1
|
López-Cánovas JL, Naranjo-Martínez B, Diaz-Ruiz A. Fasting in combination with the cocktail Sorafenib:Metformin blunts cellular plasticity and promotes liver cancer cell death via poly-metabolic exhaustion. Cell Oncol (Dordr) 2025; 48:161-182. [PMID: 38990489 PMCID: PMC11850423 DOI: 10.1007/s13402-024-00966-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2024] [Indexed: 07/12/2024] Open
Abstract
PURPOSE Dual-Interventions targeting glucose and oxidative metabolism are receiving increasing attention in cancer therapy. Sorafenib (S) and Metformin (M), two gold-standards in liver cancer, are known for their mitochondrial inhibitory capacity. Fasting, a glucose-limiting strategy, is also emerging as chemotherapy adjuvant. Herein, we explore the anti-carcinogenic response of nutrient restriction in combination with sorafenib:metformin (NR-S:M). RESULTS Our data demonstrates that, independently of liver cancer aggressiveness, fasting synergistically boosts the anti-proliferative effects of S:M co-treatment. Metabolic and Cellular plasticity was determined by the examination of mitochondrial and glycolytic activity, cell cycle modulation, activation of cellular apoptosis, and regulation of key signaling and metabolic enzymes. Under NR-S:M conditions, early apoptotic events and the pro-apoptotic Bcl-xS/Bcl-xL ratio were found increased. NR-S:M induced the highest retention in cellular SubG1 phase, consistent with the presence of DNA fragments from cellular apoptosis. Mitochondrial functionality, Mitochondrial ATP-linked respiration, Maximal respiration and Spare respiratory capacity, were all found blunted under NR-S:M conditions. Basal Glycolysis, Glycolytic reserve, and glycolytic capacity, together with the expression of glycogenic (PKM), gluconeogenic (PCK1 and G6PC3), and glycogenolytic enzymes (PYGL, PGM1, and G6PC3), were also negatively impacted by NR-S:M. Lastly, a TMT-proteomic approach corroborated the synchronization of liver cancer metabolic reprogramming with the activation of molecular pathways to drive a quiescent-like status of energetic-collapse and cellular death. CONCLUSION Altogether, we show that the energy-based polytherapy NR-S:M blunts cellular, metabolic and molecular plasticity of liver cancer. Notwithstanding the in vitro design of this study, it holds a promising therapeutic tool worthy of exploration for this tumor pathology.
Collapse
Affiliation(s)
- Juan L López-Cánovas
- Laboratory of Cellular and Molecular Gerontology, Precision Nutrition and Aging Program, Institute IMDEA Food (CEI UAM+CSIC), Crta. de Canto Blanco nº 8, Madrid, E-28049, Spain
| | - Beatriz Naranjo-Martínez
- Laboratory of Cellular and Molecular Gerontology, Precision Nutrition and Aging Program, Institute IMDEA Food (CEI UAM+CSIC), Crta. de Canto Blanco nº 8, Madrid, E-28049, Spain
| | - Alberto Diaz-Ruiz
- Laboratory of Cellular and Molecular Gerontology, Precision Nutrition and Aging Program, Institute IMDEA Food (CEI UAM+CSIC), Crta. de Canto Blanco nº 8, Madrid, E-28049, Spain.
- CIBER Pathophysiology of Obesity and Nutrition (CIBERobn), Córdoba, Spain.
| |
Collapse
|
2
|
Gadet R, Jabbour L, Nguyen TTM, Lohez O, Mikaelian I, Gonzalo P, Luyten T, Chalabi-Dchar M, Wierinckx A, Marcillat O, Bultynck G, Rimokh R, Popgeorgiev N, Gillet G. The endoplasmic reticulum pool of Bcl-xL prevents cell death through IP3R-dependent calcium release. Cell Death Discov 2024; 10:346. [PMID: 39090104 PMCID: PMC11294475 DOI: 10.1038/s41420-024-02112-1] [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: 04/12/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 08/04/2024] Open
Abstract
Apoptosis plays a role in cell homeostasis in both normal development and disease. Bcl-xL, a member of the Bcl-2 family of proteins, regulates the intrinsic mitochondrial pathway of apoptosis. It is overexpressed in several cancers. Bcl-xL has a dual subcellular localisation and is found at the mitochondria as well as the endoplasmic reticulum (ER). However, the biological significance of its ER localisation is unclear. In order to decipher the functional contributions of the mitochondrial and reticular pools of Bcl-xL, we generated genetically modified mice expressing exclusively Bcl-xL at the ER, referred to as ER-xL, or the mitochondria, referred to as Mt-xL. By performing cell death assays, we demonstrated that ER-xL MEFs show increased vulnerability to apoptotic stimuli but are more resistant to ER stress. Furthermore, ER-xL MEFs displayed reduced 1,4,5-inositol trisphosphate receptor (IP3R)-mediated ER calcium release downstream of Phospholipase C activation. Collectively, our data indicate that upon ER stress, Bcl-xL negatively regulates IP3R-mediated calcium flux from the ER, which prevents ER calcium depletion and maintains the UPR and subsequent cell death in check. This work reveals a moonlighting function of Bcl-xL at the level of the ER, in addition to its well-known role in regulating apoptosis through the mitochondria.
