1
|
Chahat, Nainwal N, Murti Y, Yadav S, Rawat P, Dhiman S, Kumar B. Advancements in targeting tumor suppressor genes (p53 and BRCA 1/2) in breast cancer therapy. Mol Divers 2025; 29:2691-2716. [PMID: 39152355 DOI: 10.1007/s11030-024-10964-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 08/07/2024] [Indexed: 08/19/2024]
Abstract
Globally, among numerous cancer subtypes, breast cancer (BC) is one of the most prevalent forms of cancer affecting the female population. A female's family history significantly increases her risk of developing breast cancer. BC is caused by aberrant breast cells that proliferate and develop into tumors. It is estimated that 5-10% of breast carcinomas are inherited and involve genetic mutations that ensure the survival and prognosis of breast cancer cells. The most common genetic variations are responsible for hereditary breast cancer but are not limited to p53, BRCA1, and BRCA2. BRCA1 and BRCA2 are involved in genomic recombination, cell cycle monitoring, programmed cell death, and transcriptional regulation. When BRCA1 and 2 genetic variations are present in breast carcinoma, p53 irregularities become more prevalent. Both BRCA1/2 and p53 genes are involved in cell cycle monitoring. The present article discusses the current status of breast cancer research, spotlighting the tumor suppressor genes (BRCA1/2 and p53) along with structural activity relationship studies, FDA-approved drugs, and several therapy modalities for treating BC. Breast cancer drugs, accessible today in the market, have different side effects including anemia, pneumonitis, nausea, lethargy, and vomiting. Thus, the development of novel p53 and BRCA1/2 inhibitors with minimal possible side effects is crucial. We have covered compounds that have been examined subsequently (2020 onwards) in this overview which may be utilized as lead compounds. Further, we have covered mechanistic pathways to showcase the critical druggable targets and clinical and post-clinical drugs targeting them for their utility in BC.
Collapse
Affiliation(s)
- Chahat
- Department of Pharmaceutical Sciences, HNB Garhwal University, Chauras Campus, Srinagar, 246174, Uttarakhand, India
| | - Nidhi Nainwal
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Premanagar, Dehradun, 248007, Uttarakhand, India
| | - Yogesh Murti
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, India
| | - Savita Yadav
- IES Institute of Technology and Management, IES University, Bhopal, 462044, Madhya Pradesh, India
| | - Pramod Rawat
- Graphic Era (Deemed to Be University), Clement Town, Dehradun, 248002, India
- Graphic Era Hill University Clement Town, Dehradun, 248002, India
| | - Sonia Dhiman
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Bhupinder Kumar
- Department of Pharmaceutical Sciences, HNB Garhwal University, Chauras Campus, Srinagar, 246174, Uttarakhand, India.
| |
Collapse
|
2
|
Zhang Y, Qiu JG, Wang W, Sun FL, Wang X, Liu WJ, Jia XY, Ji H, Wang L, Jiang BH. Suppression of CYLD by HER3 confers ovarian cancer platinum resistance via inhibiting apoptosis and by inducing drug efflux. Exp Hematol Oncol 2025; 14:21. [PMID: 40012003 DOI: 10.1186/s40164-025-00620-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 02/18/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Ovarian cancer (OC) is the most pathogenic gynecological malignant tumor in the world. Due to the difficulty of early diagnosis, most of patients developed chemo-resistance and recurrence during/after chemotherapy. METHODS CCK8 and flow cytometry were utilized to assess drug sensitivity and apoptosis in parental and drug resistant cell lines. CYLD knockdown or overexpressed cells were employed to investigate its regulatory involvement in DDP resistance. Clinical tumor samples have been utilized to investigate the clinical relevance of CYLD. The drug synergistic effects were investigated through drug combination methods and a nude mice model with ABCB1 inhibitor or HER3 inhibitor. RESULTS In this study, we found that CYLD levels were significantly reduced in DDP-resistant cancer tissues and cells compared to the normal tissues and cells. CYLD knockdown in DDP-sensitive cells was sufficient to converse the cells to become DDP resistant by reducing cell apoptosis through increasing Bcl-XL and inhibiting Bax, and by increasing drug efflux via upregulating ABCB1 expression. HER3 expression levels were substantially higher in resistant cancer tissues and cells, and HER3 was the upstream facilitator of suppressing CYLD expression via STAT3 signaling. Furthermore, overexpression of CYLD in resistant cells increased sensitivity to platinum-based chemotherapy both in vitro and in vivo. ABCB1 was a key downstream target of CYLD for regulating tumor growth and therapeutic resistance both in vitro and in vivo, CYLD knockdown promoted the translocation of p65 to nucleus which increased ABCB1 expression through transcriptional activation. High expression levels of HER3 rendered CYLD suppression, consequently, mediated DDP resistance by blocking cell apoptosis pathways and promoting the drug efflux in ovarian cancer. CONCLUSIONS Our findings identify novel HER3/CYLD/ABCB1 axis that regulate tumor growth and DDP resistance, which may be used as potential novel therapeutic target(s) to overcome ovarian cancer DDP resistance.
Collapse
Affiliation(s)
- Ye Zhang
- Department of Gynecology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou University, Zhengzhou, 450000, China
| | - Jian-Ge Qiu
- Department of Gynecology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou University, Zhengzhou, 450000, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Wei Wang
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Fan-Li Sun
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Xue Wang
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Wen-Jing Liu
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, China
| | - Xiao-Yu Jia
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Hongbin Ji
- Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Lin Wang
- Department of Gynecology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou University, Zhengzhou, 450000, China.
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China.
| | - Bing-Hua Jiang
- Department of Gynecology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou University, Zhengzhou, 450000, China.
| |
Collapse
|
3
|
Kuehnemann C, Wiley CD. Senescent cells at the crossroads of aging, disease, and tissue homeostasis. Aging Cell 2024; 23:e13988. [PMID: 37731189 PMCID: PMC10776127 DOI: 10.1111/acel.13988] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/15/2023] [Accepted: 08/18/2023] [Indexed: 09/22/2023] Open
Abstract
Originally identified as an outcome of continuous culture of primary cells, cellular senescence has moved beyond the culture dish and is now a bona fide driver of aging and disease in animal models, and growing links to human disease. This cellular stress response consists of a stable proliferative arrest coupled to multiple phenotypic changes. Perhaps the most important of these is the senescence-associated secretory phenotype, or senescence-associated secretory phenotype -a complex and variable collection of secreted molecules release by senescent cells with a number of potent biological activities. Senescent cells appear in multiple age-associated conditions in humans and mice, and interventions that eliminate these cells can prevent or even reverse multiple diseases in mouse models. Here, we review salient aspects of senescent cells in the context of human disease and homeostasis. Senescent cells increase in abundance during several diseases that associated with premature aging. Conversely, senescent cells have a key role in beneficial processes such as development and wound healing, and thus can help maintain tissue homeostasis. Finally, we speculate on mechanisms by which deleterious aspects of senescent cells might be targeted while retaining homeostatic aspects in order to improve age-related outcomes.
Collapse
Affiliation(s)
- Chisaka Kuehnemann
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts UniversityBostonMassachusettsUSA
| | - Christopher D. Wiley
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts UniversityBostonMassachusettsUSA
| |
Collapse
|
4
|
Nugroho NT, Herten M, Torsello GF, Osada N, Marchiori E, Sielker S, Torsello GB. Association of Genetic Polymorphisms with Abdominal Aortic Aneurysm in the Processes of Apoptosis, Inflammation, and Cholesterol Metabolism. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1844. [PMID: 37893562 PMCID: PMC10608078 DOI: 10.3390/medicina59101844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/26/2023] [Accepted: 10/14/2023] [Indexed: 10/29/2023]
Abstract
Background and Objectives: This study aims to identify the minor allele of the single nucleotide polymorphisms (SNPs) DAB2IP rs7025486, IL6R rs2228145, CDKN2BAS rs10757278, LPA rs3798220, LRP1 rs1466535, and SORT1 rs599839 in order to assess the risk of abdominal aortic aneurysm (AAA) formation and define the linkage among these SNPs. Materials and Methods: A case-control study with AAA patients (AAA group) and non-AAA controls (control group) was carried out in a study population. DNA was isolated from whole blood samples; the SNPs were amplified using PCR and sequenced. Results: In the AAA group of 148 patients, 87.2% of the patients were male, 64.2% had a history of smoking, and 18.2% had relatives with AAA. The mean ± SD of age, BMI, and aneurysmal diameter in the AAA group were 74.8 ± 8.3 years, 27.6 ± 4.6 kg/m2, and 56.2 ± 11.8 mm, respectively. In comparison with 50 non-AAA patients, there was a significantly elevated presence of the SNPs DAB2IP rs7025486[A], CDKN2BAS rs10757278[G], and SORT1 rs599839[G] in the AAA group (p-values 0.040, 0.024, 0.035, respectively), while LPA rs3798220[C] was significantly higher in the control group (p = 0.049). A haplotype investigation showed that the SNPs DAB2IP, CDKN2BAS, and IL6R rs2228145[C] were significantly elevated in the AAA group (p = 0.037, 0.037, and 0.046) with minor allele frequencies (MAF) of 25.5%, 10.6%, and 15.4%, respectively. Only DAB2IP and CDKN2BAS showed significantly higher occurrences of a mutation (p = 0.028 and 0.047). Except for LPA, all SNPs were associated with a large aortic diameter in AAA (p < 0.001). Linkage disequilibrium detection showed that LPA to DAB2IP, to IL6R, to CDKN2BAS, and to LRP1 rs1466535[T] had D' values of 70.9%, 80.4%, 100%, and 100%, respectively. IL6R to LRP1 and to SORT1 had values for the coefficient of determination (r2) of 3.9% and 2.2%, respectively. Conclusions: In the investigated study population, the SNPs CDKN2BAS rs10757278, LPA rs3798220, SORT1 rs599839, DAB2IP rs7025486, and IL6R rs2228145 were associated with the development of abdominal aortic aneurysms. Individuals with risk factors for atherosclerosis and/or a family history of AAA should be evaluated using genetic analysis.
Collapse
Affiliation(s)
- Nyityasmono Tri Nugroho
- Department of Vascular and Endovascular Surgery, University Hospital Münster, 48149 Münster, Germany
- Vascular and Endovascular Division, Department of Surgery, Cipto Mangunkusumo National Hospital, Faculty of Medicine, University of Indonesia, Jakarta 10430, Indonesia
| | - Monika Herten
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Duisburg-Essen, 45147 Essen, Germany
| | | | - Nani Osada
- Department of Vascular and Endovascular Surgery, University Hospital Münster, 48149 Münster, Germany
| | - Elena Marchiori
- Department of Vascular and Endovascular Surgery, University Hospital Münster, 48149 Münster, Germany
| | - Sonja Sielker
- Research Unit Vascular Biology of Oral Structures (VABOS), Department of Cranio-Maxillofacial Surgery, University Hospital Münster, 48149 Münster, Germany
| | - Giovanni B. Torsello
- Institute for Vascular Research, St. Franziskus Hospital, 48145 Münster, Germany;
| |
Collapse
|
5
|
El-Tanani M, Nsairat H, Aljabali AA, Serrano-Aroca Á, Mishra V, Mishra Y, Naikoo GA, Alshaer W, Tambuwala MM. Role of mammalian target of rapamycin (mTOR) signalling in oncogenesis. Life Sci 2023; 323:121662. [PMID: 37028545 DOI: 10.1016/j.lfs.2023.121662] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/07/2023] [Accepted: 03/31/2023] [Indexed: 04/09/2023]
Abstract
The signalling system known as mammalian target of rapamycin (mTOR) is believed to be required for several biological activities involving cell proliferation. The serine-threonine kinase identified as mTOR recognises PI3K-AKT stress signals. It is well established in the scientific literature that the deregulation of the mTOR pathway plays a crucial role in cancer growth and advancement. This review focuses on the normal functions of mTOR as well as its abnormal roles in cancer development.
Collapse
Affiliation(s)
- Mohamed El-Tanani
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman 19328, Jordan; Institute of Cancer Therapeutics, University of Bradford, Bradford, West Yorkshire BD7 1DP, United Kingdom.
| | - Hamdi Nsairat
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Alaa A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Yarmouk University, Irbid 21163, Jordan.
| | - Ángel Serrano-Aroca
- Biomaterials and Bioengineering Laboratory, Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, c/Guillem de Castro 94, 46001, Valencia, Spain.
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, Punjab, India
| | - Yachana Mishra
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, 144411, Punjab, India
| | - Gowhar A Naikoo
- Department of Mathematics and Sciences, College of Arts and Applied Sciences, Dhofar University, Salalah, PC 211, Oman.
| | - Walhan Alshaer
- Cell Therapy Center, the University of Jordan, Amman 11942, Jordan
| | - Murtaza M Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool Campus, Lincoln LN6 7TS, United Kingdom.
| |
Collapse
|
6
|
Mall R, Bynigeri RR, Karki R, Malireddi RKS, Sharma B, Kanneganti TD. Pancancer transcriptomic profiling identifies key PANoptosis markers as therapeutic targets for oncology. NAR Cancer 2022; 4:zcac033. [PMID: 36329783 PMCID: PMC9623737 DOI: 10.1093/narcan/zcac033] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 10/03/2022] [Accepted: 10/28/2022] [Indexed: 11/24/2022] Open
Abstract
Resistance to programmed cell death (PCD) is a hallmark of cancer. While some PCD components are prognostic in cancer, the roles of many molecules can be masked by redundancies and crosstalks between PCD pathways, impeding the development of targeted therapeutics. Recent studies characterizing these redundancies have identified PANoptosis, a unique innate immune-mediated inflammatory PCD pathway that integrates components from other PCD pathways. Here, we designed a systematic computational framework to determine the pancancer clinical significance of PANoptosis and identify targetable biomarkers. We found that high expression of PANoptosis genes was detrimental in low grade glioma (LGG) and kidney renal cell carcinoma (KIRC). ZBP1, ADAR, CASP2, CASP3, CASP4, CASP8 and GSDMD expression consistently had negative effects on prognosis in LGG across multiple survival models, while AIM2, CASP3, CASP4 and TNFRSF10 expression had negative effects for KIRC. Conversely, high expression of PANoptosis genes was beneficial in skin cutaneous melanoma (SKCM), with ZBP1, NLRP1, CASP8 and GSDMD expression consistently having positive prognostic effects. As a therapeutic proof-of-concept, we treated melanoma cells with combination therapy that activates ZBP1 and showed that this treatment induced PANoptosis. Overall, through our systematic framework, we identified and validated key innate immune biomarkers from PANoptosis which can be targeted to improve patient outcomes in cancers.
Collapse
Affiliation(s)
- Raghvendra Mall
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Ratnakar R Bynigeri
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Rajendra Karki
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | | - Bhesh Raj Sharma
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | |
Collapse
|
7
|
Vaishnav M, Garg S, Adhaduk B. Association of smoking, p53 and Ki-67 immunomarkers with bladder neoplasms in tribal region of India. Int J Health Sci (Qassim) 2022; 16:11-17. [PMID: 36475036 PMCID: PMC9682877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023] Open
Abstract
OBJECTIVES Going back to the basics of urinary bladder neoplasms and correlation of histopathological diagnosis with age, sex, clinical features, smoking in tribal population of India in today's day and age. Along with that we are also determining the role of p53 and Ki67 immunomarkers in grading and staging of Urinary Bladder Neoplasms and their correlation with history of smoking. MATERIALS AND METHODS This retrospective study was done in a tertiary care hospital affiliated with medical college in tribal region of India. Total 72 cases of transurethral resection of bladder tumors and bladder biopsies were studied over a period of 2 years. The histopathological grading and staging was done according to the WHO/ISUP 2016 classification. The histopathological diagnosis was further correlated with age, sex, clinical features, and history of active smoking. Along with that immunomarkers p53 and Ki-67 were done and correlated with grading and staging of tumors and with history of smoking. The cut-off used was more than 20% positivity for high expression and < 20% for low expression. For descriptive, data median (Interquartile Range) and ratio were used. For categorical variables percentage (%), Fisher exact test and Chi-square test were used. Results were evaluated with SPSS software program. P < 0.05 was considered statistically significant. RESULTS Out of 72 cases studied, 66 cases were neoplastic. Out of them, bladder neoplasms were common in age group of 41-60 years and there was a male preponderance. Most common clinical feature was hematuria. Smoking was not a risk factor for the development of bladder neoplasms but bladder neoplasms in smokers were associated with higher grading and staging. The most common neoplasm was High Grade Papillary Carcinoma (43.05%). Stage T1 and Stage T2 were seen in 3.03% and 27.8% of cases. P53 and Ki-67 immunomarkers showed higher expression in tumors of higher grade and stage. CONCLUSION The take away from this study is bladder neoplasms that are quite versatile and histopathology is the gold standard to confirm these. However, p53and ki-67 gives important information regarding prognosis of tumors and helps in stratification of patients in high risk and low risk groups. Development of bladder carcinoma was independent of history of smoking; however, smoking is associated with higher grading and staging of bladder neoplasms and is also associated with higher expression of p53 and Ki-67 immunomarkers.
