing, China
| | - Timothy P Gilliam
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC, USA
| | | | - Liza Makowski
- Division of Hematology and Oncology, Department of Medicine, University of Tennessee Health Science CenterMemphis, TN, USA
| | - Chunxiao Zhou
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel HillChapel Hill, NC, USA
| | - Victoria L Bae-Jump
- Division of Gynecologic Oncology, University of North Carolina at Chapel HillChapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel HillChapel Hill, NC, USA
| |
Collapse
|
13
|
Bange E, Marmarelis ME, Hwang WT, Yang YX, Thompson JC, Rosenbaum J, Bauml JM, Ciunci C, Alley EW, Cohen RB, Langer CJ, Carpenter E, Aggarwal C. Impact of KRAS and TP53 Co-Mutations on Outcomes After First-Line Systemic Therapy Among Patients With STK11-Mutated Advanced Non-Small-Cell Lung Cancer. JCO Precis Oncol 2019; 3. [PMID: 31428721 DOI: 10.1200/po.18.00326] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
PURPOSE The STK11 gene encodes a serine/threonine protein kinase that regulates cell polarity and functions as a tumor suppressor. Patients with non-small-cell lung cancer (NSCLC) and STK11 mutations often have other co-mutations. We evaluated the impact of KRAS and TP53 co-mutations on outcomes after first-line systemic therapy for patients with metastatic or recurrent NSCLC that harbors STK11 mutations. METHODS We conducted a retrospective review of patients with metastatic NSCLC and STK11 mutations treated at the University of Pennsylvania. STK11 mutations were identified through next-generation sequencing (NGS) in tissue or plasma. Cox proportional hazard models were used to determine the relationship between STK11 co-mutations and survival outcomes. The Kaplan-Meier method was used to estimate overall survival (OS) and progression-free survival (PFS). RESULTS From February 2013 to December 2016, samples from 1,385 patients with NSCLC were analyzed by NGS; of these, 77 patients (6%) harbored an STK11 mutation (n = 56, tissue; n = 21, plasma). Of the 62 patients included, 18 had an STK11 mutation alone, 19 had STK11/KRAS, 18 had STK11/TP53, and seven had STK11/KRAS/TP53. Patients with STK11/KRAS co-mutations had a worse median PFS (2.4 months) compared with STK11 alone (5.1 months; log-rank P = .048), STK11/TP53 (4.3 months; log-rank P = .043), and STK11/KRAS/ TP53 (13 months; log-rank P = .03). Patients with STK11/KRAS co-mutation experienced shorter median OS (7.1 months) compared with STK11 alone (16.1 months; log-rank P < .001), STK11/TP53 (28.3 months; log-rank P < .001), and STK11/KRAS/TP53 (22 months; log-rank P = .025). CONCLUSION Among patients with advanced NSCLC and STK11 mutations treated with first-line systemic therapy, co-mutation with KRAS was associated with significantly worse PFS and OS. By contrast, co-mutation of STK11 with TP53 conferred a better prognosis.
Collapse
Affiliation(s)
- Erin Bange
- University of Pennsylvania, Philadelphia, PA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Kim SH, Kim H, Lee JH, Park JW. Oxalomalate suppresses metastatic melanoma through IDH-targeted stress response to ROS. Free Radic Res 2019; 53:418-429. [DOI: 10.1080/10715762.2019.1597974] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Sung Hwan Kim
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Hyunjin Kim
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Jin Hyup Lee
- Department of Food and Biotechnology, Korea University, Sejong, Republic of Korea
| | - Jeen-Woo Park
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
15
|
Zhao P, Pang X, Jiang J, Wang L, Zhu X, Yin Y, Zhai Q, Xiang X, Feng F, Xu W. TIPE1 promotes cervical cancer progression by repression of p53 acetylation and is associated with poor cervical cancer outcome. Carcinogenesis 2018; 40:592-599. [PMID: 30445600 DOI: 10.1093/carcin/bgy163] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 10/19/2018] [Accepted: 11/13/2018] [Indexed: 01/25/2023] Open
Affiliation(s)
- Peiqing Zhao
- Department of Gynecologic Oncology, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Center of Translational Medicine, Zibo Central Hospital, Zibo, China