Collapse
Affiliation(s)
- Rudy Gadet
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, 28 rue Laennec, 69008, Lyon, France
| | - Lea Jabbour
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, 28 rue Laennec, 69008, Lyon, France
| | - Trang Thi Minh Nguyen
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, 28 rue Laennec, 69008, Lyon, France
| | - Olivier Lohez
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, 28 rue Laennec, 69008, Lyon, France
| | - Ivan Mikaelian
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, 28 rue Laennec, 69008, Lyon, France
| | - Philippe Gonzalo
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, 28 rue Laennec, 69008, Lyon, France
- Laboratoire de Biochimie, CHU de Saint-Etienne, Université Jean Monnet, Saint-Étienne, France
| | - Tomas Luyten
- KU Leuven. Laboratory Molecular and Cellular Signaling. Department Cellular and Molecular Medicine., Campus Gasthuisberg O/N-I bus 802 Herestraat 49, BE-3000, Leuven, Belgium
| | - Mounira Chalabi-Dchar
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, 28 rue Laennec, 69008, Lyon, France
| | - Anne Wierinckx
- ProfilXpert. Faculté de Médecine Lyon Est, 8 rue Guillaume Paradin, 69008, Lyon, France
| | - Olivier Marcillat
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, 28 rue Laennec, 69008, Lyon, France
| | - Geert Bultynck
- KU Leuven. Laboratory Molecular and Cellular Signaling. Department Cellular and Molecular Medicine., Campus Gasthuisberg O/N-I bus 802 Herestraat 49, BE-3000, Leuven, Belgium
| | - Ruth Rimokh
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, 28 rue Laennec, 69008, Lyon, France
| | - Nikolay Popgeorgiev
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, 28 rue Laennec, 69008, Lyon, France
- Institut Universitaire de France, Paris, France
| | - Germain Gillet
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, 28 rue Laennec, 69008, Lyon, France.
- Hospices civils de Lyon, Laboratoire d'anatomie et cytologie pathologiques, Centre Hospitalier Lyon Sud, chemin du Grand Revoyet, 69495, Pierre Bénite, France.
| |
Collapse
|
3
|
Manjunath M, Ravindran F, Sharma S, Siddiqua H, Raghavan SC, Choudhary B. Disarib, a Specific BCL2 Inhibitor, Induces Apoptosis in Triple-Negative Breast Cancer Cells and Impedes Tumour Progression in Xenografts by Altering Mitochondria-Associated Processes. Int J Mol Sci 2024; 25:6485. [PMID: 38928195 PMCID: PMC11203414 DOI: 10.3390/ijms25126485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
Targeted cancer therapy aims to disrupt the functions of proteins that regulate cancer progression, mainly by using small molecule inhibitors (SMIs). SMIs exert their effect by modulating signalling pathways, organelle integrity, chromatin components, and several biosynthetic processes essential for cell division and survival. Antiapoptotic protein BCL2 is highly upregulated in many cancers compared with normal cells, making it an ideal target for cancer therapy. Around 75% of primary breast cancers overexpress BCL2, providing an opportunity to explore BCL2 inhibitors as a therapeutic option. Disarib is an SMI that has been developed as a selective BCL2 inhibitor. Disarib works by disrupting BCL2-BAK interaction and activating intrinsic apoptotic pathways in leukemic cells while sparing normal cells. We investigated the effects of Disarib, a BCL2 specific inhibitor, on breast cancer cells and xenografts. Cytotoxicity and fluorometric assays revealed that Disarib induced cell death by increasing reactive oxygen species and activating intrinsic apoptotic pathways in Triple-Negative Breast Cancer cells (MDA-MB-231 and MDA-MB-468). Disarib also affected the colony-forming properties of these cells. MDA-MB-231- and MDA-MB-468-derived xenografts showed a significant reduction in tumours upon Disarib treatment. Through the transcriptomics approach, we also explored the influence of BCL2 inhibitors on energy metabolism, mitochondrial dynamics, and epithelial-to-mesenchymal transition (EMT). Mitochondrial dynamics and glucose metabolism mainly regulate energy metabolism. The change in energetics regulates tumour growth through epithelial-mesenchymal transition, and angiogenesis. RNA sequencing (RNAseq) analysis revealed that BCL2 inhibitors ABT-199 and Disarib maintain Oxphos levels in MDA-MB-231. However, key glycolytic genes were significantly downregulated. Mitochondrial fission genes were seen to be downregulated both in RNAseq data and semi quantitative real time polymerase chain reaction (qRTPCR) in Disarib-treated TNBC cells and xenografts. Lastly, Disarib inhibited wound healing and epithelial-to-mesenchymal transition. This study showed that Disarib disrupts mitochondrial function, activates the intrinsic apoptotic pathway in breast cancer, and inhibits epithelial-to-mesenchymal transition both in vitro and in vivo. These findings highlight Disarib's potential as a multifaceted therapeutic strategy for patients with Triple-Negative Breast Cancer.
Collapse
Affiliation(s)
- Meghana Manjunath
- Department of Biotechnology and Applied Bioinformatics, Institute of Bioinformatics and Applied Biotechnology, Electronic City Phase 1, Bengaluru 560100, India
| | - Febina Ravindran
- Department of Biotechnology and Applied Bioinformatics, Institute of Bioinformatics and Applied Biotechnology, Electronic City Phase 1, Bengaluru 560100, India
| | - Shivangi Sharma
- Department of Biotechnology and Applied Bioinformatics, Institute of Bioinformatics and Applied Biotechnology, Electronic City Phase 1, Bengaluru 560100, India
- Indian Institute of Science, Bengaluru 560012, India; (H.S.); (S.C.R.)
| | - Humaira Siddiqua
- Indian Institute of Science, Bengaluru 560012, India; (H.S.); (S.C.R.)
| | | | - Bibha Choudhary
- Department of Biotechnology and Applied Bioinformatics, Institute of Bioinformatics and Applied Biotechnology, Electronic City Phase 1, Bengaluru 560100, India
| |
Collapse
|
4
|
Davidson BA, Miranda AX, Reed SC, Bergman RE, Kemp JDJ, Reddy AP, Pantone MV, Fox EK, Dorand RD, Hurley PJ, Croessmann S, Park BH. An in vitro CRISPR screen of cell-free DNA identifies apoptosis as the primary mediator of cell-free DNA release. Commun Biol 2024; 7:441. [PMID: 38600351 PMCID: PMC11006667 DOI: 10.1038/s42003-024-06129-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 03/29/2024] [Indexed: 04/12/2024] Open
Abstract
ABTRACT Clinical circulating cell-free DNA (cfDNA) testing is now routine, however test accuracy remains limited. By understanding the life-cycle of cfDNA, we might identify opportunities to increase test performance. Here, we profile cfDNA release across a 24-cell line panel and utilize a cell-free CRISPR screen (cfCRISPR) to identify mediators of cfDNA release. Our panel outlines two distinct groups of cell lines: one which releases cfDNA fragmented similarly to clinical samples and purported as characteristic of apoptosis, and another which releases larger fragments associated with vesicular or necrotic DNA. Our cfCRISPR screens reveal that genes mediating cfDNA release are primarily involved with apoptosis, but also identify other subsets of genes such as RNA binding proteins as potential regulators of cfDNA release. We observe that both groups of cells lines identified primarily produce cfDNA through apoptosis. These results establish the utility of cfCRISPR, genetically validate apoptosis as a major mediator of DNA release in vitro, and implicate ways to improve cfDNA assays.