Collapse
Affiliation(s)
- Mitsu Vaishnav
- Department of Pathology, Namo Medical College and Research Institute, Silvassa, Gujarat, India
| | - Sameep Garg
- Department of Pathology, Namo Medical College and Research Institute, Silvassa, Gujarat, India
| | - Brinda Adhaduk
- Department of Pathology, Shri Vinoba Bhave Civil Hospital, Silvassa, Gujarat, India
| |
Collapse
|
8
|
Pinus roxburghii and Nauplius graveolens Extracts Elevate Apoptotic Gene Markers in C26 Colon Carcinoma Cells Induced in a BALB/c Mouse Model. SEPARATIONS 2022. [DOI: 10.3390/separations9100277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The present study aimed to evaluate the chemopreventive potential of Pinus roxburghii branch (P. roxburghii) and Nauplius graveolens (N. graveolens) extracts against human colorectal cancer (CRC) induced by C26 murine cells in a BALB/c mouse model. Real-time qRT-PCR was used to evaluate the apoptotic pathway by measuring the relative mRNA expression levels of the Bcl-2, Bax, Cas3, NF-κB, and PI3k genes. At the termination of the 30-day period, blood samples were collected to assay the biomarkers. The results showed a significant increase (p < 0.05) in the levels of TGF-β, CEA, CA19-9, malondialdehyde, ALT, AST, ALP, urea, and creatinine in the positive control compared to the negative control group. In addition, the glutathione reductase activity and total antioxidant activity were reduced in the positive control compared to the negative control. The biomarkers mentioned above were restored to almost normal levels after administering a safe dose (1/10) of a lethal dose of P. roxburghii and N. graveolens extracts. Administration of one-tenth of the LD50 of P. roxburghii and N. graveolens extracts caused a significant upregulation of the expression of Bax and Cas-3 and downregulation of the Bcl-2, NF-ĸB, and PI3k genes vs. the GAPDH gene as a housekeeping gene compared to the control group. Furthermore, the Bax/Bcl-2 ratio increased upon treatment. After administration of P. roxburghii and N. graveolens at a safe dose (1/10) of a lethal dose, the results showed improvement in both body weight gain and a significant decrease (p < 0.05) in tumor volume. Histopathological changes supported these improvements. Conclusively, the research outputs show that P. roxburghii and N. graveolens extracts can be utilized as potential chemopreventive agents for CRC treatment by stimulating cancer cell apoptosis and suppressing CRC survival and proliferation.
Collapse
|
9
|
Sanguinarine Induces H 2O 2-Dependent Apoptosis and Ferroptosis in Human Cervical Cancer. Biomedicines 2022; 10:biomedicines10081795. [PMID: 35892694 PMCID: PMC9331761 DOI: 10.3390/biomedicines10081795] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/15/2022] [Accepted: 06/15/2022] [Indexed: 01/31/2023] Open
Abstract
Sanguinarine (SNG) is a benzophenanthridine alkaloid isolated mainly from Sanguinaria canadensis, Chelidonium majus, and Macleaya cordata. SNG is considered an antineoplastic agent based on its cytotoxic activity against various tumors. However, the exact molecular mechanism through which SNG mediates this activity has not been elucidated. Here, we report that SNG induces death in human cervical cancer (HeLa) cells through activation of two interdependent cell death pathways—apoptosis and ferroptosis. SNG-induced apoptosis was characterized by caspase activation and PARP cleavage, while ferroptosis involved solute carrier family 7 member 11 (SLC7A11) down-regulation, glutathione (GSH) depletion, iron accumulation, and lipid peroxidation (LPO). Interestingly, incubation with caspase inhibitor z-VAD-fmk not only inhibited the features of apoptosis, but also negated markers of SNG-induced ferroptosis. Similarly, pretreatment with ferroptosis inhibitor ferrostatin-1 (Fer-1), apart from rescuing cells from SNG-induced ferroptosis, also curbed the features of SNG-induced apoptosis. Our study implies that, together, apoptosis and ferroptosis act as partners in the context of SNG mediated tumor suppression in HeLa cells. Importantly, SNG increased the generation of reactive oxygen species (ROS), and ROS inhibition blocks the induction of both apoptosis and ferroptosis. These findings highlight the value of continued investigation into the potential use of SNG as an antineoplastic agent.
Collapse
|
10
|
Oncogenic Kras-Mediated Cytokine CCL15 Regulates Pancreatic Cancer Cell Migration and Invasion through ROS. Cancers (Basel) 2022; 14:cancers14092153. [PMID: 35565279 PMCID: PMC9104113 DOI: 10.3390/cancers14092153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/19/2022] [Accepted: 04/21/2022] [Indexed: 11/30/2022] Open
Abstract
Simple Summary Oncogenic KrasG12D and tumor inflammation are critical components of the development and dissemination of pancreatic ductal adenocarcinoma (PDAC). The aim of this study is to investigate a lesser-known cytokine, CCL15, that functions as a new downstream target of KrasG12D with the purpose of regulating PDAC cell migration and invasion. We showed increased levels of CCL15 as well as the presence of its receptors, including CCR1 and CCR3, in PDAC tissues and cell lines. The knockdown of CCL15 diminished metastatic Panc-1 cell migration, whereas the treatment of CCL15 in non-metastatic BxPC-3 cells promoted BxPC-3 cell motility. Similar results were verified using murine metastatic PDAC KP-2 cells. Furthermore, we demonstrated that CCL15-modulated PDAC cell migration through the upregulation of cellular reactive oxygen species (ROS) levels and the knockdown of KrasG12D resulted in a decrease in CCL15. Altogether, our data unveiled a new mechanism of oncogenic KrasG12D in modulating PDAC inflammation and spreading. Abstract Pancreatic ductal adenocarcinoma (PDAC) is well known for its high death rate due to prompt cancer metastasis caused by cancer cell migration and invasion within the early stages of its development. Here, we reveal a new function of cytokine CCL15, namely the upregulation of PDAC cell migration and invasion. We showed increased levels of CCL15 transcripts and protein expressions in human PDAC tissue samples, as well as in cultured cell lines. Furthermore, PDAC cells also expressed CCL15 receptors, including CCR1 and CCR3. Murine PDAC cell lines and tissues strengthened this finding. The manipulation of CCL15 in metastatic Panc-1 cells through CCL15 knockdown or CCL15 neutralization decreased Panc-1 cell motility and invasiveness. In addition, treating non-metastatic BxPC-3 cells with recombinant CCL15 accelerated the cell migration of BxPC-3. A reduction in the levels of reactive oxygen species (ROS) by either N-Acetyl-L-Cysteine treatment or p22phox knockdown led to a decrease in Panc-1 cell migration and a reversed effect on recombinant CCL15-promoted BxPC-3 cell movement. Importantly, the knockdown of oncogenic Kras in Panc-1 cells abolished CCL15 protein expression and impeded cell migration without affecting PDAC cell growth. Altogether, our work elucidates an additional molecular pathway of oncogenic Kras to promote PDAC metastasis through the upregulation of cell migration and invasion by the Kras downstream CCL15, a lesser-known cytokine within the cancer research field.
Collapse
|
11
|
Dominguez-Martinez I, Joaquin-Ovalle F, Ferrer-Acosta Y, Griebenow KH. Folate-Decorated Cross-Linked Cytochrome c Nanoparticles for Active Targeting of Non-Small Cell Lung Carcinoma (NSCLC). Pharmaceutics 2022. [DOI: https://doi.org/10.3390/pharmaceutics14030490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The folate receptor alpha (FR), which is overexpressed in solid tumors including NSCLC, can be utilized for active tumor targeting to afford more effective cancer therapies. In this context, cytochrome c (Cyt c) has drawn attention to cancer research because it is non-toxic, yet, when delivered to the cytoplasm of cancer cells, can kill them by inducing apoptosis. Cyt c nanoparticles (NPs, 169 ± 9 nm) were obtained by solvent precipitation with acetonitrile, and stabilized by reversible homo-bifunctional crosslinking to accomplish a Cyt-c-based drug delivery system that combines stimulus-responsive release and active targeting. Cyt c was released under intracellular redox conditions, due to an S–S bond in the NPs linker, while NPs remained intact without any release under extracellular conditions. The NP surface was decorated with a hydrophilic folic acid–polyethylene glycol (FA–PEG) polymer for active targeting. The FA-decorated NPs specifically recognized and killed cancer cells (IC50 = 47.46 µg/mL) that overexpressed FR, but showed no toxicity against FR-negative cells. Confocal microscopy confirmed the preferential uptake and apoptosis induction of our NPs by FR-positive cancer cells. In vivo experiments using a Lewis lung carcinoma (LLC) mouse model showed visible NP accumulation within the tumor and inhibited the growth of LLC tumors.
Collapse
|
12
|
Folate-Decorated Cross-Linked Cytochrome c Nanoparticles for Active Targeting of Non-Small Cell Lung Carcinoma (NSCLC). Pharmaceutics 2022; 14:pharmaceutics14030490. [PMID: 35335867 PMCID: PMC8951294 DOI: 10.3390/pharmaceutics14030490] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 02/16/2022] [Accepted: 02/20/2022] [Indexed: 01/27/2023] Open
Abstract
The folate receptor alpha (FR), which is overexpressed in solid tumors including NSCLC, can be utilized for active tumor targeting to afford more effective cancer therapies. In this context, cytochrome c (Cyt c) has drawn attention to cancer research because it is non-toxic, yet, when delivered to the cytoplasm of cancer cells, can kill them by inducing apoptosis. Cyt c nanoparticles (NPs, 169 ± 9 nm) were obtained by solvent precipitation with acetonitrile, and stabilized by reversible homo-bifunctional crosslinking to accomplish a Cyt-c-based drug delivery system that combines stimulus-responsive release and active targeting. Cyt c was released under intracellular redox conditions, due to an S–S bond in the NPs linker, while NPs remained intact without any release under extracellular conditions. The NP surface was decorated with a hydrophilic folic acid–polyethylene glycol (FA–PEG) polymer for active targeting. The FA-decorated NPs specifically recognized and killed cancer cells (IC50 = 47.46 µg/mL) that overexpressed FR, but showed no toxicity against FR-negative cells. Confocal microscopy confirmed the preferential uptake and apoptosis induction of our NPs by FR-positive cancer cells. In vivo experiments using a Lewis lung carcinoma (LLC) mouse model showed visible NP accumulation within the tumor and inhibited the growth of LLC tumors.
Collapse
|
13
|
Dominguez-Martinez I, Joaquin-Ovalle F, Ferrer-Acosta Y, Griebenow KH. Folate-Decorated Cross-Linked Cytochrome c Nanoparticles for Active Targeting of Non-Small Cell Lung Carcinoma (NSCLC). Pharmaceutics 2022. [DOI: https:/doi.org/10.3390/pharmaceutics14030490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The folate receptor alpha (FR), which is overexpressed in solid tumors including NSCLC, can be utilized for active tumor targeting to afford more effective cancer therapies. In this context, cytochrome c (Cyt c) has drawn attention to cancer research because it is non-toxic, yet, when delivered to the cytoplasm of cancer cells, can kill them by inducing apoptosis. Cyt c nanoparticles (NPs, 169 ± 9 nm) were obtained by solvent precipitation with acetonitrile, and stabilized by reversible homo-bifunctional crosslinking to accomplish a Cyt-c-based drug delivery system that combines stimulus-responsive release and active targeting. Cyt c was released under intracellular redox conditions, due to an S–S bond in the NPs linker, while NPs remained intact without any release under extracellular conditions. The NP surface was decorated with a hydrophilic folic acid–polyethylene glycol (FA–PEG) polymer for active targeting. The FA-decorated NPs specifically recognized and killed cancer cells (IC50 = 47.46 µg/mL) that overexpressed FR, but showed no toxicity against FR-negative cells. Confocal microscopy confirmed the preferential uptake and apoptosis induction of our NPs by FR-positive cancer cells. In vivo experiments using a Lewis lung carcinoma (LLC) mouse model showed visible NP accumulation within the tumor and inhibited the growth of LLC tumors.
Collapse
|
14
|
Pouille CL, Ouaza S, Roels E, Behra J, Tourret M, Molinié R, Fontaine JX, Mathiron D, Gagneul D, Taminiau B, Daube G, Ravallec R, Rambaud C, Hilbert JL, Cudennec B, Lucau-Danila A. Chicory: Understanding the Effects and Effectors of This Functional Food. Nutrients 2022; 14:957. [PMID: 35267932 PMCID: PMC8912540 DOI: 10.3390/nu14050957] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 02/22/2022] [Indexed: 02/06/2023] Open
Abstract
Industrial chicory has been the subject of numerous studies, most of which provide clinical observations on its health effects. Whether it is the roasted root, the flour obtained from the roots or the different classes of molecules that enter into the composition of this plant, understanding the molecular mechanisms of action on the human organism remains incomplete. In this study, we were interested in three molecules or classes of molecules present in chicory root: fructose, chlorogenic acids, and sesquiterpene lactones. We conducted experiments on the murine model and performed a nutrigenomic analysis, a metabolic hormone assay and a gut microbiota analysis, associated with in vitro observations for different responses. We have highlighted a large number of effects of all these classes of molecules that suggest a pro-apoptotic activity, an anti-inflammatory, antimicrobial, antioxidant, hypolipidemic and hypoglycemic effect and also an important role in appetite regulation. A significant prebiotic activity was also identified. Fructose seems to be the most involved in these activities, contributing to approximately 83% of recorded responses, but the other classes of tested molecules have shown a specific role for these different effects, with an estimated contribution of 23-24%.
Collapse
Affiliation(s)
- Céline L. Pouille
- UMR Transfrontalière BioEcoAgro N° 1158, Univ. Lille, INRAE, Univ. Liège, UPJV, JUNIA, Univ. Artois, Univ. Littoral Côte d’Opale, ICV, SFR Condorcet FR CNRS 3417—Institut Charles Viollette, 59655 Villeneuve d’Ascq, France; (C.L.P.); (S.O.); (E.R.); (J.B.); (M.T.); (D.G.); (R.R.); (C.R.); (J.-L.H.); (B.C.)
- Joint Laboratory CHIC41H University of Lille-Florimond-Desprez, Cité scientifique, 59655 Villeneuve d’Ascq, France
| | - Souad Ouaza
- UMR Transfrontalière BioEcoAgro N° 1158, Univ. Lille, INRAE, Univ. Liège, UPJV, JUNIA, Univ. Artois, Univ. Littoral Côte d’Opale, ICV, SFR Condorcet FR CNRS 3417—Institut Charles Viollette, 59655 Villeneuve d’Ascq, France; (C.L.P.); (S.O.); (E.R.); (J.B.); (M.T.); (D.G.); (R.R.); (C.R.); (J.-L.H.); (B.C.)
- Joint Laboratory CHIC41H University of Lille-Florimond-Desprez, Cité scientifique, 59655 Villeneuve d’Ascq, France
| | - Elise Roels
- UMR Transfrontalière BioEcoAgro N° 1158, Univ. Lille, INRAE, Univ. Liège, UPJV, JUNIA, Univ. Artois, Univ. Littoral Côte d’Opale, ICV, SFR Condorcet FR CNRS 3417—Institut Charles Viollette, 59655 Villeneuve d’Ascq, France; (C.L.P.); (S.O.); (E.R.); (J.B.); (M.T.); (D.G.); (R.R.); (C.R.); (J.-L.H.); (B.C.)