| | - Xiaoming Pang
- Center of Translational Medicine, Zibo Central Hospital, Zibo, China
| | - Jie Jiang
- Department of Clinical Laboratory, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Lianqing Wang
- Center of Translational Medicine, Zibo Central Hospital, Zibo, China
| | - Xiaolan Zhu
- Department of Gynecologic Oncology, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yingchun Yin
- Center of Translational Medicine, Zibo Central Hospital, Zibo, China
| | - Qiaoli Zhai
- Center of Translational Medicine, Zibo Central Hospital, Zibo, China
| | - Xinxin Xiang
- Center of Translational Medicine, Zibo Central Hospital, Zibo, China
| | - Fan Feng
- Department of Gynecologic Oncology, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Wenlin Xu
- Department of Gynecologic Oncology, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
16
|
Wang M, Fan W, Ye M, Tian C, Zhao L, Wang J, Han W, Yang W, Gu C, Li M, Zhang Z, Wang Y, Zhang H, Meng Y. Molecular profiles and tumor mutational burden analysis in Chinese patients with gynecologic cancers. Sci Rep 2018; 8:8990. [PMID: 29895933 PMCID: PMC5997642 DOI: 10.1038/s41598-018-25583-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 04/13/2018] [Indexed: 12/27/2022] Open
Abstract
The goal of this work was to investigate the tumor mutational burden (TMB) in Chinese patients with gynecologic cancer. In total, 117 patients with gynecologic cancers were included in this study. Both tumor DNA and paired blood cell genomic DNA were isolated from formalin-fixed paraffin-embedded (FFPE) specimens and blood samples, and next-generation sequencing was performed to identify somatic mutations. TP53, PTEN, ARID1A, and PIK3CA alterations were significantly different in various types of gynecologic cancers (p = 0.001, 1.15E-07, 0.004, and 0.009, respectively). The median TMB of all 117 gynecologic tumor specimens was 0.37 mutations/Mb, with a range of 0-41.45 mutations/Mb. Despite the lack of significant difference, endometrial cancer cases had a higher median TMB than cervical and ovarian cancer cases. Younger gynecologic cancer patients (age <40 years) had a significantly lower TMB than older patients (age ≥40 years) (p = 0.04). In addition, TMB was significantly increased with increasing clinical stage of disease (p = 0.001). PTEN alterations were commonly observed in patients with a moderate to high TMB (n = 8, 38.10%, p = 9.95E-04). Although limited by sample size, all of the patients with TSC2 (n = 3, p = 3.83E-11) or POLE (n = 2, p = 0.005) mutations had a moderate to high TMB. Further large-scale, prospective studies are needed to validate our findings.
Collapse
Affiliation(s)
- Min Wang
- Department of Gynecology and Obstetrics, Chinese PLA General Hospital, Beijing, P.R. China
- Department of Gynecology and Obstetrics, The 306th Hospital of PLA, Beijing, P.R. China
| | - Wensheng Fan
- Department of Gynecology and Obstetrics, Chinese PLA General Hospital, Beijing, P.R. China
| | - Mingxia Ye
- Department of Gynecology and Obstetrics, Chinese PLA General Hospital, Beijing, P.R. China
| | - Chen Tian
- Beijing Genecast Biotechnology Co., Beijing, P.R. China
| | - Lili Zhao
- Beijing Genecast Biotechnology Co., Beijing, P.R. China
| | - Jianfei Wang
- Beijing Genecast Biotechnology Co., Beijing, P.R. China
| | - Wenbo Han
- Beijing Genecast Biotechnology Co., Beijing, P.R. China
| | - Wen Yang
- Department of Gynecology and Obstetrics, Chinese PLA General Hospital, Beijing, P.R. China
| | - Chenglei Gu
- Department of Gynecology and Obstetrics, Chinese PLA General Hospital, Beijing, P.R. China
| | - Mingxia Li
- Department of Gynecology and Obstetrics, Chinese PLA General Hospital, Beijing, P.R. China
| | - Zhe Zhang
- Department of Gynecology and Obstetrics, Chinese PLA General Hospital, Beijing, P.R. China
| | - Yongjun Wang
- Department of Gynecology and Obstetrics, Peking University International Hospital, Beijing, P.R. China
- Department of Gynecology and Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, P.R. China
| | - Henghui Zhang
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, P.R. China.