Collapse
Affiliation(s)
- Brad A Davidson
- Division of Hematology, Oncology, Department of Medicine, Vanderbilt University Medical Center and the Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - Adam X Miranda
- Division of Hematology, Oncology, Department of Medicine, Vanderbilt University Medical Center and the Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - Sarah C Reed
- Division of Hematology, Oncology, Department of Medicine, Vanderbilt University Medical Center and the Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
- Medical Scientist Training Program, Vanderbilt University, Nashville, TN, USA
| | - Riley E Bergman
- Division of Hematology, Oncology, Department of Medicine, Vanderbilt University Medical Center and the Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
- Medical Scientist Training Program, Vanderbilt University, Nashville, TN, USA
| | - Justin D J Kemp
- Division of Hematology, Oncology, Department of Medicine, Vanderbilt University Medical Center and the Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - Anvith P Reddy
- Division of Hematology, Oncology, Department of Medicine, Vanderbilt University Medical Center and the Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
- Medical Scientist Training Program, Vanderbilt University, Nashville, TN, USA
| | - Morgan V Pantone
- Division of Hematology, Oncology, Department of Medicine, Vanderbilt University Medical Center and the Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - Ethan K Fox
- Division of Hematology, Oncology, Department of Medicine, Vanderbilt University Medical Center and the Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - R Dixon Dorand
- Division of Hematology, Oncology, Department of Medicine, Vanderbilt University Medical Center and the Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - Paula J Hurley
- Division of Hematology, Oncology, Department of Medicine, Vanderbilt University Medical Center and the Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - Sarah Croessmann
- Division of Hematology, Oncology, Department of Medicine, Vanderbilt University Medical Center and the Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - Ben Ho Park
- Division of Hematology, Oncology, Department of Medicine, Vanderbilt University Medical Center and the Vanderbilt-Ingram Cancer Center, Nashville, TN, USA.
| |
Collapse
|
5
|
Li X, Xu SJ, Jin B, Lu HS, Zhao SK, Ding XF, Xu LL, Li HJ, Liu SC, Chen J, Chen G. Heparanase inhibitor OGT 2115 induces prostate cancer cell apoptosis via the downregulation of MCL‑1. Oncol Lett 2024; 27:83. [PMID: 38249815 PMCID: PMC10797316 DOI: 10.3892/ol.2024.14217] [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: 07/28/2023] [Accepted: 10/31/2023] [Indexed: 01/23/2024] Open
Abstract
Heparanase (HPSE), an endo-β-D-glucuronidase, cleaves heparan sulfate and serves an important role in the tumor microenvironment and thus in tumorigenesis. HPSE is known to promote tumor cell evasion of apoptosis. However, the underlying mechanism of this requires further study. In the present study, the results demonstrated that myeloid cell leukemia-1 (MCL-1), an antiapoptotic protein, and HPSE were upregulated in prostate cancer tissues compared with adjacent normal tissues. In addition, the HPSE inhibitor, OGT 2115, inhibited PC-3 and DU-145 prostate cancer cell viability in a dose-dependent manner, with IC50 values of 20.2 and 97.2 µM, respectively. Furthermore, annexin V/PI double-staining assays demonstrated that OGT 2115 induced apoptosis in prostate cancer cells. OGT 2115 treatment markedly decreased MCL-1 protein expression levels, whereas RNA interference-mediated downregulation of MCL-1 and OGT 2115 drug treatment synergistically induced apoptosis in PC-3 and DU-145 cells. In vivo, OGT 2115 40 mg/kg (ig) significantly inhibited PC-3 cell xenograft growth in nude mice and increased the positive TUNEL staining rate of xenograft tissues. It was therefore hypothesized that MCL-1 was an important signaling molecule in OGT 2115-induced apoptosis. The results of the present study also demonstrated that the proteasome inhibitor, MG-132, markedly inhibited the downregulation of MCL-1 protein expression levels induced by OGT 2115. However, the protein synthesis inhibitor, cycloheximide, did not affect the role of OGT 2115 in regulating MCL-1. In summary, the results of the present study demonstrated that the proapoptotic activity of OGT 2115 was achieved by downregulating MCL-1.