- Joint Laboratory CHIC41H University of Lille-Florimond-Desprez, Cité scientifique, 59655 Villeneuve d’Ascq, France
| | - Josette Behra
- UMR Transfrontalière BioEcoAgro N° 1158, Univ. Lille, INRAE, Univ. Liège, UPJV, JUNIA, Univ. Artois, Univ. Littoral Côte d’Opale, ICV, SFR Condorcet FR CNRS 3417—Institut Charles Viollette, 59655 Villeneuve d’Ascq, France; (C.L.P.); (S.O.); (E.R.); (J.B.); (M.T.); (D.G.); (R.R.); (C.R.); (J.-L.H.); (B.C.)
| | - Melissa Tourret
- UMR Transfrontalière BioEcoAgro N° 1158, Univ. Lille, INRAE, Univ. Liège, UPJV, JUNIA, Univ. Artois, Univ. Littoral Côte d’Opale, ICV, SFR Condorcet FR CNRS 3417—Institut Charles Viollette, 59655 Villeneuve d’Ascq, France; (C.L.P.); (S.O.); (E.R.); (J.B.); (M.T.); (D.G.); (R.R.); (C.R.); (J.-L.H.); (B.C.)
| | - Roland Molinié
- UMR Transfontalière BioEcoAgro N° 1158, Univ. Lille, INRAE, Univ. Liège, UPJV, JUNIA, Univ. Artois, Univ. Littoral Côte d’Opale, ICV, SFR Condorcet FR CNRS 3417—BIOlogie des Plantes et Innovation (BIOPI), 80025 Amiens, France; (R.M.); (J.-X.F.)
| | - Jean-Xavier Fontaine
- UMR Transfontalière BioEcoAgro N° 1158, Univ. Lille, INRAE, Univ. Liège, UPJV, JUNIA, Univ. Artois, Univ. Littoral Côte d’Opale, ICV, SFR Condorcet FR CNRS 3417—BIOlogie des Plantes et Innovation (BIOPI), 80025 Amiens, France; (R.M.); (J.-X.F.)
| | - David Mathiron
- Plateforme Analytique UFR des Sciences, UPJV, Bâtiment Serres-Transfert Rue Dallery-Passage du Sourire d’Avril, 80039 Amiens, France;
| | - David Gagneul
- UMR Transfrontalière BioEcoAgro N° 1158, Univ. Lille, INRAE, Univ. Liège, UPJV, JUNIA, Univ. Artois, Univ. Littoral Côte d’Opale, ICV, SFR Condorcet FR CNRS 3417—Institut Charles Viollette, 59655 Villeneuve d’Ascq, France; (C.L.P.); (S.O.); (E.R.); (J.B.); (M.T.); (D.G.); (R.R.); (C.R.); (J.-L.H.); (B.C.)
- Joint Laboratory CHIC41H University of Lille-Florimond-Desprez, Cité scientifique, 59655 Villeneuve d’Ascq, France
| | - Bernard Taminiau
- Department of Food Sciences–Microbiology, FARAH, University of Liege, 4000 Liege, Belgium; (B.T.); (G.D.)
| | - Georges Daube
- Department of Food Sciences–Microbiology, FARAH, University of Liege, 4000 Liege, Belgium; (B.T.); (G.D.)
| | - Rozenn Ravallec
- UMR Transfrontalière BioEcoAgro N° 1158, Univ. Lille, INRAE, Univ. Liège, UPJV, JUNIA, Univ. Artois, Univ. Littoral Côte d’Opale, ICV, SFR Condorcet FR CNRS 3417—Institut Charles Viollette, 59655 Villeneuve d’Ascq, France; (C.L.P.); (S.O.); (E.R.); (J.B.); (M.T.); (D.G.); (R.R.); (C.R.); (J.-L.H.); (B.C.)
| | - Caroline Rambaud
- UMR Transfrontalière BioEcoAgro N° 1158, Univ. Lille, INRAE, Univ. Liège, UPJV, JUNIA, Univ. Artois, Univ. Littoral Côte d’Opale, ICV, SFR Condorcet FR CNRS 3417—Institut Charles Viollette, 59655 Villeneuve d’Ascq, France; (C.L.P.); (S.O.); (E.R.); (J.B.); (M.T.); (D.G.); (R.R.); (C.R.); (J.-L.H.); (B.C.)
- Joint Laboratory CHIC41H University of Lille-Florimond-Desprez, Cité scientifique, 59655 Villeneuve d’Ascq, France
| | - Jean-Louis Hilbert
- UMR Transfrontalière BioEcoAgro N° 1158, Univ. Lille, INRAE, Univ. Liège, UPJV, JUNIA, Univ. Artois, Univ. Littoral Côte d’Opale, ICV, SFR Condorcet FR CNRS 3417—Institut Charles Viollette, 59655 Villeneuve d’Ascq, France; (C.L.P.); (S.O.); (E.R.); (J.B.); (M.T.); (D.G.); (R.R.); (C.R.); (J.-L.H.); (B.C.)
- Joint Laboratory CHIC41H University of Lille-Florimond-Desprez, Cité scientifique, 59655 Villeneuve d’Ascq, France
| | - Benoit Cudennec
- UMR Transfrontalière BioEcoAgro N° 1158, Univ. Lille, INRAE, Univ. Liège, UPJV, JUNIA, Univ. Artois, Univ. Littoral Côte d’Opale, ICV, SFR Condorcet FR CNRS 3417—Institut Charles Viollette, 59655 Villeneuve d’Ascq, France; (C.L.P.); (S.O.); (E.R.); (J.B.); (M.T.); (D.G.); (R.R.); (C.R.); (J.-L.H.); (B.C.)
| | - Anca Lucau-Danila
- UMR Transfrontalière BioEcoAgro N° 1158, Univ. Lille, INRAE, Univ. Liège, UPJV, JUNIA, Univ. Artois, Univ. Littoral Côte d’Opale, ICV, SFR Condorcet FR CNRS 3417—Institut Charles Viollette, 59655 Villeneuve d’Ascq, France; (C.L.P.); (S.O.); (E.R.); (J.B.); (M.T.); (D.G.); (R.R.); (C.R.); (J.-L.H.); (B.C.)
- Joint Laboratory CHIC41H University of Lille-Florimond-Desprez, Cité scientifique, 59655 Villeneuve d’Ascq, France
| |
Collapse
|
15
|
Karki R, Sundaram B, Sharma BR, Lee S, Malireddi RKS, Nguyen LN, Christgen S, Zheng M, Wang Y, Samir P, Neale G, Vogel P, Kanneganti TD. ADAR1 restricts ZBP1-mediated immune response and PANoptosis to promote tumorigenesis. Cell Rep 2021; 37:109858. [PMID: 34686350 PMCID: PMC8853634 DOI: 10.1016/j.celrep.2021.109858] [Citation(s) in RCA: 226] [Impact Index Per Article: 56.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 08/12/2021] [Accepted: 09/28/2021] [Indexed: 01/04/2023] Open
Abstract
Cell death provides host defense and maintains homeostasis. Zα-containing molecules are essential for these processes. Z-DNA binding protein 1 (ZBP1) activates inflammatory cell death, PANoptosis, whereas adenosine deaminase acting on RNA 1 (ADAR1) serves as an RNA editor to maintain homeostasis. Here, we identify and characterize ADAR1's interaction with ZBP1, defining its role in cell death regulation and tumorigenesis. Combining interferons (IFNs) and nuclear export inhibitors (NEIs) activates ZBP1-dependent PANoptosis. ADAR1 suppresses this PANoptosis by interacting with the Zα2 domain of ZBP1 to limit ZBP1 and RIPK3 interactions. Adar1fl/flLysMcre mice are resistant to development of colorectal cancer and melanoma, but deletion of the ZBP1 Zα2 domain restores tumorigenesis in these mice. In addition, treating wild-type mice with IFN-γ and the NEI KPT-330 regresses melanoma in a ZBP1-dependent manner. Our findings suggest that ADAR1 suppresses ZBP1-mediated PANoptosis, promoting tumorigenesis. Defining the functions of ADAR1 and ZBP1 in cell death is fundamental to informing therapeutic strategies for cancer and other diseases.
Collapse
MESH Headings
- Adenosine Deaminase/genetics
- Adenosine Deaminase/metabolism
- Animals
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Cell Death
- Cell Transformation, Neoplastic/immunology
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Colorectal Neoplasms/drug therapy
- Colorectal Neoplasms/enzymology
- Colorectal Neoplasms/immunology
- Colorectal Neoplasms/pathology
- Female
- Gene Expression Regulation, Neoplastic
- HEK293 Cells
- Humans
- Hydrazines/pharmacology
- Interferon-gamma/pharmacology
- Male
- Melanoma, Experimental/drug therapy
- Melanoma, Experimental/enzymology
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Mice, Inbred C57BL
- Mice, Knockout
- Necroptosis
- Pyroptosis
- RNA-Binding Proteins/genetics
- RNA-Binding Proteins/metabolism
- Receptor-Interacting Protein Serine-Threonine Kinases/genetics
- Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
- Signal Transduction
- Skin Neoplasms/drug therapy
- Skin Neoplasms/enzymology
- Skin Neoplasms/immunology
- Skin Neoplasms/pathology
- Triazoles/pharmacology
- Mice
Collapse
Affiliation(s)
- Rajendra Karki
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Balamurugan Sundaram
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Bhesh Raj Sharma
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - SangJoon Lee
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | | - Lam Nhat Nguyen
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Shelbi Christgen
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Min Zheng
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yaqiu Wang
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Parimal Samir
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Geoffrey Neale
- Hartwell Center for Bioinformatics & Biotechnology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Peter Vogel
- Animal Resources Center and Veterinary Pathology Core, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | |
Collapse
|
16
|
Bostan M, Mihaila M, Petrica-Matei GG, Radu N, Hainarosie R, Stefanescu CD, Roman V, Diaconu CC. Resveratrol Modulation of Apoptosis and Cell Cycle Response to Cisplatin in Head and Neck Cancer Cell Lines. Int J Mol Sci 2021; 22:6322. [PMID: 34204834 PMCID: PMC8231609 DOI: 10.3390/ijms22126322] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/04/2021] [Accepted: 06/08/2021] [Indexed: 12/13/2022] Open
Abstract
In head and neck cancers, the effectiveness of cisplatin (CisPt) treatment is limited by its toxicity, especially when higher doses are necessary, and the possible occurrence of cisplatin resistance. This study evaluated the effects of resveratrol (RSV) on the expression of different genes involved in the response of human tumor cells (FaDu, PE/CA-PJ49) to cisplatin therapy. Our results revealed that RSV induced apoptosis amplification in both FaDu and PE/CA-PJ49 cells and modulated the expression of specific genes differently than in normal HaCaT cells. In FaDu cells, combined CisPt + RSV treatment induced an increase in apoptosis, which was associated with an increase in c-MYC and TP53 and a decrease in BCL-2 expression. While CisPt + RSV treatment induced apoptosis in PE/CA-PJ49 cells by inhibition of BCL-2 associated with high levels of MDM-2 and subsequently led to inhibition of TP53 gene expression. Decreased c-MYC expression in PE/CA-PJ49 treated with CisPt + RSV was accompanied by cell cycle blockage in G0/G1 phase. In conclusion, RSV influences tumor cell response to CisPt by inducing apoptosis and modulating gene expression. In addition, in normal HaCaT cells, RSV was able to reduce the harmful effects of CisPt.
Collapse
Affiliation(s)
- Marinela Bostan
- Center of Immunology, Stefan S. Nicolau Institute of Virology, 030304 Bucharest, Romania; (M.B.); (M.M.)
- Department of Immunology, Victor Babes National Institute of Pathology, 050096 Bucharest, Romania
| | - Mirela Mihaila
- Center of Immunology, Stefan S. Nicolau Institute of Virology, 030304 Bucharest, Romania; (M.B.); (M.M.)
| | | | - Nicoleta Radu
- Department of Biotechnology, University of Agronomic Sciences and Veterinary Medicine of Bucharest, 011464 Bucharest, Romania
- Biotechnology Department, National Institute for Chemistry and Petrochemistry R&D of Bucharest, 060021 Bucharest, Romania
| | - Razvan Hainarosie
- Otorhinolaryngology and Head and Neck Surgery Department—Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (R.H.); (C.D.S.)
| | - Cristian Dragos Stefanescu
- Otorhinolaryngology and Head and Neck Surgery Department—Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (R.H.); (C.D.S.)
| | - Viviana Roman
- Center of Immunology, Stefan S. Nicolau Institute of Virology, 030304 Bucharest, Romania; (M.B.); (M.M.)
| | - Carmen Cristina Diaconu
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, 030304 Bucharest, Romania;
| |
Collapse
|
17
|
Arulananda S, Lee EF, Fairlie WD, John T. The role of BCL-2 family proteins and therapeutic potential of BH3-mimetics in malignant pleural mesothelioma. Expert Rev Anticancer Ther 2020; 21:413-424. [PMID: 33238762 DOI: 10.1080/14737140.2021.1856660] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction: With limited recent therapeutic changes, malignant pleural mesothelioma (MPM) is associated with poor survival and death within 12 months, making it one of the most lethal malignancies. Due to unregulated asbestos use in developing countries and home renovation exposures, cases of MPM are likely to present for decades. As MPM is largely driven by dysregulation of tumor suppressor genes, researchers have examined other mechanisms of subverting tumor proliferation and spread. Over-expression of pro-survival BCL-2 family proteins impairs cells from undergoing apoptosis, and BH3-mimetics targeting them are a novel treatment option across various cancers, though have not been widely investigated in MPM.Areas covered: This review provides an overview of MPM and its current treatment landscape. It summarizes the role of BCL-2 family proteins in tumorigenesis and the therapeutic potential of BH3-mimetics . Finally, it discusses the role of BCL-2 proteins in MPM and the pre-clinical rationale for investigating BH3-mimetics as a therapeutic strategy.Expert opinion: As a disease without readily actionable oncogene driver mutations and with modest benefit from immune checkpoint inhibition, novel therapeutic options are urgently needed for MPM. Hence, BH3-mimetics provide a promising treatment option, with evidence supporting dependence on pro-survival BCL-2 proteins for MPM cell survival.
Collapse
Affiliation(s)
- Surein Arulananda
- Department of Medical Oncology, Austin Health, Heidelberg, Australia.,Olivia Newton-John Cancer Research Institute, Heidelberg, Australia.,School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Erinna F Lee
- Olivia Newton-John Cancer Research Institute, Heidelberg, Australia.,School of Cancer Medicine, La Trobe University, Heidelberg, Australia.,Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Victoria, Australia
| | - W Douglas Fairlie
- Olivia Newton-John Cancer Research Institute, Heidelberg, Australia.,School of Cancer Medicine, La Trobe University, Heidelberg, Australia.,Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Victoria, Australia
| | - Thomas John
- Olivia Newton-John Cancer Research Institute, Heidelberg, Australia.,School of Cancer Medicine, La Trobe University, Heidelberg, Australia.,Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| |
Collapse
|
18
|
NPD1 rapidly targets mitochondria-mediated apoptosis after acute injection protecting brain against ischemic injury. Exp Neurol 2020; 335:113495. [PMID: 33038416 DOI: 10.1016/j.expneurol.2020.113495] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/20/2020] [Accepted: 10/05/2020] [Indexed: 12/19/2022]
Abstract
Mitochondria-related cell death pathways play a major role in ischemic brain injury. Thus, mitochondrial "protective" molecules could be considered for new therapeutic regimens. We recently reported that acute administration of docosahexaenoic acid (DHA) triglyceride lipid emulsion, immediately after hypoxic-ischemic (HI) insult, markedly attenuated brain infarct size. This was associated with an early change of DHA-derived specialized pro-resolving mediator (SPM) profiles. Specifically, DHA treatment induced a 50% increase of neuroprotectin D1 (NPD1) levels in ischemic brain. Based on these findings, we questioned if direct administration of NPD1 after HI injury also affords neuroprotection, and if so, by what mechanisms. Using HI insult to mimic ischemic stroke in neonatal mice, we observed that acute intraperitoneal injection of NPD1 immediately after HI injury prevented the expansion of the ischemic core by ~40% and improved coordination and motor abilities compared to the control group. At 7 days after HI injury, NPD1 treatment decreased ipsilateral hemisphere atrophy and preserved motor functions in wire-holding and bridge-crossing tests compared to control littermates. Brain mitochondria, isolated at 4 h after reperfusion from mice treated with NPD1, showed an increase in the capacity to buffer calcium after HI injury, as result of the preservation of mitochondrial membranes. Further, NPD1 induced a reduction of mitochondrial BAX translocation and oligomerization, attenuated cytochrome C release and decreased AIF nuclear translocation. To confirm whether NPD1 acts as BAX inhibitor, we evaluated NPD1 action co-administrated with a pro-apoptotic agent, staurosporine, using mouse embryonic fibroblasts as in vitro model of apoptosis. NPD1 exposure markedly decreased mitochondria-mediated apoptosis, blocking BAX translocation from cytosol to mitochondria and subsequently reducing caspase-3 activation. Our findings provide novel evidence that the neuroprotective action of NPD1 is elicited rapidly in the first few hours after ischemic injury and is associated with both preserved mitochondrial membrane structure and reduced BAX mitochondrial translocation and activation.