- Beijing Genecast Biotechnology Co., Beijing, P.R. China.
| | - Yuanguang Meng
- Department of Gynecology and Obstetrics, Chinese PLA General Hospital, Beijing, P.R. China.
| |
Collapse
|
17
|
LKB1 as a Tumor Suppressor in Uterine Cancer: Mouse Models and Translational Studies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 943:211-241. [PMID: 27910069 DOI: 10.1007/978-3-319-43139-0_7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The LKB1 tumor suppressor was identified in 1998 as the gene mutated in the Peutz-Jeghers Syndrome (PJS), a hereditary cancer predisposition characterized by gastrointestinal polyposis and a high incidence of cancers, particularly carcinomas, at a variety of anatomic sites including the gastrointestinal tract, lung, and female reproductive tract. Women with PJS have a high incidence of carcinomas of the uterine corpus (endometrium) and cervix. The LKB1 gene is also somatically mutated in human cancers arising at these sites. Work in mouse models has highlighted the potency of LKB1 as an endometrial tumor suppressor and its distinctive roles in driving invasive and metastatic growth. These in vivo models represent tractable experimental systems for the discovery of underlying biological principles and molecular processes regulated by LKB1 in the context of tumorigenesis and also serve as useful preclinical model systems for experimental therapeutics. Here we review LKB1's known roles in mTOR signaling, metabolism, and cell polarity, with an emphasis on human pathology and mouse models relevant to uterine carcinogenesis, including cancers of the uterine corpus and cervix.
Collapse
|
18
|
Xie B, Nagalingam A, Kuppusamy P, Muniraj N, Langford P, Győrffy B, Saxena NK, Sharma D. Benzyl Isothiocyanate potentiates p53 signaling and antitumor effects against breast cancer through activation of p53-LKB1 and p73-LKB1 axes. Sci Rep 2017; 7:40070. [PMID: 28071670 PMCID: PMC5223184 DOI: 10.1038/srep40070] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 11/30/2016] [Indexed: 11/09/2022] Open
Abstract
Functional reactivation of p53 pathway, although arduous, can potentially provide a broad-based strategy for cancer therapy owing to frequent p53 inactivation in human cancer. Using a phosphoprotein-screening array, we found that Benzyl Isothiocynate, (BITC) increases p53 phosphorylation in breast cancer cells and reveal an important role of ERK and PRAS40/MDM2 in BITC-mediated p53 activation. We show that BITC rescues and activates p53-signaling network and inhibits growth of p53-mutant cells. Mechanistically, BITC induces p73 expression in p53-mutant cells, disrupts the interaction of p73 and mutant-p53, thereby releasing p73 from sequestration and allowing it to be transcriptionally active. Furthermore, BITC-induced p53 and p73 axes converge on tumor-suppressor LKB1 which is transcriptionally upregulated by p53 and p73 in p53-wild-type and p53-mutant cells respectively; and in a feed-forward mechanism, LKB1 tethers with p53 and p73 to get recruited to p53-responsive promoters. Analyses of BITC-treated xenografts using LKB1-null cells corroborate in vitro mechanistic findings and establish LKB1 as the key node whereby BITC potentiates as well as rescues p53-pathway in p53-wild-type as well as p53-mutant cells. These data provide first in vitro and in vivo evidence of the integral role of previously unrecognized crosstalk between BITC, p53/LKB1 and p73/LKB1 axes in breast tumor growth-inhibition.