Collapse
Affiliation(s)
- Xin Li
- Department of Urology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang 318000, P.R. China
| | - Shuai-Jun Xu
- Graduate School of Medicine, Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Bin Jin
- Graduate School of Medicine, Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Hong-Sheng Lu
- Department of Pathology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang 318000, P.R. China
| | - Shan-Kun Zhao
- Department of Urology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang 318000, P.R. China
| | - Xiao-Fei Ding
- Department of Pharmacology, Taizhou University, Taizhou, Zhejiang 318000, P.R. China
| | - Ling-Long Xu
- Department of Hematology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang 318000, P.R. China
| | - Hai-Jun Li
- Department of Neurology, Taizhou Second People's Hospital, Taizhou University, Taizhou, Zhejiang 318000, P.R. China
| | - Shuang-Chun Liu
- Laboratory Department, Municipal Hospital Affiliated to Taizhou University, Taizhou, Zhejiang 318000, P.R. China
| | - Jie Chen
- Department of Pharmacology, Taizhou University, Taizhou, Zhejiang 318000, P.R. China
| | - Guang Chen
- Department of Pharmacology, Taizhou University, Taizhou, Zhejiang 318000, P.R. China
- Department of Hematology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang 318000, P.R. China
| |
Collapse
|
6
|
Choi S, Cho N, Kim EM, Kim KK. The role of alternative pre-mRNA splicing in cancer progression. Cancer Cell Int 2023; 23:249. [PMID: 37875914 PMCID: PMC10594706 DOI: 10.1186/s12935-023-03094-3] [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: 09/06/2023] [Accepted: 10/06/2023] [Indexed: 10/26/2023] Open
Abstract
Alternative pre-mRNA splicing is a critical mechanism that generates multiple mRNA from a single gene, thereby increasing the diversity of the proteome. Recent research has highlighted the significance of specific splicing isoforms in cellular processes, particularly in regulating cell numbers. In this review, we examine the current understanding of the role of alternative splicing in controlling cancer cell growth and discuss specific splicing factors and isoforms and their molecular mechanisms in cancer progression. These isoforms have been found to intricately control signaling pathways crucial for cell cycle progression, proliferation, and apoptosis. Furthermore, studies have elucidated the characteristics and functional importance of splicing factors that influence cell numbers. Abnormal expression of oncogenic splicing isoforms and splicing factors, as well as disruptions in splicing caused by genetic mutations, have been implicated in the development and progression of tumors. Collectively, these findings provide valuable insights into the complex interplay between alternative splicing and cell proliferation, thereby suggesting the potential of alternative splicing as a therapeutic target for cancer.
Collapse
Affiliation(s)
- Sunkyung Choi
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Namjoon Cho
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Eun-Mi Kim
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea.
| | - Kee K Kim
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon, 34134, Republic of Korea.
| |
Collapse
|
7
|
Nakamura M, Keller MA, Fefelova N, Zhai P, Liu T, Tian Y, Ikeda S, Del Re DP, Li H, Xie LH, Sadoshima J. Ser14 phosphorylation of Bcl-xL mediates compensatory cardiac hypertrophy in male mice. Nat Commun 2023; 14:5805. [PMID: 37726310 PMCID: PMC10509265 DOI: 10.1038/s41467-023-41595-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 09/08/2023] [Indexed: 09/21/2023] Open
Abstract
The anti-apoptotic function of Bcl-xL in the heart during ischemia/reperfusion is diminished by K-Ras-Mst1-mediated phosphorylation of Ser14, which allows dissociation of Bcl-xL from Bax and promotes cardiomyocyte death. Here we show that Ser14 phosphorylation of Bcl-xL is also promoted by hemodynamic stress in the heart, through the H-Ras-ERK pathway. Bcl-xL Ser14 phosphorylation-resistant knock-in male mice develop less cardiac hypertrophy and exhibit contractile dysfunction and increased mortality during acute pressure overload. Bcl-xL Ser14 phosphorylation enhances the Ca2+ transient by blocking the inhibitory interaction between Bcl-xL and IP3Rs, thereby promoting Ca2+ release and activation of the calcineurin-NFAT pathway, a Ca2+-dependent mechanism that promotes cardiac hypertrophy. These results suggest that phosphorylation of Bcl-xL at Ser14 in response to acute pressure overload plays an essential role in mediating compensatory hypertrophy by inducing the release of Bcl-xL from IP3Rs, alleviating the negative constraint of Bcl-xL upon the IP3R-NFAT pathway.
Collapse
Affiliation(s)
- Michinari Nakamura
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, 185 South Orange Ave, Newark, NJ, 07103, USA.
| | - Mariko Aoyagi Keller
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, 185 South Orange Ave, Newark, NJ, 07103, USA
| | - Nadezhda Fefelova
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, 185 South Orange Ave, Newark, NJ, 07103, USA
| | - Peiyong Zhai
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, 185 South Orange Ave, Newark, NJ, 07103, USA
| | - Tong Liu
- Center for Advanced Proteomics Research, Department of Biochemistry & Molecular Biology, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Yimin Tian
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, 185 South Orange Ave, Newark, NJ, 07103, USA
| | - Shohei Ikeda
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, 185 South Orange Ave, Newark, NJ, 07103, USA
| | - Dominic P Del Re
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, 185 South Orange Ave, Newark, NJ, 07103, USA
| | - Hong Li
- Center for Advanced Proteomics Research, Department of Biochemistry & Molecular Biology, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Lai-Hua Xie
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, 185 South Orange Ave, Newark, NJ, 07103, USA
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers-New Jersey Medical School, 185 South Orange Ave, Newark, NJ, 07103, USA.
| |
Collapse
|
8
|
Stark MC, Joubert AM, Visagie MH. Molecular Farming of Pembrolizumab and Nivolumab. Int J Mol Sci 2023; 24:10045. [PMID: 37373192 DOI: 10.3390/ijms241210045] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/09/2023] [Accepted: 06/10/2023] [Indexed: 06/29/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) are a class of immunotherapy agents capable of alleviating the immunosuppressive effects exerted by tumorigenic cells. The programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) immune checkpoint is one of the most ubiquitous checkpoints utilized by tumorigenic cells for immune evasion by inducing apoptosis and inhibiting the proliferation and cytokine production of T lymphocytes. Currently, the most frequently used ICIs targeting the PD-1/PD-L1 checkpoint include monoclonal antibodies (mAbs) pembrolizumab and nivolumab that bind to PD-1 on T lymphocytes and inhibit interaction with PD-L1 on tumorigenic cells. However, pembrolizumab and nivolumab are costly, and thus their accessibility is limited in low- and middle-income countries (LMICs). Therefore, it is essential to develop novel biomanufacturing platforms capable of reducing the cost of these two therapies. Molecular farming is one such platform utilizing plants for mAb production, and it has been demonstrated to be a rapid, low-cost, and scalable platform that can be potentially implemented in LMICs to diminish the exorbitant prices, ultimately leading to a significant reduction in cancer-related mortalities within these countries.