Collapse
|
19
|
El‐Mesery M, Seher A, El‐Shafey M, El‐Dosoky M, Badria FA. Repurposing of quinoline alkaloids identifies their ability to enhance doxorubicin‐induced sub‐G0/G1 phase cell cycle arrest and apoptosis in cervical and hepatocellular carcinoma cells. Biotechnol Appl Biochem 2020; 68:832-840. [DOI: 10.1002/bab.1999] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/27/2020] [Indexed: 12/24/2022]
Affiliation(s)
- Mohamed El‐Mesery
- Department of Biochemistry, Faculty of Pharmacy Mansoura University Mansoura Egypt
| | - Axel Seher
- Department of Oral and Maxillofacial Plastic Surgery University Hospital Wuerzburg Wuerzburg Germany
| | - Mohamed El‐Shafey
- Department of Anatomy and Embryology, Faculty of Medicine Mansoura University Egypt
- Physiological Sciences Department Fakeeh College for Medical Sciences Jeddah Saudi Arabia
| | - Mohamed El‐Dosoky
- Department of Neuroscience Technology, College of Applied Medical Science in Jubail Imam Abdulalrahman Bin Faisal University Dammam Saudi Arabia
| | - Farid A Badria
- Department of Pharmacognosy, Faculty of Pharmacy Mansoura University Egypt
| |
Collapse
|
20
|
Mishra RK, Ahmad A, Vyawahare A, Kumar A, Khan R. Understanding the Monoclonal Antibody Involvement in Targeting the Activation of Tumor Suppressor Genes. Curr Top Med Chem 2020; 20:1810-1823. [PMID: 32543361 DOI: 10.2174/1568026620666200616133814] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/28/2020] [Accepted: 05/08/2020] [Indexed: 12/14/2022]
Abstract
Monoclonal antibodies (mAbs) have always provided outstanding therapeutic arsenal in the
treatment of cancer, be it hematological malignancies or solid tumors. Monoclonal antibodies mediated
targeting of cancer genes in general and tumor-suppressor genes, in particular, have appreciably allowed
the possibilities of trafficking these antibodies to specific tumor mechanisms and aim for the pin-point
maneuvered tumor treatment strategies. The conventional cancer treatment options are associated with
enormous limitations like drug resistance, acute and pan-toxic side effects and collateral damage to other
unrelated cells and organs. Therefore, monoclonal antibody-mediated treatments have some special advantages
of specific targeting of cancer-related genes and minimizing the off-target side effects. A large
number of monoclonal antibody-mediated treatment regimen viz. use of immunoconjugates, clinically
targeting TGFβ with pan-TGFβ monoclonal antibodies, p53 by its monoclonal antibodies and EGFRtargeted
monoclonal antibodies, etc. have been observed in the recent past. In this review, the authors
have discussed some of the significant advances in the context of targeting tumor suppressor genes with
monoclonal antibodies. Approximately 250 articles were scanned from research databases like PubMed
central, Europe PubMed Central and google scholar up to the date of inception, and relevant reports on
monoclonal antibody-mediated targeting of cancer genes were selected. mAb mediated targeting of tumor
suppressor genes is a recent grey paradigm, which has not been explored up to its maximum potential.
Therefore, this review will be of appreciable significance that it will boost further in-depth understanding
of various aspects of mAb arbitrated cancer targeting and will warrant and promote further rigorous
research initiatives in this regard. The authors expect that this review will acquaint the readers
with the current status regarding the recent progress in the domain of mAbs and their employability and
targetability towards tumor suppressor genes in anti-cancer therapeutics.
Collapse
Affiliation(s)
- Rakesh Kumar Mishra
- Department of Nano-Therapeutics, Institute of Nano Science and Technology, Habitat Centre, Phase 10, Sector 64, Mohali, Punjab 160062, India
| | - Anas Ahmad
- Department of Nano-Therapeutics, Institute of Nano Science and Technology, Habitat Centre, Phase 10, Sector 64, Mohali, Punjab 160062, India
| | - Akshay Vyawahare
- Department of Nano-Therapeutics, Institute of Nano Science and Technology, Habitat Centre, Phase 10, Sector 64, Mohali, Punjab 160062, India
| | - Ajay Kumar
- Department of Nano-Therapeutics, Institute of Nano Science and Technology, Habitat Centre, Phase 10, Sector 64, Mohali, Punjab 160062, India
| | - Rehan Khan
- Department of Nano-Therapeutics, Institute of Nano Science and Technology, Habitat Centre, Phase 10, Sector 64, Mohali, Punjab 160062, India
| |
Collapse
|
21
|
Queiroz FC, Vargas AMP, Oliveira MGA, Comarela GV, Silveira SA. ppiGReMLIN: a graph mining based detection of conserved structural arrangements in protein-protein interfaces. BMC Bioinformatics 2020; 21:143. [PMID: 32293241 PMCID: PMC7158050 DOI: 10.1186/s12859-020-3474-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 03/27/2020] [Indexed: 12/15/2022] Open
Abstract
Background Protein-protein interactions (PPIs) are fundamental in many biological processes and understanding these interactions is key for a myriad of applications including drug development, peptide design and identification of drug targets. The biological data deluge demands efficient and scalable methods to characterize and understand protein-protein interfaces. In this paper, we present ppiGReMLIN, a graph based strategy to infer interaction patterns in a set of protein-protein complexes. Our method combines an unsupervised learning strategy with frequent subgraph mining in order to detect conserved structural arrangements (patterns) based on the physicochemical properties of atoms on protein interfaces. To assess the ability of ppiGReMLIN to point out relevant conserved substructures on protein-protein interfaces, we compared our results to experimentally determined patterns that are key for protein-protein interactions in 2 datasets of complexes, Serine-protease and BCL-2. Results ppiGReMLIN was able to detect, in an automatic fashion, conserved structural arrangements that represent highly conserved interactions at the specificity binding pocket of trypsin and trypsin-like proteins from Serine-protease dataset. Also, for the BCL-2 dataset, our method pointed out conserved arrangements that include critical residue interactions within the conserved motif LXXXXD, pivotal to the binding specificity of BH3 domains of pro-apoptotic BCL-2 proteins towards apoptotic suppressors. Quantitatively, ppiGReMLIN was able to find all of the most relevant residues described in literature for our datasets, showing precision of at least 69% up to 100% and recall of 100%. Conclusions ppiGReMLIN was able to find highly conserved structures on the interfaces of protein-protein complexes, with minimum support value of 60%, in datasets of similar proteins. We showed that the patterns automatically detected on protein interfaces by our method are in agreement with interaction patterns described in the literature.
Collapse
Affiliation(s)
- Felippe C Queiroz
- Department of Computer Science, Universidade Federal de Viçosa, Av Peter Henry Rolfs, Viçosa, MG, Brazil.
| | - Adriana M P Vargas
- Department of Biochemistry and Molecular Biology, Universidade Federal de Viçosa, Av Peter Henry Rolfs, Viçosa, MG, Brazil
| | - Maria G A Oliveira
- Department of Biochemistry and Molecular Biology, Universidade Federal de Viçosa, Av Peter Henry Rolfs, Viçosa, MG, Brazil.,Instituto de Biotecnologia aplicada a Agropecuaria, BIOAGRO-UFV, Av Peter Henry Rolfs, Viçosa MG, Brazil
| | - Giovanni V Comarela
- Department of Computer Science, Universidade Federal do Espírito Santo, Av Fernando Ferrari, Vitória, ES, Brazil
| | - Sabrina A Silveira
- Department of Computer Science, Universidade Federal de Viçosa, Av Peter Henry Rolfs, Viçosa, MG, Brazil.,European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, CB10 1SD, UK
| |
Collapse
|
22
|
Merino-Valverde I, Greco E, Abad M. The microproteome of cancer: From invisibility to relevance. Exp Cell Res 2020; 392:111997. [PMID: 32302626 DOI: 10.1016/j.yexcr.2020.111997] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 04/02/2020] [Accepted: 04/03/2020] [Indexed: 01/08/2023]
Abstract
Recent findings have revealed that many genomic regions previously annotated as non-protein coding actually contain small open reading frames, smaller that 300 bp, that are transcribed and translated into evolutionary conserved microproteins. To date, only a small subset of them have been functionally characterized, but they play key functions in fundamental processes such as DNA repair, RNA processing and metabolism regulation. This emergent field seems to hide a new category of molecular regulators with clinical potential. In this review, we focus on its relevance for cancer. Following Hanahan and Weinberg's classification of the hallmarks of cancer, we provide an overview of those microproteins known to be implicated in cancer or those that, based on their function, are likely to play a role in cancer. The resulting picture is that while we are at the very early times of this field, it holds the promise to provide crucial information to understand cancer biology.
Collapse
Affiliation(s)
| | - Emanuela Greco
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, 08035, Spain
| | - María Abad
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, 08035, Spain.
| |
Collapse
|
23
|
Seervi M, Sumi S, Chandrasekharan A, Sharma AK, SanthoshKumar TR. Molecular profiling of anastatic cancer cells: potential role of the nuclear export pathway. Cell Oncol (Dordr) 2019; 42:645-661. [PMID: 31147963 DOI: 10.1007/s13402-019-00451-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2019] [Indexed: 01/11/2023] Open
Abstract
PURPOSE Anastasis is newly discovered process by which cells recover from late-stage apoptosis upon removal of a death stimulus. Recent reports suggest that cells may recover, even after the initiation of mitochondrial outer-membrane permeabilization (MOMP) and caspase activation. Here, we specifically studied the reversibility of late-stage apoptosis in cervical (HeLa) and breast (MDA-MB-231) cancer cells in relation to the extent of MOMP (limited or widespread). In addition, we explored the molecular factors involved in the anastatic process. METHODS The extent of MOMP was assessed using time lapse confocal microscopic imaging, considering mitochondrial cytochrome c-GFP release as a marker for MOMP. Anastatic cells were generated by specifically recovering late-stage apoptotic (annexin V/PI positive) cervical and breast cancer cells. Molecular signaling events involved in death reversal were assessed using LC-MS/MS and qRT-PCR. Targeted chemical inhibition and shRNA-based gene silencing studies were employed to explore the role of the nuclear export pathway in anastasis and increased oncogenicity. RESULTS Time-lapse imaging of drug-treated Cyt-c-GFP expressing cancer cells revealed cell recovery despite widespread MOMP. A few recovered anastatic cells were noted and these were found to proliferate through a selection-type of survival. They showed increased drug-resistance, migration and invasive potential compared to non-anastatic cancer cells. Network analysis using 49 proteins uniquely expressed in anastatic cells indicated upregulation of nuclear export/import, redox and Ras signaling pathways in both HeLa and MDA-MB-231 anastatic cells, indicating common molecular mechanisms in different cell types. Inhibition of XPO1 significantly reduced the recovery of apoptotic cells and abrogated acquired oncogenic transformation in the anastatic cancer cells. CONCLUSIONS Our study indicates that cancer cells can revert from apoptosis even after the induction of widespread MOMP. We noted a significant role of the nuclear-export pathway in the anastatic process of cancer cells. Inhibition of anastasis through the nuclear export pathway may be a potential therapeutic strategy for targeting drug-resistance, metastasis and recurrence problems during cancer treatment.
Collapse
Affiliation(s)
- Mahendra Seervi
- DBT-PU-IPLS, Department of Botany/Biotechnology, Patna University, Patna, Bihar, India.
- Department of Biological Sciences, P. D. Patel Institute of Applied Sciences, Charotar University of Science and Technology (CHARUSAT), Changa, Anand, Gujarat, India.
| | - S Sumi
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Aneesh Chandrasekharan
- Cancer Research Division 1, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Abhay K Sharma
- DBT-PU-IPLS, Department of Botany/Biotechnology, Patna University, Patna, Bihar, India
| | - T R SanthoshKumar
- Cancer Research Division 1, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| |
Collapse
|
24
|
Yin S, Qiu Y, Jin C, Wang R, Wu S, Liu H, Koo S, Han L, Zhang Y, Gao X, Pang X, Wang T, Yu H. 7‐Deoxynarciclasine shows promising antitumor efficacy by targeting Akt against hepatocellular carcinoma. Int J Cancer 2019; 145:3334-3346. [DOI: 10.1002/ijc.32395] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 05/02/2019] [Indexed: 01/17/2023]
Affiliation(s)
- Shuangshuang Yin
- Tianjin State Key Laboratory of Modern Chinese MedicineTianjin University of Traditional Chinese Medicine Tianjin China
| | - Yuling Qiu
- School of PharmacyTianjin Medical University Tianjin China
| | - Chengyun Jin
- School of Pharmaceutical Sciences, Key Laboratory of State Ministry of Education, Key Laboratory of Henan province for Drug Quality Control and EvaluationZhengzhou University Zhengzhou Henan China
| | - Rui Wang
- Tianjin State Key Laboratory of Modern Chinese MedicineTianjin University of Traditional Chinese Medicine Tianjin China
| | - Song Wu
- Tianjin State Key Laboratory of Modern Chinese MedicineTianjin University of Traditional Chinese Medicine Tianjin China
| | - Hongwei Liu
- Tianjin State Key Laboratory of Modern Chinese MedicineTianjin University of Traditional Chinese Medicine Tianjin China
| | - Sangho Koo
- Department of ChemistryMyongji University Seoul South Korea
| | - Lifeng Han
- Tianjin State Key Laboratory of Modern Chinese MedicineTianjin University of Traditional Chinese Medicine Tianjin China
| | - Yi Zhang
- Tianjin State Key Laboratory of Modern Chinese MedicineTianjin University of Traditional Chinese Medicine Tianjin China
| | - Xiumei Gao
- Tianjin State Key Laboratory of Modern Chinese MedicineTianjin University of Traditional Chinese Medicine Tianjin China
| | - Xu Pang
- Tianjin State Key Laboratory of Modern Chinese MedicineTianjin University of Traditional Chinese Medicine Tianjin China
| | - Tao Wang
- Tianjin State Key Laboratory of Modern Chinese MedicineTianjin University of Traditional Chinese Medicine Tianjin China
| | - Haiyang Yu
- Tianjin State Key Laboratory of Modern Chinese MedicineTianjin University of Traditional Chinese Medicine Tianjin China
| |
Collapse
|
25
|
Lyu X, Song AL, Bai YL, Xu XD, He DQ, Zhang YC. Inhibitory effects of petasin on human colon carcinoma cells mediated by inactivation of Akt/mTOR pathway. Chin Med J (Engl) 2019; 132:1071-1078. [PMID: 30896562 PMCID: PMC6595872 DOI: 10.1097/cm9.0000000000000199] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Colorectal cancer is the third most common cancer worldwide and still lack of effective therapy so far. Petasin, a natural product found in plants of the genus Petasites, has been reported to possess anticancer activity. The present study aimed to investigate the anticolon cancer activity of petasin both in vitro and in vivo. The molecular mechanism of petasin was also further explored. METHODS Caco-2, LoVo, SW-620, and HT-29 cell lines were used to detect the inhibitory effect of petasin on colon cancer proliferation. Cell viability was determined using the MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide) assay. Cell apoptosis was analyzed by flow cytometry. Hoechst 33258 staining was used to visualize morphological changes. Cell migration was assessed using a wound-healing migration assay, and cell invasion was investigated using Transwell chambers. Western blotting assays were employed to evaluate the expression levels of proteins in the protein kinase B/mammalian target of rapamycin (Akt/mTOR) signaling pathway. Finally, in vivo activity of petasin was evaluated using the SW-620 subcutaneous tumor model established in Balb/c nude mice. Twelve rats were randomly divided into control group and 10 mg/kg petasin group. The tumor volume was calculated every 7 days for 28 days. Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay was performed to assess the apoptotic effect of petasin. Differences between two groups were assessed by analysis of independent-sample t tests. RESULTS Petasin significantly inhibited the proliferation of human colon carcinoma cell lines, induced apoptosis, and suppressed migration and invasion in SW-620 cells. Western blotting results showed that petasin decreased the phosphorylation of Akt (1.01 ± 0.16 vs. 0.74 ± 0.06, P = 0.042), mTOR (0.71 ± 0.12 vs. 0.32 ± 0.11, P = 0.013), and P70S6K (1.23 ± 0.21 vs. 0.85 ± 0.14, P = 0.008), elevated the expression of caspase-3 (0.41 ± 0.09 vs. 0.74 ± 0.12, P = 0.018) and caspase-9 (1.10 ± 0.27 vs. 1.98 ± 0.22, P = 0.009), decreased the Bcl-2 protein (2.75 ± 0.47 vs. 1.51 ± 0.36, P = 0.008), downregulated the expression of matrix metalloproteinase (MMP)-3 (1.51 ± 0.31 vs. 0.82 ± 0.11, P = 0.021) and MMP-9 (1.56 ± 0.32 vs. 0.94 ± 0.15, P = 0.039) in SW-620 cell. In vivo, 10 mg/kg petasin inhibited tumor growth in Balb/c nude mice (924.18 ± 101.23 vs. 577.67 ± 75.12 mm at day 28, P = 0.001) and induced apoptosis (3.6 ± 0.7% vs. 36.0 ± 4.9%, P = 0.001) in tumor tissues. CONCLUSIONS Petasin inhibits the proliferation of colon cancer SW-620 cells via inactivating the Akt/mTOR pathway. Our findings suggest petasin as a potential candidate for colon cancer therapy.