Collapse
Affiliation(s)
- Bei Xie
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore MD 21231, USA
| | - Arumugam Nagalingam
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore MD 21231, USA
| | - Panjamurthy Kuppusamy
- Department of Medicine, University of Maryland School of Medicine, Baltimore MD 21201, USA
| | - Nethaji Muniraj
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore MD 21231, USA
| | - Peter Langford
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore MD 21231, USA
| | - Balázs Győrffy
- MTA TTK Momentum Cancer Biomarker Research Group, H-1117 Budapest, Semmelweis University, 2nd Dept. of Pediatrics, H-1094 Budapest, Hungary
| | - Neeraj K Saxena
- Department of Medicine, University of Maryland School of Medicine, Baltimore MD 21201, USA
| | - Dipali Sharma
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore MD 21231, USA
| |
Collapse
|
19
|
Makker A, Goel MM. Tumor progression, metastasis, and modulators of epithelial-mesenchymal transition in endometrioid endometrial carcinoma: an update. Endocr Relat Cancer 2016; 23:R85-R111. [PMID: 26538531 DOI: 10.1530/erc-15-0218] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/02/2015] [Indexed: 12/17/2022]
Abstract
Endometrioid endometrial carcinoma (EEC), also known as type 1 endometrial cancer (EC), accounts for over 70-80% of all cases that are usually associated with estrogen stimulation and often develops in a background of atypical endometrial hyperplasia. The increased incidence of EC is mainly confined to this type of cancer. Most EEC patients present at an early stage and generally have a favorable prognosis; however, up to 30% of EEC present as high risk tumors, which have invaded deep into the myometrium at diagnosis and progressively lead to local or extra pelvic metastasis. The poor survival of advanced EC is related to the lack of effective therapies, which can be attributed to poor understanding of the molecular mechanisms underlying the progression of disease toward invasion and metastasis. Multiple lines of evidence illustrate that epithelial-mesenchymal transition (EMT)-like events are central to tumor progression and malignant transformation, endowing the incipient cancer cell with invasive and metastatic properties. The aim of this review is to summarize the current knowledge on molecular events associated with EMT in progression, invasion, and metastasis of EEC. Further, the role of epigenetic modifications and microRNA regulation, tumor microenvironment, and microcystic elongated and fragmented glands like invasion pattern have been discussed. We believe this article may perhaps stimulate further research in this field that may aid in identifying high risk patients within this clinically challenging patient group and also lead to the recognition of novel targets for the prevention of metastasis - the most fatal consequence of endometrial carcinogenesis.
Collapse
Affiliation(s)
- Annu Makker
- Post Graduate Department of PathologyKing George's Medical University, Lucknow 226003, Uttar Pradesh, India
| | - Madhu Mati Goel
- Post Graduate Department of PathologyKing George's Medical University, Lucknow 226003, Uttar Pradesh, India
| |
Collapse
|
20
|
Grossi V, Lucarelli G, Forte G, Peserico A, Matrone A, Germani A, Rutigliano M, Stella A, Bagnulo R, Loconte D, Galleggiante V, Sanguedolce F, Cagiano S, Bufo P, Trabucco S, Maiorano E, Ditonno P, Battaglia M, Resta N, Simone C. Loss of STK11 expression is an early event in prostate carcinogenesis and predicts therapeutic response to targeted therapy against MAPK/p38. Autophagy 2015; 11:2102-2113. [PMID: 26391455 DOI: 10.1080/15548627.2015.1091910] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Prostate cancer (PCa) is the second leading cause of cancer-related death in men; however, the molecular mechanisms leading to its development and progression are not yet fully elucidated. Of note, it has been recently shown that conditional stk11 knockout mice develop atypical hyperplasia and prostate intraepithelial neoplasia (PIN). We recently reported an inverse correlation between the activity of the STK11/AMPK pathway and the MAPK/p38 cascade in HIF1A-dependent malignancies. Furthermore, MAPK/p38 overactivation was detected in benign prostate hyperplasia, PIN and PCa in mice and humans. Here we report that STK11 expression is significantly decreased in PCa compared to normal tissues. Moreover, STK11 protein levels decreased throughout prostate carcinogenesis. To gain insight into the role of STK11-MAPK/p38 activity balance in PCa, we treated PCa cell lines and primary biopsies with a well-established MAPK14-MAPK11 inhibitor (SB202190), which has been extensively used in vitro and in vivo. Our results indicate that inhibition of MAPK/p38 significantly affects PCa cell survival in an STK11-dependent manner. Indeed, we found that pharmacologic inactivation of MAPK/p38 does not affect viability of STK11-proficient PCa cells due to the triggering of the AMPK-dependent autophagic pathway, while it induces apoptosis in STK11-deficient cells irrespective of androgen receptor (AR) status. Of note, AMPK inactivation or autophagy inhibition in STK11-proficient cells sensitize SB202190-treated PCa cells to apoptosis. On the other end, reconstitution of functional STK11 in STK11-deficient PCa cells abrogates apoptosis. Collectively, our data show that STK11 is a key factor involved in the early phases of prostate carcinogenesis, and suggest that it might be used as a predictive marker of therapeutic response to MAPK/p38 inhibitors in PCa patients.