Collapse
Affiliation(s)
- Michael C Stark
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Private Bag X323, Pretoria 0031, South Africa
| | - Anna M Joubert
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Private Bag X323, Pretoria 0031, South Africa
| | - Michelle H Visagie
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Private Bag X323, Pretoria 0031, South Africa
| |
Collapse
|
9
|
Qureshi QUA, Audas TE, Morin RD, Coyle KM. Emerging roles for heterogeneous ribonuclear proteins in normal and malignant B cells. Biochem Cell Biol 2023; 101:160-171. [PMID: 36745874 DOI: 10.1139/bcb-2022-0332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Heterogeneous nuclear ribonucleoproteins (hnRNPs) are among the most abundantly expressed RNA binding proteins in the cell and play major roles in all facets of RNA metabolism. hnRNPs are increasingly appreciated as essential for mammalian B cell development by regulating the carefully ordered expression of specific genes. Due to this tight regulation of the hnRNP-RNA network, it is no surprise that a growing number of genes encoding hnRNPs have been causally associated with the onset or progression of many cancers, including B cell neoplasms. Here we discuss our current understanding of hnRNP-driven regulation in normal, perturbed, and malignant B cells, and the most recent and emerging therapeutic innovations aimed at targeting the hnRNP-RNA network in lymphoma.
Collapse
Affiliation(s)
- Qurat Ul Ain Qureshi
- Department of Molecular Biology & Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Timothy E Audas
- Department of Molecular Biology & Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Ryan D Morin
- Department of Molecular Biology & Biochemistry, Simon Fraser University, Burnaby, BC, Canada.,Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Krysta M Coyle
- Department of Molecular Biology & Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| |
Collapse
|
10
|
Kwon JW, Seok SH, Kim S, An HW, Choudhury AD, Woo SH, Oh JS, Kim JK, Voon DC, Kim DY, Park JW. A synergistic partnership between IL-33/ST2 and Wnt pathway through Bcl-xL drives gastric cancer stemness and metastasis. Oncogene 2023; 42:501-515. [PMID: 36526851 DOI: 10.1038/s41388-022-02575-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 12/04/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022]
Abstract
ST2 functions as a receptor for the cytokine IL-33. It has been implicated in carcinogenesis. In this study, we sought to mechanistically determine how ST2 and IL-33 function to support cancer stem cell (CSC) activity and drive gastric cancer (GC) pathogenesis. ST2+ subpopulation spontaneously arose during gastric tumorigenesis. A thorough evaluation of ST2 and IL-33 expression in gastric tumors revealed that they show an overlapping expression pattern, notably in poor differentiated GC and metastasis foci. Moreover, their expression levels are clinically correlated to cancer progression. Using a genetic model of CSC-driven gastric carcinogenesis, ST2+ subpopulation displays increased tumorigenicity, chemoresistance and metastatic potentials through increased survival fitness endowed by an elevated MAPK-regulated Bcl-xL. The IL-33/ST2 axis enhances the self-renewal and survival of GC stem cells and organoids. Importantly, we observed a synergistic cooperation between IL-33/ST2 and the canonical Wnt pathway in transactivating Wnt-dependent transcription and supporting CSC activity, a partnership that was abrogated by inhibiting Bcl-xL. Concordant with this, ST2+ subpopulation was targeted by MEK1/2 and Bcl-xL-specific inhibitors. These findings establish ST2 as a functional CSC marker that fortifies the Wnt signal while availing a novel therapeutic strategy to suppress GC progression by targeting the IL-33/ST2/Bcl-xL signaling axis.
Collapse
Affiliation(s)
- Jong-Wan Kwon
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, 1 Kangwondaehak-gil, ChunCheon-si, Gangwon-do, 24341, South Korea
| | - Sang-Hyuk Seok
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, 1 Kangwondaehak-gil, ChunCheon-si, Gangwon-do, 24341, South Korea
| | - Somi Kim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, 37673, South Korea
| | - Hyeok-Won An
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, 1 Kangwondaehak-gil, ChunCheon-si, Gangwon-do, 24341, South Korea
| | - Anahita Dev Choudhury
- Cancer Research Institute, Kanazawa University, Kanazawa, 920-1192, Japan.,Innovative Cancer Model Research Unit, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Sang-Ho Woo
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea
| | - Jeong-Seop Oh
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea
| | - Jong Kyoung Kim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, 37673, South Korea
| | - Dominic C Voon
- Cancer Research Institute, Kanazawa University, Kanazawa, 920-1192, Japan. .,Innovative Cancer Model Research Unit, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, 920-1192, Japan.
| | - Dae-Yong Kim
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea.
| | - Jun Won Park
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, 1 Kangwondaehak-gil, ChunCheon-si, Gangwon-do, 24341, South Korea.
| |
Collapse
|
11
|
Raimondi V, Iannozzi NT, Burroughs-Garcìa J, Toscani D, Storti P, Giuliani N. A personalized molecular approach in multiple myeloma: the possible use of RAF/RAS/MEK/ERK and BCL-2 inhibitors. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2022; 3:463-479. [PMID: 36071980 PMCID: PMC9446161 DOI: 10.37349/etat.2022.00095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 07/08/2022] [Indexed: 11/19/2022] Open
Abstract
Multiple myeloma (MM) is a blood cancer that derives from plasma cells (PCs), which will accumulate in the bone marrow (BM). Over time, several drugs have been developed to treat this disease that is still uncurable. The therapies used to treat the disease target immune activity, inhibit proteasome activity, and involve the use of monoclonal antibodies. However, MM is a highly heterogeneous disease, in fact, there are several mutations in signaling pathways that are particularly important for MM cell biology and that are possible therapeutic targets. Indeed, some studies suggest that MM is driven by mutations within the rat sarcoma virus (RAS) signaling cascade, which regulates cell survival and proliferation. The RAS/proto-oncogene, serine/threonine kinase (RAF)/mitogen-activated extracellular signal-regulated kinase (ERK) kinase (MEK)/ERK signaling pathway is deregulated in several cancers, for which drugs have been developed to inhibit these pathways. In addition to the signaling pathways, the disease implements mechanisms to ensure the survival and consequently a high replicative capacity. This strategy consists in the deregulation of apoptosis. In particular, some cases of MM show overexpression of anti-apoptotic proteins belonging to the B cell lymphoma 2 (BCL-2) family that represent a possible druggable target. Venetoclax is an anti-BCL-2 molecule used in hematological malignancies that may be used in selected MM patients based on their molecular profile. We focused on the possible effects in MM of off-label drugs that are currently used for other cancers with the same molecular characteristics. Their use, combined with the current treatments, could be a good strategy against MM.