Collapse
Affiliation(s)
- Xi Lyu
- The 5th Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China
| | - Ai-Lin Song
- The 5th Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China
| | - Yin-Liang Bai
- Department of Pharmacy, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China
| | - Xiao-Dong Xu
- The 2nd Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China
| | - Dong-Qiang He
- The 5th Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China
| | - You-Cheng Zhang
- The 2nd Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China
| |
Collapse
|
26
|
Abstract
Ferroptosis is a nonapoptotic, iron-dependent form of cell death that can be activated in cancer cells by natural stimuli and synthetic agents. Three essential hallmarks define ferroptosis, namely: the loss of lipid peroxide repair capacity by the phospholipid hydroperoxidase GPX4, the availability of redox-active iron, and oxidation of polyunsaturated fatty acid (PUFA)-containing phospholipids. Several processes including RAS/MAPK signaling, amino acid and iron metabolism, ferritinophagy, epithelial-to-mesenchymal transition, cell adhesion, and mevalonate and phospholipid biosynthesis can modulate susceptibility to ferroptosis. Ferroptosis sensitivity is also governed by p53 and KEAP1/NRF2 activity, linking ferroptosis to the function of key tumor suppressor pathways. Together these findings highlight the role of ferroptosis as an emerging concept in cancer biology and an attractive target for precision cancer medicine discovery.
Collapse
Affiliation(s)
- Scott J. Dixon
- Department of Biology, Stanford University, Stanford, California 94305, USA
| | - Brent R. Stockwell
- Department of Biological Sciences and Department of Chemistry, Columbia University, New York, NY 10027, USA
| |
Collapse
|
27
|
Li Z, Mbah NE, Overmeyer JH, Sarver JG, George S, Trabbic CJ, Erhardt PW, Maltese WA. The JNK signaling pathway plays a key role in methuosis (non-apoptotic cell death) induced by MOMIPP in glioblastoma. BMC Cancer 2019; 19:77. [PMID: 30651087 PMCID: PMC6335761 DOI: 10.1186/s12885-019-5288-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 01/07/2019] [Indexed: 12/12/2022] Open
Abstract
Background Synthetic indolyl- pyridinyl- propenones (IPPs) induce methuosis, a form of non-apoptotic cell death, in glioblastoma and other cancer cell lines. Methuosis is characterized by accumulation of cytoplasmic vacuoles derived from macropinosomes and late endosomes, followed by metabolic failure and rupture of the plasma membrane. However, not all IPPs that cause vacuolization are cytotoxic. The main goals of the present study were to identify key signaling pathways that contribute to methuosis induced by cytotoxic IPPs and to evaluate the anti-tumor potential of a prototype IPP in vivo. Methods We utilized metabolic flux analysis, glucose uptake, immunoblotting, and selective pharmacological inhibitors to compare the effects of closely related cytotoxic and non-cytotoxic IPPs in cultured glioblastoma cells. To determine whether the use of methuosis-inducing IPPs might be feasible in a therapeutic context, we quantified the distribution of our lead IPP compound, MOMIPP, in mouse plasma and brain, and tested its ability to inhibit tumor growth in an intracerebral glioblastoma xenograft model. Results The cytotoxic IPP compound, MOMIPP, causes early disruptions of glucose uptake and glycolytic metabolism. Coincident with these metabolic changes, MOMIPP selectively activates the JNK1/2 stress kinase pathway, resulting in phosphorylation of c-Jun, Bcl-2 and Bcl-xL. At the same concentration, the non-cytotoxic analog, MOPIPP, does not activate these pathways. Pharmacologic inhibition of JNK activity promotes survival, even when cells are extensively vacuolated, but suppression of c-Jun transcriptional activity offers no protection. MOMIPP readily penetrates the blood-brain barrier and is moderately effective in suppressing progression of intracerebral glioblastoma xenografts. Conclusions The results suggest that interference with glucose uptake and induction of JNK-mediated phosphorylation of pro-survival members of the Bcl-2 family represent key events in the methuosis death process. In addition to providing new insights into the underlying molecular mechanism of methuosis, the results indicate that compounds of the cytotoxic IPP class may have potential for further development as therapeutic agents for brain tumors. Electronic supplementary material The online version of this article (10.1186/s12885-019-5288-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zehui Li
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio, 43614, United States
| | - Nneka E Mbah
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio, 43614, United States
| | - Jean H Overmeyer
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio, 43614, United States
| | - Jeffrey G Sarver
- Center for Drug Design and Development, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, 43606, USA
| | - Sage George
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio, 43614, United States
| | - Christopher J Trabbic
- Center for Drug Design and Development, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, 43606, USA
| | - Paul W Erhardt
- Center for Drug Design and Development, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, 43606, USA
| | - William A Maltese
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio, 43614, United States.
| |
Collapse
|
28
|
Induction of apoptosis by in vitro and in vivo plant extracts derived from Menyanthes trifoliata L. in human cancer cells. Cytotechnology 2019; 71:165-180. [PMID: 30610508 PMCID: PMC6368494 DOI: 10.1007/s10616-018-0274-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 10/25/2018] [Indexed: 12/14/2022] Open
Abstract
Menyanthes trifoliata L. has been used in traditional medicine for centuries. It exists in Asia, Europe, North America and in Morocco and is exploited as a remedy for anemia and lack of appetite. This plant shows many pharmacological properties, but its most interesting one is its anti-cancer potential. The present study examines the induction of apoptosis in grade IV glioma cells after treatment with the extracts from aerial part and root of M. trifoliata plants derived from in vitro (MtAPV and MtRV, respectively) and from soil (MtAPS and MtRS, respectively) and presents the first comparison of the biological effects of four different extracts of M. trifoliata against glioblastoma cells. The root extracts of M. trifoliata plants were found to exhibit cytotoxic effects against grade IV glioma cells, but not normal human astrocytes. HPLC analysis demonstrated the presence of various polyphenolic compounds, including sinapinic acid, ferulic acid, syringic acid and vanilic acid. Higher amount of pentacyclic triterpene (betulinic acid) was also found in MtRV extract. The growth inhibition of human grade IV glioma cells mediated by MtRV extract appears to be associated with apoptosis and G2/M phase cell cycle arrest, and altered expression of the pro- and anti-apoptotic genes (Bax, Bcl-2, Cas-3 and TP53) and proteins (Bax, Bcl-2, Cas-3 and p53), as well as decreased mitochondrial membrane potential. Our results indicate that M. trifoliata gives promising results as an anti-cancer agent for human glioblastoma cell lines. However, further research is necessary in view of its therapeutic use.
Collapse
|
29
|
Jackson M, Marks L, May GHW, Wilson JB. The genetic basis of disease. Essays Biochem 2018; 62:643-723. [PMID: 30509934 PMCID: PMC6279436 DOI: 10.1042/ebc20170053] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 10/02/2018] [Accepted: 10/05/2018] [Indexed: 12/13/2022]
Abstract
Genetics plays a role, to a greater or lesser extent, in all diseases. Variations in our DNA and differences in how that DNA functions (alone or in combinations), alongside the environment (which encompasses lifestyle), contribute to disease processes. This review explores the genetic basis of human disease, including single gene disorders, chromosomal imbalances, epigenetics, cancer and complex disorders, and considers how our understanding and technological advances can be applied to provision of appropriate diagnosis, management and therapy for patients.
Collapse
Affiliation(s)
- Maria Jackson
- School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, U.K
| | - Leah Marks
- School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, U.K
| | - Gerhard H W May
- School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, U.K.
| | - Joanna B Wilson
- School of Life Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, U.K
| |
Collapse
|
30
|
Sondka Z, Bamford S, Cole CG, Ward SA, Dunham I, Forbes SA. The COSMIC Cancer Gene Census: describing genetic dysfunction across all human cancers. Nat Rev Cancer 2018; 18:696-705. [PMID: 30293088 PMCID: PMC6450507 DOI: 10.1038/s41568-018-0060-1] [Citation(s) in RCA: 978] [Impact Index Per Article: 139.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The Catalogue of Somatic Mutations in Cancer (COSMIC) Cancer Gene Census (CGC) is an expert-curated description of the genes driving human cancer that is used as a standard in cancer genetics across basic research, medical reporting and pharmaceutical development. After a major expansion and complete re-evaluation, the 2018 CGC describes in detail the effect of 719 cancer-driving genes. The recent expansion includes functional and mechanistic descriptions of how each gene contributes to disease generation in terms of the key cancer hallmarks and the impact of mutations on gene and protein function. These functional characteristics depict the extraordinary complexity of cancer biology and suggest multiple cancer-related functions for many genes, which are often highly tissue-dependent or tumour stage-dependent. The 2018 CGC encompasses a second tier, describing an expanding list of genes (currently 145) from more recent cancer studies that show supportive but less detailed indications of a role in cancer.
Collapse
Affiliation(s)
- Zbyslaw Sondka
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK.
- Open Targets, Wellcome Genome Campus, Hinxton, Cambridge, UK.
| | - Sally Bamford
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Charlotte G Cole
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Sari A Ward
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Ian Dunham
- Open Targets, Wellcome Genome Campus, Hinxton, Cambridge, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Simon A Forbes
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| |
Collapse
|
31
|
Elkady N, Sultan M, Elkhouly E. Evaluation of topoisomerase II, ki-67, and P53 expression in non-muscle-invasive urothelial carcinoma and their clinical significance. INDIAN J PATHOL MICR 2018; 61:526-531. [PMID: 30303142 DOI: 10.4103/ijpm.ijpm_588_17] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Background Transurethral resection of tumor is the main treatment of non-muscle-invasive urothelial carcinoma, but it is associated with high rate of recurrence and/or progression and this arouses the need for adjuvant therapy. Topoisomerase II (Top II), KI-67, and P53 are proliferation and cell cycle regulation markers that may predict tumor response to therapy. Aim This study aimed to assess Top II, KI-67, and P53 expression and their effect on clinical outcome and response to therapy of non-muscle-invasive urothelial carcinoma. Materials and Methods Fifty cases of non-muscle invasive urothelial carcinoma were collected; Top II, KI-67, and P53 expression was evaluated. Patients received treatment then tumor recurrence was correlated with the expression of previous markers. Results There was a significant association between high Top II score, P53, and KI-67 and high tumor grade (P = 0.0001, 0.001, and 0.0001), submucosal infiltration (P = 0.0001 and 0.01), and recurrence (P = 0.01, 0.001, and 0.001). Conclusion Top II, P53, and KI-67 may predict tumor response to therapy and the clinical outcome in non-muscle-invasive urothelial carcinoma.
Collapse
Affiliation(s)
- Noha Elkady
- Department of Pathology, Faculty of Medicine, Menoufia University, Shibin El-Kom, Egypt
| | - Mohamed Sultan
- Department of Urology, Faculty of Medicine, Menoufia University, Shibin El-Kom, Egypt
| | - Enas Elkhouly
- Department of Clinical Oncology, Faculty of Medicine, Menoufia University, Shibin El-Kom, Egypt
| |
Collapse
|
32
|
Lu Y, Li L, Lin Z, Li M, Hu X, Zhang Y, Peng M, Xia H, Han G. Enhancing Osteosarcoma Killing and CT Imaging Using Ultrahigh Drug Loading and NIR-Responsive Bismuth Sulfide@Mesoporous Silica Nanoparticles. Adv Healthc Mater 2018; 7:e1800602. [PMID: 30102469 PMCID: PMC6504251 DOI: 10.1002/adhm.201800602] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 07/14/2018] [Indexed: 01/13/2023]
Abstract
Despite its 5-year event-free survival rate increasing to 60-65% due to surgery and chemotherapy, osteosarcoma (OS) remains one of the most threatening malignant human tumors, especially in young patients. Therefore, a new approach that combines early diagnosis with efficient tumor eradication and bioimaging is urgently needed. Here, a new type of mesoporous silica-coated bismuth sulfide nanoparticles (Bi2 S3 @MSN NPs) is developed. The well distributed mesoporous pores and large surface areas hold great promise for drug protection and encapsulation (doxorubicin (DOX), 99.85%). Moreover, the high photothermal efficiency of Bi2 S3 @MSNs (36.62%) offers great possibility for cancer synergistic treatment and highly near-infrared-triggered drug release (even at an ultralow power density of 0.3 W cm-2 ). After covalently conjugated to arginine-glycine-aspartic acid (RGD) peptide [c(RGDyC)], the NPs exhibit a high specificity for osteosarcoma and finally accumulate in the tumor cells (tenfold more than peritumoral tissues) for computed tomography (CT) imaging and tumor ablation. Importantly, the synergistic photothermal therapy-chemotherapy of the RGD-Bi2 S3 @MSN/DOX significantly ablates the highly malignant OS. It is further proved that the superior combined killing effect is achieved by activating the mitochondrial apoptosis pathway. Hence, the smart RGD-Bi2 S3 @MSN/DOX theranostic platform is a promising candidate for future applications in CT monitoring and synergistic treatment of malignant tumors.
Collapse
Affiliation(s)
- Yao Lu
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, 253 Gongye Road, Guangzhou 510282, China
- Department of Orthopedics, Guangdong Key Lab of Orthopedic Technology and Implant Key Laboratory of Trauma & Tissue Repair of Tropical Area of PLA Guangzhou General Hospital of Guangzhou Military Command of PLA 111 Liuhua Road, Guangzhou, Guangdong 510010, China
| | - Lihua Li
- Department of Orthopedics, Guangdong Key Lab of Orthopedic Technology and Implant Key Laboratory of Trauma & Tissue Repair of Tropical Area of PLA Guangzhou General Hospital of Guangzhou Military Command of PLA 111 Liuhua Road, Guangzhou, Guangdong 510010, China
- China–Germany Research Center for Photonic Materials and Device the State Key Laboratory ofLuminescent Materials and Devices School of Materials Science and Engineering South China University of Technology 381 Wushan Road, Guangzhou 510641, China
| | - Zefeng Lin
- Department of Orthopedics, Guangdong Key Lab of Orthopedic Technology and Implant Key Laboratory of Trauma & Tissue Repair of Tropical Area of PLA Guangzhou General Hospital of Guangzhou Military Command of PLA 111 Liuhua Road, Guangzhou, Guangdong 510010, China
| | - Mei Li
- Department of Orthopedics, Guangdong Key Lab of Orthopedic Technology and Implant Key Laboratory of Trauma & Tissue Repair of Tropical Area of PLA Guangzhou General Hospital of Guangzhou Military Command of PLA 111 Liuhua Road, Guangzhou, Guangdong 510010, China
| | - Xiaoming Hu
- Department of Orthopedics, Guangdong Key Lab of Orthopedic Technology and Implant Key Laboratory of Trauma & Tissue Repair of Tropical Area of PLA Guangzhou General Hospital of Guangzhou Military Command of PLA 111 Liuhua Road, Guangzhou, Guangdong 510010, China
| | - Yu Zhang
- Department of Orthopedics, Guangdong Key Lab of Orthopedic Technology and Implant Key Laboratory of Trauma & Tissue Repair of Tropical Area of PLA Guangzhou General Hospital of Guangzhou Military Command of PLA 111 Liuhua Road, Guangzhou, Guangdong 510010, China
| | - Mingying Peng
- China–Germany Research Center for Photonic Materials and Device the State Key Laboratory ofLuminescent Materials and Devices School of Materials Science and Engineering South China University of Technology 381 Wushan Road, Guangzhou 510641, China
| | - Hong Xia
- Department of Orthopedics, Guangdong Key Lab of Orthopedic Technology and Implant Key Laboratory of Trauma & Tissue Repair of Tropical Area of PLA Guangzhou General Hospital of Guangzhou Military Command of PLA 111 Liuhua Road, Guangzhou, Guangdong 510010, China
| | - Gang Han
- Department of Biochemistry and Molecular Pharmacology University of Massachusetts Medical School Worcester, MA 01605, USA
| |
Collapse
|
33
|
Han B, Jiang P, Li Z, Yu Y, Huang T, Ye X, Li X. Coptisine-induced apoptosis in human colon cancer cells (HCT-116) is mediated by PI3K/Akt and mitochondrial-associated apoptotic pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 48:152-160. [PMID: 30195873 DOI: 10.1016/j.phymed.2017.12.027] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 11/16/2017] [Accepted: 12/25/2017] [Indexed: 06/08/2023]
Abstract
BACKGROUND Colorectal cancer is the third leading cause of cancer-related deaths in the word. Coptisine (COP), an isoquinoline alkaloid derived from Coptis chinensis Franch, possesses a wide variety of pharmacological effects. However, its anti-proliferative effect on colon cancer is not fully elucidated. In the present study, we aimed to ascertain whether COP inhibits HCT-116 cell growth and to further explore the molecular mechanism in vitro and in vivo. METHODS Cell viability was determined by MTT assay. Cell migration was detected using wound healing assay. Apoptosis, mitochondrial membrane potential (Δψm) and reactive oxygen species (ROS) was analysis via flow cytometry. Hoechst 33342 was used for morphology observation. The expression levels of proteins related to mitochondrial-mediated apoptotic pathway were detected by western blotting. In addition, the antitumor ability of COP was further measured in athymic nude mice. RESULTS COP significantly decreased cell viability and migration in HCT-116 cells. Flow cytometry and Hoechst 33342 analysis confirmed that COP suppressed cell proliferation by inducing apoptosis. COP decreased Δψm dose-dependently and induced intracellular ROS production time-dependently. Western blotting showed that COP activated mitochondrial-associated apoptosis by down-regulating Bcl-2, Bcl-XL, pro-caspase 3, XIAP level and up-regulating Bax, Bad, cytochrome c, Apaf-1, AIF and cleaved caspase-3 expression. In addition, COP also attenuated PI3K/Akt signaling pathway. In vivo study showed that 150 mg/kg COP significantly delayed the tumor development in BALB/c nude mice. Immunohistochemical analysis also confirmed the activated apoptosis in tumor tissue. CONCLUSION The results demonstrated that COP induces apoptosis in HCT-116 cells through PI3K/Akt and mitochondrial-associated apoptotic pathway. Our findings suggest that COP has potential to be a therapeutic candidate for colon cancer patients.