Collapse
Affiliation(s)
- Valentina Grossi
- a Division of Medical Genetics; Department of Biomedical Sciences and Human Oncology (DIMO) ; University of Bari 'Aldo Moro' ; Bari , Italy
| | - Giuseppe Lucarelli
- b Urology, Andrology and Kidney Transplantation Unit ; Department of Emergency and Organ Transplantation (DETO) ; University of Bari 'Aldo Moro' ; Bari , Italy
| | - Giovanna Forte
- c Cancer Genetics Laboratory; IRCCS "S. de Bellis" ; Castellana Grotte ( BA ), Italy
| | - Alessia Peserico
- a Division of Medical Genetics; Department of Biomedical Sciences and Human Oncology (DIMO) ; University of Bari 'Aldo Moro' ; Bari , Italy.,d National Cancer Institute; IRCCS Oncologico Giovanni Paolo II ; Bari , Italy
| | - Antonio Matrone
- e Developmental Biology and Cancer; UCL Institute of Child Health ; London , UK
| | - Aldo Germani
- a Division of Medical Genetics; Department of Biomedical Sciences and Human Oncology (DIMO) ; University of Bari 'Aldo Moro' ; Bari , Italy
| | - Monica Rutigliano
- b Urology, Andrology and Kidney Transplantation Unit ; Department of Emergency and Organ Transplantation (DETO) ; University of Bari 'Aldo Moro' ; Bari , Italy
| | - Alessandro Stella
- a Division of Medical Genetics; Department of Biomedical Sciences and Human Oncology (DIMO) ; University of Bari 'Aldo Moro' ; Bari , Italy
| | - Rosanna Bagnulo
- a Division of Medical Genetics; Department of Biomedical Sciences and Human Oncology (DIMO) ; University of Bari 'Aldo Moro' ; Bari , Italy
| | - Daria Loconte
- a Division of Medical Genetics; Department of Biomedical Sciences and Human Oncology (DIMO) ; University of Bari 'Aldo Moro' ; Bari , Italy
| | - Vanessa Galleggiante
- b Urology, Andrology and Kidney Transplantation Unit ; Department of Emergency and Organ Transplantation (DETO) ; University of Bari 'Aldo Moro' ; Bari , Italy
| | | | - Simona Cagiano
- f Department of Pathology ; University of Foggia ; Foggia , Italy
| | - Pantaleo Bufo
- f Department of Pathology ; University of Foggia ; Foggia , Italy
| | - Senia Trabucco
- g Department of Pathology ; University of Bari 'Aldo Moro' ; Bari , Italy
| | - Eugenio Maiorano
- g Department of Pathology ; University of Bari 'Aldo Moro' ; Bari , Italy
| | - Pasquale Ditonno
- b Urology, Andrology and Kidney Transplantation Unit ; Department of Emergency and Organ Transplantation (DETO) ; University of Bari 'Aldo Moro' ; Bari , Italy
| | - Michele Battaglia
- b Urology, Andrology and Kidney Transplantation Unit ; Department of Emergency and Organ Transplantation (DETO) ; University of Bari 'Aldo Moro' ; Bari , Italy
| | - Nicoletta Resta
- a Division of Medical Genetics; Department of Biomedical Sciences and Human Oncology (DIMO) ; University of Bari 'Aldo Moro' ; Bari , Italy
| | - Cristiano Simone
- a Division of Medical Genetics; Department of Biomedical Sciences and Human Oncology (DIMO) ; University of Bari 'Aldo Moro' ; Bari , Italy.,c Cancer Genetics Laboratory; IRCCS "S. de Bellis" ; Castellana Grotte ( BA ), Italy
| |
Collapse
|
21
|
Mao KS, Li MS, Zhou J. Update on the roles of liver kinase B1 in pancreatic cancer. Shijie Huaren Xiaohua Zazhi 2015; 23:3086-3093. [DOI: 10.11569/wcjd.v23.i19.3086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Germline mutations of the liver kinase B1 (STK11/LKB1) gene which encodes a serine/threonine kinase is responsible for Peutz-Jeghers syndrome. There are 14 AMP-activated protein kinase (AMPK)-related kinases in pathways downstream of LKB1, which are involved in many physiological and pathological processes such as regulation of energy metabolism, cell polarity and apoptosis in cells. LKB1 gene mutation has been investigated extensively in a variety of cancers, including pancreatic cancer. Pancreatic cancer is commonly recognized as a disease with extremely poor prognosis. Therefore, a full understanding of its molecular pathology is critical. This review aims to elucidate the structure, distribution, and function of LKB1, and the relationship with pancreatic cancer. In addition, we also point out that in some scenarios, LKB1 may play a role as a tumor protector.
Collapse
|