Collapse
Affiliation(s)
- Vincenzo Raimondi
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | | | | | - Denise Toscani
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Paola Storti
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Nicola Giuliani
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy;Hematology, “Azienda Ospedaliero-Universitaria di Parma”, 43126 Parma, Italy
| |
Collapse
|
12
|
Keeping Cell Death Alive: An Introduction into the French Cell Death Research Network. Biomolecules 2022; 12:biom12070901. [PMID: 35883457 PMCID: PMC9313292 DOI: 10.3390/biom12070901] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/16/2022] [Accepted: 06/20/2022] [Indexed: 02/01/2023] Open
Abstract
Since the Nobel Prize award more than twenty years ago for discovering the core apoptotic pathway in C. elegans, apoptosis and various other forms of regulated cell death have been thoroughly characterized by researchers around the world. Although many aspects of regulated cell death still remain to be elucidated in specific cell subtypes and disease conditions, many predicted that research into cell death was inexorably reaching a plateau. However, this was not the case since the last decade saw a multitude of cell death modalities being described, while harnessing their therapeutic potential reached clinical use in certain cases. In line with keeping research into cell death alive, francophone researchers from several institutions in France and Belgium established the French Cell Death Research Network (FCDRN). The research conducted by FCDRN is at the leading edge of emerging topics such as non-apoptotic functions of apoptotic effectors, paracrine effects of cell death, novel canonical and non-canonical mechanisms to induce apoptosis in cell death-resistant cancer cells or regulated forms of necrosis and the associated immunogenic response. Collectively, these various lines of research all emerged from the study of apoptosis and in the next few years will increase the mechanistic knowledge into regulated cell death and how to harness it for therapy.
Collapse
|
13
|
Krstic A, Pavic A, Avdovic E, Markovic Z, Stevanovic M, Petrovic I. Coumarin-Palladium(II) Complex Acts as a Potent and Non-Toxic Anticancer Agent against Pancreatic Carcinoma Cells. Molecules 2022; 27:2115. [PMID: 35408514 PMCID: PMC9000835 DOI: 10.3390/molecules27072115] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/18/2022] [Accepted: 03/19/2022] [Indexed: 12/30/2022] Open
Abstract
Pancreatic carcinoma still represents one of the most lethal malignant diseases in the world although some progress has been made in treating the disease in the past decades. Current multi-agent treatment options have improved the overall survival of patients, however, more effective treatment strategies are still needed. In this paper we have characterized the anticancer potential of coumarin-palladium(II) complex against pancreatic carcinoma cells. Cells viability, colony formation and migratory potential of pancreatic carcinoma cells were assessed in vitro, followed by evaluation of apoptosis induction and in vivo testing on zebrafish. Presented results showed remarkable reduction in pancreatic carcinoma cells growth both in vitro and in vivo, being effective at micromolar concentrations (0.5 μM). Treatments induced apoptosis, increased BAX/BCL-2 ratio and suppressed the expression of SOX9 and SOX18, genes shown to be significantly up-regulated in pancreatic ductal adenocarcinoma. Importantly, treatments of the zebrafish-pancreatic adenocarcinoma xenografts resulted in significant reduction in tumor mass, without provoking any adverse toxic effects including hepatotoxicity. Presented results indicate the great potential of the tested compound and the perspective of its further development towards pancreatic cancer therapy.
Collapse
Affiliation(s)
- Aleksandra Krstic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (A.K.); (A.P.); (M.S.)
| | - Aleksandar Pavic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (A.K.); (A.P.); (M.S.)
| | - Edina Avdovic
- Department of Science, Institute of Information Technologies, University of Kragujevac, Jovana Cvijica bb, 34000 Kragujevac, Serbia; (E.A.); (Z.M.)
| | - Zoran Markovic
- Department of Science, Institute of Information Technologies, University of Kragujevac, Jovana Cvijica bb, 34000 Kragujevac, Serbia; (E.A.); (Z.M.)
| | - Milena Stevanovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (A.K.); (A.P.); (M.S.)
- Faculty of Biology, University of Belgrade, Studentski trg 16, 11000 Belgrade, Serbia
- Department of Chemical and Biological Sciences, Serbian Academy of Sciences and Arts, Kneza Mihaila 35, 11000 Belgrade, Serbia
| | - Isidora Petrovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (A.K.); (A.P.); (M.S.)
| |
Collapse
|
14
|
Lu J, Xiao Z, Xu M, Li L. New Insights into LINC00346 and its Role in Disease. Front Cell Dev Biol 2022; 9:819785. [PMID: 35096842 PMCID: PMC8794746 DOI: 10.3389/fcell.2021.819785] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 12/28/2021] [Indexed: 12/12/2022] Open
Abstract
Accumulating evidence has shown that long intergenic non-protein-coding RNA 346 (LINC00346) functions as an oncogene in the tumorigenesis of several cancers. The expression level of LINC00346 has been shown to be obviously correlated with prognosis, lymphoma metastasis, histological grade, TNM stage, tumor size and pathologic stage. LINC00346 has been found to regulate specific cellular functions by interacting with several molecules and signaling pathways. In this review, we summarize recent evidence concerning the role of LINC00346 in the occurrence and development of diseases. We also discuss the potential clinical utility of LINC00346, thereby providing new insight into the diagnosis and treatment of diseases. In addition, we further discuss the potential clinical utility of LINC00346 in the diagnosis, prognostication, and treatment of diseases.