Collapse
Affiliation(s)
- Bing Han
- Chongqing productivity promotion center for the modernization of Chinese traditional medicine, School of Pharmaceutical Sciences, Southwest University, Chongqing, 400716, China
| | - Pu Jiang
- Chongqing productivity promotion center for the modernization of Chinese traditional medicine, School of Pharmaceutical Sciences, Southwest University, Chongqing, 400716, China
| | - Zhaoxing Li
- Chongqing productivity promotion center for the modernization of Chinese traditional medicine, School of Pharmaceutical Sciences, Southwest University, Chongqing, 400716, China
| | - Yang Yu
- Chongqing productivity promotion center for the modernization of Chinese traditional medicine, School of Pharmaceutical Sciences, Southwest University, Chongqing, 400716, China
| | - Tao Huang
- Chongqing productivity promotion center for the modernization of Chinese traditional medicine, School of Pharmaceutical Sciences, Southwest University, Chongqing, 400716, China
| | - Xiaoli Ye
- School of Life Sciences, Southwest University, Chongqing, 400715, China; Chongqing Engineering Research Center for Pharmaceutical Process and Quality Control, Southwest University, Chongqing, 400716, China
| | - Xuegang Li
- Chongqing productivity promotion center for the modernization of Chinese traditional medicine, School of Pharmaceutical Sciences, Southwest University, Chongqing, 400716, China; Chongqing Engineering Research Center for Pharmaceutical Process and Quality Control, Southwest University, Chongqing, 400716, China.
| |
Collapse
|
34
|
Hagenbuchner J, Lungkofler L, Kiechl-Kohlendorfer U, Viola G, Ferlin MG, Ausserlechner MJ, Obexer P. The tubulin inhibitor MG-2477 induces autophagy-regulated cell death, ROS accumulation and activation of FOXO3 in neuroblastoma. Oncotarget 2018; 8:32009-32026. [PMID: 28415610 PMCID: PMC5458265 DOI: 10.18632/oncotarget.16434] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 03/08/2017] [Indexed: 12/29/2022] Open
Abstract
Neuroblastoma is the most frequent extra-cranial solid tumor in children with still high mortality in stage M. Here we studied the tubulin-inhibitor MG-2477 as a possible therapeutic agent for neuroblastoma therapy and uncovered that MG-2477 induces death in neuroblastoma cells independent of PKB-activation status and stage. MG-2477 triggers within 30 minutes extensive autophagosome-formation that finally leads to cell death associated with mitotic catastrophe. Autophagy is critical for MG-2477-induced death and is regulated by the BH3-only protein PMAIP1/NOXA which sequesters the anti-apoptotic BCL2-protein BCLXL and thereby displaces and activates the autophagy-regulator BECN1/beclin1. Knockdown of NOXA or overexpression of its pro-survival binding partners MCL1 and BCLXL counteracts MG-2477-induced cell death. MG-2477 also rapidly induces the repression of the anti-apoptotic protein Survivin, which promotes autophagy and cell death. We further observed the accumulation of reactive oxygen species (ROS) that triggers autophagy induction suggesting a change of the PI3 kinase-III/BECN1 complex and activates the transcription factor FOXO3, which contributes to final cell death induction. The combined data suggest that MG-2477 induces a sequential process of ROS-accumulation, autophagy and FOXO3-activation that leads to cell death in neuroblastoma cells.
Collapse
Affiliation(s)
- Judith Hagenbuchner
- Department of Pediatrics II, Medical University Innsbruck, Innsbruck, Austria
| | | | | | - Giampietro Viola
- Department of Woman's and Child's Health, Oncohematology Laboratory University of Padova, Padova, Italy
| | - Maria Grazia Ferlin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | | | - Petra Obexer
- Department of Pediatrics II, Medical University Innsbruck, Innsbruck, Austria.,Tyrolean Cancer Research Institute, Innsbruck, Austria
| |
Collapse
|
35
|
Carmona-Gutierrez D, Bauer MA, Zimmermann A, Aguilera A, Austriaco N, Ayscough K, Balzan R, Bar-Nun S, Barrientos A, Belenky P, Blondel M, Braun RJ, Breitenbach M, Burhans WC, Büttner S, Cavalieri D, Chang M, Cooper KF, Côrte-Real M, Costa V, Cullin C, Dawes I, Dengjel J, Dickman MB, Eisenberg T, Fahrenkrog B, Fasel N, Fröhlich KU, Gargouri A, Giannattasio S, Goffrini P, Gourlay CW, Grant CM, Greenwood MT, Guaragnella N, Heger T, Heinisch J, Herker E, Herrmann JM, Hofer S, Jiménez-Ruiz A, Jungwirth H, Kainz K, Kontoyiannis DP, Ludovico P, Manon S, Martegani E, Mazzoni C, Megeney LA, Meisinger C, Nielsen J, Nyström T, Osiewacz HD, Outeiro TF, Park HO, Pendl T, Petranovic D, Picot S, Polčic P, Powers T, Ramsdale M, Rinnerthaler M, Rockenfeller P, Ruckenstuhl C, Schaffrath R, Segovia M, Severin FF, Sharon A, Sigrist SJ, Sommer-Ruck C, Sousa MJ, Thevelein JM, Thevissen K, Titorenko V, Toledano MB, Tuite M, Vögtle FN, Westermann B, Winderickx J, Wissing S, Wölfl S, Zhang ZJ, Zhao RY, Zhou B, Galluzzi L, Kroemer G, Madeo F. Guidelines and recommendations on yeast cell death nomenclature. MICROBIAL CELL (GRAZ, AUSTRIA) 2018; 5:4-31. [PMID: 29354647 PMCID: PMC5772036 DOI: 10.15698/mic2018.01.607] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 12/29/2017] [Indexed: 12/18/2022]
Abstract
Elucidating the biology of yeast in its full complexity has major implications for science, medicine and industry. One of the most critical processes determining yeast life and physiology is cel-lular demise. However, the investigation of yeast cell death is a relatively young field, and a widely accepted set of concepts and terms is still missing. Here, we propose unified criteria for the defi-nition of accidental, regulated, and programmed forms of cell death in yeast based on a series of morphological and biochemical criteria. Specifically, we provide consensus guidelines on the differ-ential definition of terms including apoptosis, regulated necrosis, and autophagic cell death, as we refer to additional cell death rou-tines that are relevant for the biology of (at least some species of) yeast. As this area of investigation advances rapidly, changes and extensions to this set of recommendations will be implemented in the years to come. Nonetheless, we strongly encourage the au-thors, reviewers and editors of scientific articles to adopt these collective standards in order to establish an accurate framework for yeast cell death research and, ultimately, to accelerate the pro-gress of this vibrant field of research.
Collapse
Affiliation(s)
| | - Maria Anna Bauer
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Andreas Zimmermann
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Andrés Aguilera
- Centro Andaluz de Biología, Molecular y Medicina Regenerativa-CABIMER, Universidad de Sevilla, Sevilla, Spain
| | | | - Kathryn Ayscough
- Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | - Rena Balzan
- Department of Physiology and Biochemistry, University of Malta, Msida, Malta
| | - Shoshana Bar-Nun
- Department of Biochemistry and Molecular Biology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Antonio Barrientos
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, USA
- Department of Neurology, University of Miami Miller School of Medi-cine, Miami, USA
| | - Peter Belenky
- Department of Molecular Microbiology and Immunology, Brown University, Providence, USA
| | - Marc Blondel
- Institut National de la Santé et de la Recherche Médicale UMR1078, Université de Bretagne Occidentale, Etablissement Français du Sang Bretagne, CHRU Brest, Hôpital Morvan, Laboratoire de Génétique Moléculaire, Brest, France
| | - Ralf J. Braun
- Institute of Cell Biology, University of Bayreuth, Bayreuth, Germany
| | | | - William C. Burhans
- Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Sabrina Büttner
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | | | - Michael Chang
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Katrina F. Cooper
- Dept. Molecular Biology, Graduate School of Biomedical Sciences, Rowan University, Stratford, USA
| | - Manuela Côrte-Real
- Center of Molecular and Environmental Biology, Department of Biology, University of Minho, Braga, Portugal
| | - Vítor Costa
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Departamento de Biologia Molecular, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | | | - Ian Dawes
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Jörn Dengjel
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Martin B. Dickman
- Institute for Plant Genomics and Biotechnology, Texas A&M University, Texas, USA
| | - Tobias Eisenberg
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Birthe Fahrenkrog
- Laboratory Biology of the Nucleus, Institute for Molecular Biology and Medicine, Université Libre de Bruxelles, Charleroi, Belgium
| | - Nicolas Fasel
- Department of Biochemistry, University of Lausanne, Lausanne, Switzerland
| | - Kai-Uwe Fröhlich
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Ali Gargouri
- Laboratoire de Biotechnologie Moléculaire des Eucaryotes, Center de Biotechnologie de Sfax, Sfax, Tunisia
| | - Sergio Giannattasio
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council, Bari, Italy
| | - Paola Goffrini
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Campbell W. Gourlay
- Kent Fungal Group, School of Biosciences, University of Kent, Canterbury, United Kingdom
| | - Chris M. Grant
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Michael T. Greenwood
- Department of Chemistry and Chemical Engineering, Royal Military College, Kingston, Ontario, Canada
| | - Nicoletta Guaragnella
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council, Bari, Italy
| | | | - Jürgen Heinisch
- Department of Biology and Chemistry, University of Osnabrück, Osnabrück, Germany
| | - Eva Herker
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | | | - Sebastian Hofer
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | | | - Helmut Jungwirth
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Katharina Kainz
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Dimitrios P. Kontoyiannis
- Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Paula Ludovico
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Minho, Portugal
- ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Stéphen Manon
- Institut de Biochimie et de Génétique Cellulaires, UMR5095, CNRS & Université de Bordeaux, Bordeaux, France
| | - Enzo Martegani
- Department of Biotechnolgy and Biosciences, University of Milano-Bicocca, Milano, Italy
| | - Cristina Mazzoni
- Instituto Pasteur-Fondazione Cenci Bolognetti - Department of Biology and Biotechnology "C. Darwin", La Sapienza University of Rome, Rome, Italy
| | - Lynn A. Megeney
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
- Department of Medicine, Division of Cardiology, University of Ottawa, Ottawa, Canada
| | - Chris Meisinger
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
- Novo Nordisk Foundation Center for Biosustainability, Chalmers University of Technology, Gothenburg, Sweden
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, DK2800 Lyngby, Denmark
| | - Thomas Nyström
- Institute for Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Heinz D. Osiewacz
- Institute for Molecular Biosciences, Goethe University, Frankfurt am Main, Germany
| | - Tiago F. Outeiro
- Department of Experimental Neurodegeneration, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
- Max Planck Institute for Experimental Medicine, Göttingen, Germany
- Institute of Neuroscience, The Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, United Kingdom
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Hay-Oak Park
- Department of Molecular Genetics, The Ohio State University, Columbus, OH, USA
| | - Tobias Pendl
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Dina Petranovic
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
- Novo Nordisk Foundation Center for Biosustainability, Chalmers University of Technology, Gothenburg, Sweden
| | - Stephane Picot
- Malaria Research Unit, SMITh, ICBMS, UMR 5246 CNRS-INSA-CPE-University Lyon, Lyon, France
- Institut of Parasitology and Medical Mycology, Hospices Civils de Lyon, Lyon, France
| | - Peter Polčic
- Department of Biochemistry, Faculty of Natural Sciences, Comenius University in Bratislava, Bratislava, Slovak Republic
| | - Ted Powers
- Department of Molecular and Cellular Biology, College of Biological Sciences, UC Davis, Davis, California, USA
| | - Mark Ramsdale
- Biosciences, University of Exeter, Exeter, United Kingdom
| | - Mark Rinnerthaler
- Department of Cell Biology and Physiology, Division of Genetics, University of Salzburg, Salzburg, Austria
| | - Patrick Rockenfeller
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- Kent Fungal Group, School of Biosciences, University of Kent, Canterbury, United Kingdom
| | | | - Raffael Schaffrath
- Institute of Biology, Division of Microbiology, University of Kassel, Kassel, Germany
| | - Maria Segovia
- Department of Ecology, Faculty of Sciences, University of Malaga, Malaga, Spain
| | - Fedor F. Severin
- A.N. Belozersky Institute of physico-chemical biology, Moscow State University, Moscow, Russia
| | - Amir Sharon
- School of Plant Sciences and Food Security, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Stephan J. Sigrist
- Institute for Biology/Genetics, Freie Universität Berlin, Berlin, Germany
| | - Cornelia Sommer-Ruck
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Maria João Sousa
- Center of Molecular and Environmental Biology, Department of Biology, University of Minho, Braga, Portugal
| | - Johan M. Thevelein
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, KU Leuven, Leuven, Belgium
- Center for Microbiology, VIB, Leuven-Heverlee, Belgium
| | - Karin Thevissen
- Centre of Microbial and Plant Genetics, KU Leuven, Leuven, Belgium
| | | | - Michel B. Toledano
- Institute for Integrative Biology of the Cell (I2BC), SBIGEM, CEA-Saclay, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Mick Tuite
- Kent Fungal Group, School of Biosciences, University of Kent, Canterbury, United Kingdom
| | - F.-Nora Vögtle
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Joris Winderickx
- Department of Biology, Functional Biology, KU Leuven, Leuven-Heverlee, Belgium
| | | | - Stefan Wölfl
- Institute of Pharmacy and Molecu-lar Biotechnology, Heidelberg University, Heidelberg, Germany
| | - Zhaojie J. Zhang
- Department of Zoology and Physiology, University of Wyoming, Laramie, USA
| | - Richard Y. Zhao
- Department of Pathology, University of Maryland School of Medicine, Baltimore, USA
| | - Bing Zhou
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Université Paris Descartes/Paris V, Paris, France
| | - Guido Kroemer
- Université Paris Descartes/Paris V, Paris, France
- Equipe 11 Labellisée Ligue Contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Cell Biology and Metabolomics Platforms, Gustave Roussy Comprehensive Cancer Center, Villejuif, France
- INSERM, U1138, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, Paris, France
- Institute, Department of Women’s and Children’s Health, Karolinska University Hospital, Stockholm, Sweden
| | - Frank Madeo
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| |
Collapse
|
36
|
Calmon MF, Bortolozo Oliveira A, Adum de Matos R, Stuqui B, Torrezan G, Polaquini C, Regasini L, Rahal P. Cytotoxicity and antitumoral activity by apoptosis induction of AC13: A brominated curcumin analogue. Pharmacogn Mag 2018. [DOI: 10.4103/pm.pm_272_17] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
37
|
Pereira KMA, Costa SFDS, Pereira NB, Diniz MG, Castro WH, Gomes CC, Gomez RS. DNA methylation profiles of 22 apoptosis-related genes in odontogenic keratocysts before and after marsupialization. Oral Surg Oral Med Oral Pathol Oral Radiol 2017; 124:483-489. [DOI: 10.1016/j.oooo.2017.07.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Revised: 07/13/2017] [Accepted: 07/26/2017] [Indexed: 01/30/2023]
|
38
|
Loss of BIM augments resistance of ATM-deficient thymocytes to DNA damage-induced apoptosis but does not accelerate lymphoma development. Cell Death Differ 2017; 24:1987-1988. [PMID: 28885618 DOI: 10.1038/cdd.2017.138] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
39
|
A Small-Molecule Inhibitor of Bax and Bak Oligomerization Prevents Genotoxic Cell Death and Promotes Neuroprotection. Cell Chem Biol 2017; 24:493-506.e5. [PMID: 28392146 DOI: 10.1016/j.chembiol.2017.03.011] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 12/29/2016] [Accepted: 03/13/2017] [Indexed: 11/24/2022]
Abstract
Aberrant apoptosis can lead to acute or chronic degenerative diseases. Mitochondrial outer membrane permeabilization (MOMP) triggered by the oligomerization of the Bcl-2 family proteins Bax/Bak is an irreversible step leading to execution of apoptosis. Here, we describe the discovery of small-molecule inhibitors of Bax/Bak oligomerization that prevent MOMP. We demonstrate that these molecules disrupt multiple, but not all, interactions between Bax dimer interfaces thereby interfering with the formation of higher-order oligomers in the MOM, but not recruitment of Bax to the MOM. Small-molecule inhibition of Bax/Bak oligomerization allowed cells to evade apoptotic stimuli and rescued neurons from death after excitotoxicity, demonstrating that oligomerization of Bax is essential for MOMP. Our discovery of small-molecule Bax/Bak inhibitors provides novel tools for the investigation of the mechanisms leading to MOMP and will ultimately facilitate development of compounds inhibiting Bax/Bak in acute and chronic degenerative diseases.
Collapse
|
40
|
Bernard MM, McConnery JR, Hoskin DW. [10]-Gingerol, a major phenolic constituent of ginger root, induces cell cycle arrest and apoptosis in triple-negative breast cancer cells. Exp Mol Pathol 2017; 102:370-376. [PMID: 28315687 DOI: 10.1016/j.yexmp.2017.03.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 03/15/2017] [Indexed: 11/30/2022]
Abstract
The ginger rhizome is rich in bioactive compounds, including [6]-gingerol, [8]-gingerol, and [10]-gingerol; however, to date, most research on the anti-cancer activities of gingerols have focused on [6]-gingerol. In this study, we compared [10]-gingerol with [8]-gingerol and [6]-gingerol in terms of their ability to inhibit the growth of human and mouse mammary carcinoma cells. A colorimetric assay based on the enzymatic reduction of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide revealed that [10]-gingerol was more potent than [6]-gingerol and at least as potent as [8]-gingerol for the inhibition of triple-negative human (MDA-MB-231, MDA-MB-468) and mouse (4T1, E0771) mammary carcinoma cell growth. Further investigation of [10]-gingerol showed that it suppressed the growth of estrogen receptor-bearing (MCF-7, T47D) and HER2-overexpressing (SKBR3) breast cancer cells. The inhibitory effect of [10]-gingerol on the growth of MDA-MB-231 cells was associated with a reduction in the number of rounds of cell division and evidence of S phase-cell cycle arrest, as well as induction of apoptosis due to mitochondrial outer membrane permeabilization and the release of proapoptotic mitochondrial cytochrome c and SMAC/DIABLO into the cytoplasm. Surprisingly, killing of MDA-MB-231 cells by [10]-gingerol was not affected by a pan-caspase inhibitor (zVAD-fmk) or an anti-oxidant (N-acetylcysteine), suggesting that the cytotoxic effect of [10]-gingerol did not require caspase activation or the accumulation of reactive oxygen species. These findings suggest that further investigation of [10]-gingerol is warranted for its possible use in the treatment of breast cancer.
Collapse
Affiliation(s)
- Megan M Bernard
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Jason R McConnery
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - David W Hoskin
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada; Department of Pathology, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada; Department of Surgery, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada.
| |
Collapse
|
41
|
Huang T, Xiao Y, Yi L, Li L, Wang M, Tian C, Ma H, He K, Wang Y, Han B, Ye X, Li X. Coptisine from Rhizoma Coptidis Suppresses HCT-116 Cells-related Tumor Growth in vitro and in vivo. Sci Rep 2017; 7:38524. [PMID: 28165459 PMCID: PMC5292956 DOI: 10.1038/srep38524] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 11/11/2016] [Indexed: 12/21/2022] Open
Abstract
Colorectal cancer is one of the most common causes of cancer-related death in humans. Coptisine (COP) is a natural alkaloid from Coptidis Rhizoma with unclear antitumor mechanism. Human colon cancer cells (HCT-116) and xenograft mice were used to systematically explore the anti-tumor activity of COP in this study. The results indicated that COP exhibited remarkably cytotoxic activities against the HCT-116 cells by inducing G1-phase cell cycle arrest and increasing apoptosis, and preferentially inhibited the survival pathway and induced the activation of caspase proteases family of HCT-116 cells. Experimental results on male BALB/c nude mice confirmed that orally administration of COP at high-dose (150 mg/kg) could suppress tumor growth, and may reduce cancer metastasis risk by inhibiting the RAS-ERK pathway in vivo. Taken together, the results suggested that COP may be potential as a novel anti-tumor candidate in the HCT-116 cells-related colon cancer, further studies are still needed to suggest COP for the further use.
Collapse
Affiliation(s)
- Tao Huang
- School of Chinese Traditional Medicine, School of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Yubo Xiao
- School of Chinese Traditional Medicine, School of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
- Department of Clinical Laboratory, Hunan University of Medicine, Hunan 418000, China
| | - Lin Yi
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing Cancer Institute & Hospital & Cancer Center, Chongqing 400030, China
| | - Ling Li
- School of Chinese Traditional Medicine, School of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Meimei Wang
- School of Chinese Traditional Medicine, School of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Cheng Tian
- School of Chinese Traditional Medicine, School of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Hang Ma
- School of Chinese Traditional Medicine, School of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Kai He
- School of Chinese Traditional Medicine, School of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
- Department of Clinical Laboratory, Hunan University of Medicine, Hunan 418000, China
| | - Yue Wang
- School of Life Sciences, Southwest University, Chongqing 400715, China
| | - Bing Han
- School of Chinese Traditional Medicine, School of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Xiaoli Ye
- School of Life Sciences, Southwest University, Chongqing 400715, China
- Chongqing Engineering Research Center for Pharmaceutical Process and Quality Control, Chongqing 400716, China
| | - Xuegang Li
- School of Chinese Traditional Medicine, School of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
- Chongqing Engineering Research Center for Pharmaceutical Process and Quality Control, Chongqing 400716, China
| |
Collapse
|
42
|
Larsen BD, Sørensen CS. The caspase-activated DNase: apoptosis and beyond. FEBS J 2016; 284:1160-1170. [PMID: 27865056 DOI: 10.1111/febs.13970] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/04/2016] [Accepted: 11/17/2016] [Indexed: 01/13/2023]
Abstract
Organismal development and function requires multiple and accurate signal transduction pathways to ensure that proper balance between cell proliferation, differentiation, inactivation, and death is achieved. Cell death via apoptotic caspase signal transduction is extensively characterized and integral to this balance. Importantly, the view of apoptotic signal transduction has expanded over the previous decades. Subapoptotic caspase signaling has surfaced as mechanism that can promote the adoption of a range of cellular fates. An emerging mechanism of subapoptotic caspase signaling is the activation of the caspase-activated DNase (CAD) through controlled cleavage of the inhibitor of CAD (ICAD). CAD-induced DNA breaks incite a DNA damage response, frequently invoking p53 signaling, that transduces a change in cell fate. Cell differentiation and senescence are fates demonstrated to arise from CAD-induced DNA breaks. Furthermore, an apparent consequence of CAD activity is also emerging, as a potential source of oncogenic mutations. This review will discuss the mechanisms underlying CAD-induced DNA breaks and highlight how CAD activity promotes diverse cell fates.
Collapse
Affiliation(s)
- Brian D Larsen
- Biotech Research and Innovation Centre, University of Copenhagen, Denmark
| | - Claus S Sørensen
- Biotech Research and Innovation Centre, University of Copenhagen, Denmark
| |
Collapse
|
43
|
Delbridge ARD, Pang SHM, Vandenberg CJ, Grabow S, Aubrey BJ, Tai L, Herold MJ, Strasser A. RAG-induced DNA lesions activate proapoptotic BIM to suppress lymphomagenesis in p53-deficient mice. J Exp Med 2016; 213:2039-48. [PMID: 27621418 PMCID: PMC5030795 DOI: 10.1084/jem.20150477] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 08/08/2016] [Indexed: 01/29/2023] Open
Abstract
Delbridge, Strasser, and collaborators show that potentially oncogenic RAG1/2-dependent DNA lesions trigger apoptosis through the induction of BIM, which functions as an efficient tumor suppressor. Neoplastic transformation is driven by oncogenic lesions that facilitate unrestrained cell expansion and resistance to antiproliferative signals. These oncogenic DNA lesions, acquired through errors in DNA replication, gene recombination, or extrinsically imposed damage, are thought to activate multiple tumor suppressive pathways, particularly apoptotic cell death. DNA damage induces apoptosis through well-described p53-mediated induction of PUMA and NOXA. However, loss of both these mediators (even together with defects in p53-mediated induction of cell cycle arrest and cell senescence) does not recapitulate the tumor susceptibility observed in p53−/− mice. Thus, potentially oncogenic DNA lesions are likely to also trigger apoptosis through additional, p53-independent processes. We found that loss of the BH3-only protein BIM accelerated lymphoma development in p53-deficient mice. This process was negated by concomitant loss of RAG1/2-mediated antigen receptor gene rearrangement. This demonstrates that BIM is critical for the induction of apoptosis caused by potentially oncogenic DNA lesions elicited by RAG1/2-induced gene rearrangement. Furthermore, this highlights the role of a BIM-mediated tumor suppressor pathway that acts in parallel to the p53 pathway and remains active even in the absence of wild-type p53 function, suggesting this may be exploited in the treatment of p53-deficient cancers.
Collapse
Affiliation(s)
- Alex R D Delbridge
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Swee Heng Milon Pang
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Cassandra J Vandenberg
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Stephanie Grabow
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Brandon J Aubrey
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia Department of Clinical Haematology and Bone Marrow Transplant Service, the Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - Lin Tai
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Marco J Herold
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
44
|
Enhancing glioblastoma cell sensitivity to chemotherapeutics: A strategy involving survivin gene silencing mediated by gemini surfactant-based complexes. Eur J Pharm Biopharm 2016; 104:7-18. [DOI: 10.1016/j.ejpb.2016.04.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 03/23/2016] [Accepted: 04/18/2016] [Indexed: 11/23/2022]
|
45
|
Trabbic CJ, George SM, Alexander EM, Du S, Offenbacher JM, Crissman EJ, Overmeyer JH, Maltese WA, Erhardt PW. Synthesis and biological evaluation of isomeric methoxy substitutions on anti-cancer indolyl-pyridinyl-propenones: Effects on potency and mode of activity. Eur J Med Chem 2016; 122:79-91. [PMID: 27343855 DOI: 10.1016/j.ejmech.2016.06.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 06/07/2016] [Accepted: 06/10/2016] [Indexed: 12/20/2022]
Abstract
Certain indolyl-pyridinyl-propenone analogues kill glioblastoma cells that have become resistant to conventional therapeutic drugs. Some of these analogues induce a novel form of non-apoptotic cell death called methuosis, while others primarily cause microtubule disruption. Ready access to 5-indole substitution has allowed characterization of this position to be important for both types of mechanisms when a simple methoxy group is present. We now report the syntheses and biological effects of isomeric methoxy substitutions on the indole ring. Additionally, analogues containing a trimethoxyphenyl group in place of the pyridinyl moiety were evaluated for anticancer activity. The results demonstrate that the location of the methoxy group can alter both the potency and the mechanism of cell death. Remarkably, changing the methoxy from the 5-position to the 6-position switched the biological activity from induction of methuosis to disruption of microtubules. The latter may represent a prototype for a new class of mitotic inhibitors with potential therapeutic utility.
Collapse
Affiliation(s)
- Christopher J Trabbic
- Center for Drug Design and Development, Department of Medicinal and Biological Chemistry, University of Toledo College of Pharmacy and Pharmaceutical Sciences, 2801 W. Bancroft Ave., Toledo, OH 4360, USA
| | - Sage M George
- Department of Biochemistry and Cancer Biology, University of Toledo College of Medicine and Life Sciences, 3000 Arlington Ave., Toledo, OH 43614, USA
| | - Evan M Alexander
- Center for Drug Design and Development, Department of Medicinal and Biological Chemistry, University of Toledo College of Pharmacy and Pharmaceutical Sciences, 2801 W. Bancroft Ave., Toledo, OH 4360, USA
| | - Shengnan Du
- Department of Biochemistry and Cancer Biology, University of Toledo College of Medicine and Life Sciences, 3000 Arlington Ave., Toledo, OH 43614, USA
| | - Jennifer M Offenbacher
- Center for Drug Design and Development, Department of Medicinal and Biological Chemistry, University of Toledo College of Pharmacy and Pharmaceutical Sciences, 2801 W. Bancroft Ave., Toledo, OH 4360, USA
| | - Emily J Crissman
- Center for Drug Design and Development, Department of Medicinal and Biological Chemistry, University of Toledo College of Pharmacy and Pharmaceutical Sciences, 2801 W. Bancroft Ave., Toledo, OH 4360, USA
| | - Jean H Overmeyer
- Department of Biochemistry and Cancer Biology, University of Toledo College of Medicine and Life Sciences, 3000 Arlington Ave., Toledo, OH 43614, USA
| | - William A Maltese
- Department of Biochemistry and Cancer Biology, University of Toledo College of Medicine and Life Sciences, 3000 Arlington Ave., Toledo, OH 43614, USA.
| | - Paul W Erhardt
- Center for Drug Design and Development, Department of Medicinal and Biological Chemistry, University of Toledo College of Pharmacy and Pharmaceutical Sciences, 2801 W. Bancroft Ave., Toledo, OH 4360, USA.
| |
Collapse
|
46
|
Grabow S, Delbridge ARD, Aubrey BJ, Vandenberg CJ, Strasser A. Loss of a Single Mcl-1 Allele Inhibits MYC-Driven Lymphomagenesis by Sensitizing Pro-B Cells to Apoptosis. Cell Rep 2016; 14:2337-47. [PMID: 26947081 DOI: 10.1016/j.celrep.2016.02.039] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 12/22/2015] [Accepted: 02/03/2016] [Indexed: 01/19/2023] Open
Abstract
MCL-1 is critical for progenitor cell survival during emergency hematopoiesis, but its role in sustaining cells undergoing transformation and in lymphomagenesis is only poorly understood. We investigated the importance of MCL-1 in the survival of B lymphoid progenitors undergoing MYC-driven transformation and its functional interactions with pro-apoptotic BIM and PUMA and the tumor suppressor p53 in lymphoma development. Loss of one Mcl-1 allele almost abrogated MYC-driven-lymphoma development owing to a reduction in lymphoma initiating pre-B cells. Although loss of the p53 target PUMA had minor impact, loss of one p53 allele substantially accelerated lymphoma development when MCL-1 was limiting, most likely because p53 loss also causes defects in non-apoptotic tumor suppressive processes. Remarkably, loss of BIM restored the survival of lymphoma initiating cells and rate of tumor development. Thus, MCL-1 has a major role in lymphoma initiating pro-B cells to oppose BIM, which is upregulated in response to oncogenic stress.
Collapse
Affiliation(s)
- Stephanie Grabow
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC 3052, Australia; The Department of Medical Biology, University of Melbourne, Melbourne, VIC 3010, Australia.
| | - Alex R D Delbridge
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC 3052, Australia; The Department of Medical Biology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Brandon J Aubrey
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC 3052, Australia; The Department of Medical Biology, University of Melbourne, Melbourne, VIC 3010, Australia; Department of Clinical Haematology and Bone Marrow Transplant Service, the Royal Melbourne Hospital, Melbourne, VIC 3050, Australia
| | - Cassandra J Vandenberg
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC 3052, Australia; The Department of Medical Biology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC 3052, Australia; The Department of Medical Biology, University of Melbourne, Melbourne, VIC 3010, Australia.
| |
Collapse
|
47
|
Giampazolias E, Tait SWG. Mitochondria and the hallmarks of cancer. FEBS J 2015; 283:803-14. [PMID: 26607558 DOI: 10.1111/febs.13603] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 10/28/2015] [Accepted: 11/19/2015] [Indexed: 01/19/2023]
Abstract
Mitochondria have traditionally been viewed as the powerhouse of the cell, where they serve, amongst other functions, as a major source of ATP generation. More recently, mitochondria have also been shown to have active roles in a variety of other processes, including apoptotic cell death and inflammation. Here we review the various ways in which mitochondrial functions affect cancer. Although there are many diverse types of cancer, hallmarks have been defined that are applicable to most cancer types. We provide an overview of how mitochondrial functions affect some of these hallmarks, which include evasion of cell death, de-regulated bioenergetics, genome instability, tumour-promoting inflammation and metastasis. In addition to discussing the underlying mitochondrial roles in each of these processes, we also highlight the considerable potential of targeting mitochondrial functions to improve cancer treatment.