Collapse
Affiliation(s)
- Juan Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Zhaoying Xiao
- Department of Infectious Diseases Shengzhou People' Hospital, Shengzhou Branch, The Fisrt Affiliated Hospital of Zhejiang University, Shengzhou, China
| | - Mengqiu Xu
- Department of Infectious Diseases Shengzhou People' Hospital, Shengzhou Branch, The Fisrt Affiliated Hospital of Zhejiang University, Shengzhou, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
15
|
Fuentes-Fayos AC, Pérez-Gómez JM, G-García ME, Jiménez-Vacas JM, Blanco-Acevedo C, Sánchez-Sánchez R, Solivera J, Breunig JJ, Gahete MD, Castaño JP, Luque RM. SF3B1 inhibition disrupts malignancy and prolongs survival in glioblastoma patients through BCL2L1 splicing and mTOR/ß-catenin pathways imbalances. J Exp Clin Cancer Res 2022; 41:39. [PMID: 35086552 PMCID: PMC8793262 DOI: 10.1186/s13046-022-02241-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 01/03/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Glioblastoma is one of the most devastating cancer worldwide based on its locally aggressive behavior and because it cannot be cured by current therapies. Defects in alternative splicing process are frequent in cancer. Recently, we demonstrated that dysregulation of the spliceosome is directly associated with glioma development, progression, and aggressiveness. METHODS Different human cohorts and a dataset from different glioma mouse models were analyzed to determine the mutation frequency as well as the gene and protein expression levels between tumor and control samples of the splicing-factor-3B-subunit-1 (SF3B1), an essential and druggable spliceosome component. SF3B1 expression was also explored at the single-cell level across all cell subpopulations and transcriptomic programs. The association of SF3B1 expression with relevant clinical data (e.g., overall survival) in different human cohorts was also analyzed. Different functional (proliferation/migration/tumorspheres and colonies formation/VEGF secretion/apoptosis) and mechanistic (gene expression/signaling pathways) assays were performed in three different glioblastomas cell models (human primary cultures and cell lines) in response to SF3B1 blockade (using pladienolide B treatment). Moreover, tumor progression and formation were monitored in response to SF3B1 blockade in two preclinical xenograft glioblastoma mouse models. RESULTS Our data provide novel evidence demonstrating that the splicing-factor-3B-subunit-1 (SF3B1, an essential and druggable spliceosome component) is low-frequency mutated in human gliomas (~ 1 %) but widely overexpressed in glioblastoma compared with control samples from the different human cohorts and mouse models included in the present study, wherein SF3B1 levels are associated with key molecular and clinical features (e.g., overall survival, poor prognosis and/or drug resistance). Remarkably, in vitro and in vivo blockade of SF3B1 activity with pladienolide B drastically altered multiple glioblastoma pathophysiological processes (i.e., reduction in proliferation, migration, tumorspheres formation, VEGF secretion, tumor initiation and increased apoptosis) likely by suppressing AKT/mTOR/ß-catenin pathways, and an imbalance of BCL2L1 splicing. CONCLUSIONS Together, we highlight SF3B1 as a potential diagnostic and prognostic biomarker and an efficient pharmacological target in glioblastoma, offering a clinically relevant opportunity worth to be explored in humans.
Collapse
Affiliation(s)
- Antonio C Fuentes-Fayos
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004, Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004, Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004, Cordoba, Spain
| | - Jesús M Pérez-Gómez
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004, Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004, Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004, Cordoba, Spain
| | - Miguel E G-García
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004, Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004, Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004, Cordoba, Spain
| | - Juan M Jiménez-Vacas
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004, Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004, Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004, Cordoba, Spain
| | - Cristóbal Blanco-Acevedo
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Córdoba, Spain
- Reina Sofia University Hospital (HURS), 14004, Cordoba, Spain
- Department of Neurosurgery, Reina Sofia University Hospital, 14004, Cordoba, Spain
| | - Rafael Sánchez-Sánchez
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Córdoba, Spain
- Reina Sofia University Hospital (HURS), 14004, Cordoba, Spain
- Pathology Service, Reina Sofia University Hospital, 14004, Cordoba, Spain
| | - Juan Solivera
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Córdoba, Spain
- Reina Sofia University Hospital (HURS), 14004, Cordoba, Spain
- Department of Neurosurgery, Reina Sofia University Hospital, 14004, Cordoba, Spain
| | - Joshua J Breunig
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
- Center for Neural Sciences in Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Manuel D Gahete
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004, Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004, Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004, Cordoba, Spain
| | - Justo P Castaño
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Córdoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004, Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004, Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004, Cordoba, Spain
| | - Raúl M Luque
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004, Córdoba, Spain.
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004, Cordoba, Spain.
- Reina Sofia University Hospital (HURS), 14004, Cordoba, Spain.
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004, Cordoba, Spain.
| |
Collapse
|
16
|
Bcl-2 Family Members and the Mitochondrial Import Machineries: The Roads to Death. Biomolecules 2022; 12:biom12020162. [PMID: 35204663 PMCID: PMC8961529 DOI: 10.3390/biom12020162] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 01/27/2023] Open
Abstract
The localization of Bcl-2 family members at the mitochondrial outer membrane (MOM) is a crucial step in the implementation of apoptosis. We review evidence showing the role of the components of the mitochondrial import machineries (translocase of the outer membrane (TOM) and the sorting and assembly machinery (SAM)) in the mitochondrial localization of Bcl-2 family members and how these machineries regulate the function of pro- and anti-apoptotic proteins in resting cells and in cells committed into apoptosis.