Collapse
Affiliation(s)
- Evangelos Giampazolias
- Cancer Research UK Beatson Institute, University of Glasgow, UK.,Institute of Cancer Sciences, University of Glasgow, UK
| | - Stephen W G Tait
- Cancer Research UK Beatson Institute, University of Glasgow, UK.,Institute of Cancer Sciences, University of Glasgow, UK
| |
Collapse
|
48
|
Pearson HB, McGlinn E, Phesse TJ, Schlüter H, Srikumar A, Gödde NJ, Woelwer CB, Ryan A, Phillips WA, Ernst M, Kaur P, Humbert P. The polarity protein Scrib mediates epidermal development and exerts a tumor suppressive function during skin carcinogenesis. Mol Cancer 2015; 14:169. [PMID: 26376988 PMCID: PMC4574215 DOI: 10.1186/s12943-015-0440-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 08/31/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The establishment and maintenance of polarity is vital for embryonic development and loss of polarity is a frequent characteristic of epithelial cancers, however the underlying molecular mechanisms remain unclear. Here, we identify a novel role for the polarity protein Scrib as a mediator of epidermal permeability barrier acquisition, skeletal morphogenesis, and as a potent tumor suppressor in cutaneous carcinogenesis. METHODS To explore the role of Scrib during epidermal development, we compared the permeability of toluidine blue dye in wild-type, Scrib heterozygous and Scrib KO embryonic epidermis at E16.5, E17.5 and E18.5. Mouse embryos were stained with alcian blue and alizarin red for skeletal analysis. To establish whether Scrib plays a tumor suppressive role during skin tumorigenesis and/or progression, we evaluated an autochthonous mouse model of skin carcinogenesis in the context of Scrib loss. We utilised Cre-LoxP technology to conditionally deplete Scrib in adult epidermis, since Scrib KO embryos are neonatal lethal. RESULTS We establish that Scrib perturbs keratinocyte maturation during embryonic development, causing impaired epidermal barrier formation, and that Scrib is required for skeletal morphogenesis in mice. Analysis of conditional transgenic mice deficient for Scrib specifically within the epidermis revealed no skin pathologies, indicating that Scrib is dispensable for normal adult epidermal homeostasis. Nevertheless, bi-allelic loss of Scrib significantly enhanced tumor multiplicity and progression in an autochthonous model of epidermal carcinogenesis in vivo, demonstrating Scrib is an epidermal tumor suppressor. Mechanistically, we show that apoptosis is the critical effector of Scrib tumor suppressor activity during skin carcinogenesis and provide new insight into the function of polarity proteins during DNA damage repair. CONCLUSIONS For the first time, we provide genetic evidence of a unique link between skin carcinogenesis and loss of the epithelial polarity regulator Scrib, emphasizing that Scrib exerts a wide-spread tumor suppressive function in epithelia.
Collapse
Affiliation(s)
- Helen B Pearson
- Peter MacCallum Cancer Centre, St Andrew's Place, East Melbourne, VIC, 3002, Australia. .,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia.
| | - Edwina McGlinn
- EMBL Australia, Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Toby J Phesse
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, 3052, Australia.,Present address: Olivia Newton-John Cancer Research Institute and School of Cancer Medicine at La Trobe University, Heidelberg, VIC, 3084, Australia
| | - Holger Schlüter
- Peter MacCallum Cancer Centre, St Andrew's Place, East Melbourne, VIC, 3002, Australia.,Present address: National Center for Tumor Diseases Heidelberg (NCT), German Cancer Research Centre (DKFZ), 69120, Heidelberg, Germany
| | - Anuratha Srikumar
- Peter MacCallum Cancer Centre, St Andrew's Place, East Melbourne, VIC, 3002, Australia
| | - Nathan J Gödde
- Peter MacCallum Cancer Centre, St Andrew's Place, East Melbourne, VIC, 3002, Australia
| | - Christina B Woelwer
- Peter MacCallum Cancer Centre, St Andrew's Place, East Melbourne, VIC, 3002, Australia
| | - Andrew Ryan
- TissuPath Laboratories, Mount Waverley, VIC, 3149, Australia
| | - Wayne A Phillips
- Peter MacCallum Cancer Centre, St Andrew's Place, East Melbourne, VIC, 3002, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia.,Department of Surgery (St. Vincent's Hospital), The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Matthias Ernst
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, 3052, Australia.,Present address: Olivia Newton-John Cancer Research Institute and School of Cancer Medicine at La Trobe University, Heidelberg, VIC, 3084, Australia
| | - Pritinder Kaur
- Peter MacCallum Cancer Centre, St Andrew's Place, East Melbourne, VIC, 3002, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Patrick Humbert
- Peter MacCallum Cancer Centre, St Andrew's Place, East Melbourne, VIC, 3002, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia.,Department of Pathology, The University of Melbourne, Parkville, VIC, 3010, Australia.,Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| |
Collapse
|
49
|
Aktipis CA, Boddy AM, Jansen G, Hibner U, Hochberg ME, Maley CC, Wilkinson GS. Cancer across the tree of life: cooperation and cheating in multicellularity. Philos Trans R Soc Lond B Biol Sci 2015; 370:20140219. [PMID: 26056363 PMCID: PMC4581024 DOI: 10.1098/rstb.2014.0219] [Citation(s) in RCA: 232] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2015] [Indexed: 02/06/2023] Open
Abstract
Multicellularity is characterized by cooperation among cells for the development, maintenance and reproduction of the multicellular organism. Cancer can be viewed as cheating within this cooperative multicellular system. Complex multicellularity, and the cooperation underlying it, has evolved independently multiple times. We review the existing literature on cancer and cancer-like phenomena across life, not only focusing on complex multicellularity but also reviewing cancer-like phenomena across the tree of life more broadly. We find that cancer is characterized by a breakdown of the central features of cooperation that characterize multicellularity, including cheating in proliferation inhibition, cell death, division of labour, resource allocation and extracellular environment maintenance (which we term the five foundations of multicellularity). Cheating on division of labour, exhibited by a lack of differentiation and disorganized cell masses, has been observed in all forms of multicellularity. This suggests that deregulation of differentiation is a fundamental and universal aspect of carcinogenesis that may be underappreciated in cancer biology. Understanding cancer as a breakdown of multicellular cooperation provides novel insights into cancer hallmarks and suggests a set of assays and biomarkers that can be applied across species and characterize the fundamental requirements for generating a cancer.
Collapse
Affiliation(s)
- C Athena Aktipis
- Center for Evolution and Cancer, University of California San Francisco, San Francisco, CA 94143, USA Department of Psychology, Arizona State University, Tempe, AZ 85287-4501, USA Centre for Evolution and Cancer, Institute for Cancer Research, 123 Old Brompton Road, London SW7 3RP, UK Institute for Advanced Study, Wissenschaftskolleg zu Berlin, Berlin, Germany
| | - Amy M Boddy
- Center for Evolution and Cancer, University of California San Francisco, San Francisco, CA 94143, USA Department of Psychology, Arizona State University, Tempe, AZ 85287-4501, USA Institute for Advanced Study, Wissenschaftskolleg zu Berlin, Berlin, Germany
| | - Gunther Jansen
- Department of Evolutionary Ecology and Genetics, University of Kiel, Am Botanischen Garten 1-9, 24118 Kiel, Germany Institute for Advanced Study, Wissenschaftskolleg zu Berlin, Berlin, Germany
| | - Urszula Hibner
- CNRS, UMR 5535, Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Montpellier, France Institute for Advanced Study, Wissenschaftskolleg zu Berlin, Berlin, Germany
| | - Michael E Hochberg
- Institut des Sciences de l'Evolution, CNRS UMR5554, Université Montpellier, 34095 Montpellier, France Santa Fe Institute, 1399 Hyde Park Road, Santa Fe, NM 87501, USA Institute for Advanced Study, Wissenschaftskolleg zu Berlin, Berlin, Germany
| | - Carlo C Maley
- Center for Evolution and Cancer, University of California San Francisco, San Francisco, CA 94143, USA Centre for Evolution and Cancer, Institute for Cancer Research, 123 Old Brompton Road, London SW7 3RP, UK Biodesign Institute, School of Life Sciences, Arizona State University, PO Box 8724501, Tempe, AZ 85287-4501, USA Institute for Advanced Study, Wissenschaftskolleg zu Berlin, Berlin, Germany
| | - Gerald S Wilkinson
- Department of Biology, University of Maryland, College Park, MD 20742, USA Institute for Advanced Study, Wissenschaftskolleg zu Berlin, Berlin, Germany
| |
Collapse
|
50
|
Narayanan KB, Ali M, Barclay BJ, Cheng Q(S, D’Abronzo L, Dornetshuber-Fleiss R, Ghosh PM, Gonzalez Guzman MJ, Lee TJ, Leung PS, Li L, Luanpitpong S, Ratovitski E, Rojanasakul Y, Romano MF, Romano S, Sinha RK, Yedjou C, Al-Mulla F, Al-Temaimi R, Amedei A, Brown DG, Ryan EP, Colacci AM, Hamid RA, Mondello C, Raju J, Salem HK, Woodrick J, Scovassi A, Singh N, Vaccari M, Roy R, Forte S, Memeo L, Kim SY, Bisson WH, Lowe L, Park HH. Disruptive environmental chemicals and cellular mechanisms that confer resistance to cell death. Carcinogenesis 2015; 36 Suppl 1:S89-S110. [PMID: 26106145 PMCID: PMC4565614 DOI: 10.1093/carcin/bgv032] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Revised: 01/28/2015] [Accepted: 02/03/2015] [Indexed: 12/12/2022] Open
Abstract
Cell death is a process of dying within biological cells that are ceasing to function. This process is essential in regulating organism development, tissue homeostasis, and to eliminate cells in the body that are irreparably damaged. In general, dysfunction in normal cellular death is tightly linked to cancer progression. Specifically, the up-regulation of pro-survival factors, including oncogenic factors and antiapoptotic signaling pathways, and the down-regulation of pro-apoptotic factors, including tumor suppressive factors, confers resistance to cell death in tumor cells, which supports the emergence of a fully immortalized cellular phenotype. This review considers the potential relevance of ubiquitous environmental chemical exposures that have been shown to disrupt key pathways and mechanisms associated with this sort of dysfunction. Specifically, bisphenol A, chlorothalonil, dibutyl phthalate, dichlorvos, lindane, linuron, methoxychlor and oxyfluorfen are discussed as prototypical chemical disruptors; as their effects relate to resistance to cell death, as constituents within environmental mixtures and as potential contributors to environmental carcinogenesis.
Collapse
Affiliation(s)
- Kannan Badri Narayanan
- Department of Chemistry and Biochemistry, Yeungnam University, Gyeongsan 712-749, South Korea
- Sultan Zainal Abidin University, Malaysia
- Plant Biotechnologies Inc, St. Albert AB, Canada
- Computer Science Department, Southern Illinois University, Carbondale, IL 62901, USA
- Department of Urology, University of California Davis, Sacramento, CA 95817, USA
- Department of Pharmacology and Toxicology, University of Vienna, Austria
- University of Puerto Rico, Medical Sciences Campus, School of Public Health, Nutrition Program, San Juan Puerto Rico 00936-5067, USA
- Department of Anatomy, College of Medicine, Yeungnam University, Daegu, 705-717, South Korea
- School of Biomedical Science, The Chinese University Of Hong Kong, Hong Kong, China
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
- Department of Otolaryngology/Head and Neck Surgery, Head and Neck Cancer Research Division, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Pharmaceutical Sciences, Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV 26506, USA
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, 80131 Naples, Italy
- Department of Molecular and Experimental Medicine, MEM 180, The Scripps Research Institute, La Jolla, CA 92037, USA
- Department of Biology, Jackson State University, Jackson, MS 39217, USA
- Department of Pathology, Kuwait University, Safat 13110, Kuwait
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze, 50134, Italy
- Department of Environmental and Radiological Health Sciences, Colorado state University/ Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna, 40126, Italy
- Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor 43400, Malaysia
- Institute of Molecular Genetics, National Research Council, Pavia, 27100, Italy
- Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario, K1A0K9, Canada
- Urology Department, Kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo, 12515, Egypt
- Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, 20057, USA
- Advenced Molecular Science Research Centre, King George’s Medical University, Lucknow, Uttar Pradesh, 226003, India
- Mediterranean Institute of Oncology, Viagrande, 95029, Italy
- Department of Internal Medicine, Korea Cancer Center Hospital, Seoul 139-706, South Korea
- Environmental and Molecular Toxicology, Environmental Health Science Center, Oregon State University, Corvallis, OR 97331, USA and
- Getting to Know Cancer, Truro, Nova Scotia, Canada
| | - Manaf Ali
- Sultan Zainal Abidin University, Malaysia
| | | | - Qiang (Shawn) Cheng
- Computer Science Department, Southern Illinois University, Carbondale, IL 62901, USA
| | - Leandro D’Abronzo
- Department of Urology, University of California Davis, Sacramento, CA 95817, USA
| | | | - Paramita M. Ghosh
- Department of Urology, University of California Davis, Sacramento, CA 95817, USA
| | - Michael J. Gonzalez Guzman
- University of Puerto Rico, Medical Sciences Campus, School of Public Health, Nutrition Program, San Juan Puerto Rico 00936-5067, USA
| | - Tae-Jin Lee
- Department of Anatomy, College of Medicine, Yeungnam University, Daegu, 705-717, South Korea
| | - Po Sing Leung
- School of Biomedical Science, The Chinese University Of Hong Kong, Hong Kong, China
| | - Lin Li
- School of Biomedical Science, The Chinese University Of Hong Kong, Hong Kong, China
| | - Suidjit Luanpitpong
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Edward Ratovitski
- Department of Otolaryngology/Head and Neck Surgery, Head and Neck Cancer Research Division, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Yon Rojanasakul
- Department of Pharmaceutical Sciences, Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV 26506, USA
| | - Maria Fiammetta Romano
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, 80131 Naples, Italy
| | - Simona Romano
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, 80131 Naples, Italy
| | - Ranjeet K. Sinha
- Department of Molecular and Experimental Medicine, MEM 180, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Clement Yedjou
- Department of Biology, Jackson State University, Jackson, MS 39217, USA
| | - Fahd Al-Mulla
- Department of Pathology, Kuwait University, Safat 13110, Kuwait
| | | | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Firenze, Firenze, 50134, Italy
| | - Dustin G. Brown
- Department of Environmental and Radiological Health Sciences, Colorado state University/ Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Elizabeth P. Ryan
- Department of Environmental and Radiological Health Sciences, Colorado state University/ Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Anna Maria Colacci
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna, 40126, Italy
| | - Roslida A. Hamid
- Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor 43400, Malaysia
| | - Chiara Mondello
- Institute of Molecular Genetics, National Research Council, Pavia, 27100, Italy
| | - Jayadev Raju
- Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario, K1A0K9, Canada
| | - Hosni K. Salem
- Urology Department, Kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo, 12515, Egypt
| | - Jordan Woodrick
- Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, 20057, USA
| | - A.Ivana Scovassi
- Institute of Molecular Genetics, National Research Council, Pavia, 27100, Italy
| | - Neetu Singh
- Advenced Molecular Science Research Centre, King George’s Medical University, Lucknow, Uttar Pradesh, 226003, India
| | - Monica Vaccari
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna, 40126, Italy
| | - Rabindra Roy
- Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC, 20057, USA
| | - Stefano Forte
- Mediterranean Institute of Oncology, Viagrande, 95029, Italy
| | - Lorenzo Memeo
- Mediterranean Institute of Oncology, Viagrande, 95029, Italy
| | - Seo Yun Kim
- Department of Internal Medicine, Korea Cancer Center Hospital, Seoul 139-706, South Korea
| | - William H. Bisson
- Environmental and Molecular Toxicology, Environmental Health Science Center, Oregon State University, Corvallis, OR 97331, USA and
| | - Leroy Lowe
- Getting to Know Cancer, Truro, Nova Scotia, Canada
| | - Hyun Ho Park
- *To whom correspondence should be addressed. Tel: +82 53 810 3015; Fax: +82 53 810 4619;
| |
Collapse
|