Collapse
|
17
|
Khojasteh Poor F, Keivan M, Ramazii M, Ghaedrahmati F, Anbiyaiee A, Panahandeh S, Khoshnam SE, Farzaneh M. Mini review: The FDA-approved prescription drugs that target the MAPK signaling pathway in women with breast cancer. Breast Dis 2021; 40:51-62. [PMID: 33896802 DOI: 10.3233/bd-201063] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Breast cancer (BC) is the most common cancer and the prevalent type of malignancy among women. Multiple risk factors, including genetic changes, biological age, dense breast tissue, and obesity are associated with BC. The mitogen-activated protein kinases (MAPK) signaling pathway has a pivotal role in regulating biological functions such as cell proliferation, differentiation, apoptosis, and survival. It has become evident that the MAPK pathway is associated with tumorigenesis and may promote breast cancer development. The MAPK/RAS/RAF cascade is closely associated with breast cancer. RAS signaling can enhance BC cell growth and progression. B-Raf is an important kinase and a potent RAF isoform involved in breast tumor initiation and differentiation. Depending on the reasons for cancer, there are different strategies for treatment of women with BC. Till now, several FDA-approved treatments have been investigated that inhibit the MAPK pathway and reduce metastatic progression in breast cancer. The most common breast cancer drugs that regulate or inhibit the MAPK pathway may include Farnesyltransferase inhibitors (FTIs), Sorafenib, Vemurafenib, PLX8394, Dabrafenib, Ulixertinib, Simvastatin, Alisertib, and Teriflunomide. In this review, we will discuss the roles of the MAPK/RAS/RAF/MEK/ERK pathway in BC and summarize the FDA-approved prescription drugs that target the MAPK signaling pathway in women with BC.
Collapse
Affiliation(s)
- Fatemeh Khojasteh Poor
- Department of Obstetrics and Gynecology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mona Keivan
- Fertility and Infertility Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Ramazii
- Kerman University of Medical Sciences, University of Kerman, Kerman, Iran
| | - Farhoodeh Ghaedrahmati
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Amir Anbiyaiee
- Department of Surgery, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Samira Panahandeh
- School of Health, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed Esmaeil Khoshnam
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
18
|
Dou Z, Zhao D, Chen X, Xu C, Jin X, Zhang X, Wang Y, Xie X, Li Q, Di C, Zhang H. Aberrant Bcl-x splicing in cancer: from molecular mechanism to therapeutic modulation. J Exp Clin Cancer Res 2021; 40:194. [PMID: 34118966 PMCID: PMC8196531 DOI: 10.1186/s13046-021-02001-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 05/30/2021] [Indexed: 12/13/2022] Open
Abstract
Bcl-x pre-mRNA splicing serves as a typical example to study the impact of alternative splicing in the modulation of cell death. Dysregulation of Bcl-x apoptotic isoforms caused by precarious equilibrium splicing is implicated in genesis and development of multiple human diseases, especially cancers. Exploring the mechanism of Bcl-x splicing and regulation has provided insight into the development of drugs that could contribute to sensitivity of cancer cells to death. On this basis, we review the multiple splicing patterns and structural characteristics of Bcl-x. Additionally, we outline the cis-regulatory elements, trans-acting factors as well as epigenetic modifications involved in the splicing regulation of Bcl-x. Furthermore, this review highlights aberrant splicing of Bcl-x involved in apoptosis evade, autophagy, metastasis, and therapy resistance of various cancer cells. Last, emphasis is given to the clinical role of targeting Bcl-x splicing correction in human cancer based on the splice-switching oligonucleotides, small molecular modulators and BH3 mimetics. Thus, it is highlighting significance of aberrant splicing isoforms of Bcl-x as targets for cancer therapy.
Collapse
Affiliation(s)
- Zhihui Dou
- Department of Heavy Ion Radiation Medicine, Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 101408, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Dapeng Zhao
- Department of Heavy Ion Radiation Medicine, Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 101408, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Xiaohua Chen
- Department of Heavy Ion Radiation Medicine, Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 101408, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Caipeng Xu
- Department of Heavy Ion Radiation Medicine, Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 101408, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Xiaodong Jin
- Department of Heavy Ion Radiation Medicine, Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
| | - Xuetian Zhang
- Department of Heavy Ion Radiation Medicine, Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 101408, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Yupei Wang
- Medical Genetics Center of Gansu Maternal and Child Health Care Center, Lanzhou, 730000, China
| | - Xiaodong Xie
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Qiang Li
- Department of Heavy Ion Radiation Medicine, Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 101408, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing, 101408, China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516029, China
| | - Cuixia Di
- Department of Heavy Ion Radiation Medicine, Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China.
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China.
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 101408, China.
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing, 101408, China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516029, China.
| | - Hong Zhang
- Department of Heavy Ion Radiation Medicine, Bio-Medical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China.
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, 730000, China.
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 101408, China.
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing, 101408, China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516029, China.
| |
Collapse
|
19
|
Voutsadakis IA. Chromosome 20q11.21 Amplifications in Colorectal Cancer. Cancer Genomics Proteomics 2021; 18:487-496. [PMID: 33994370 DOI: 10.21873/cgp.20274] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/22/2021] [Accepted: 04/26/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Colorectal cancer is the most common gastrointestinal carcinoma in western countries. Prognosis of metastatic colorectal cancer has improved in the last decades, but the disease continues to carry an adverse outcome in most cases. An improved understanding of molecular pathogenesis has provided incremental benefits in survival outcomes with the introduction of targeted therapies for specific sub-types and gives hope for further improvements. MATERIALS AND METHODS Publicly available data from genomic series of colorectal cancer published by the TCGA were analyzed with the aim of characterizing the sub-set of colorectal cancers carrying amplifications of chromosome 20q11.21, compared with cancers with no amplifications in this locus. Associations of 20q11.21-amplified cancers with other molecular lesions commonly observed in colorectal cancer were explored. mRNA expression of genes from the locus in amplified cases was analyzed. An exploratory survival analysis was also performed. RESULTS Amplifications of genes at chromosome arm 20q are observed in 7% to 9% of colorectal cancers, representing the most commonly amplified loci in this type of cancer. The 20q11.21 presents the highest amplification rate in the 20q arm. 20q11.21 amplified cancers display concomitant mutations in the KRAS pathway and SMAD4 less often than non-amplified cancers. Mutations in DNA repair genes are also less often encountered in 20q11.21 amplified colorectal cancers than non-amplified ones. CONCLUSION Amplification of genes at locus 20q11.21, representing the most frequently amplified locus in colorectal cancers, is associated with specific molecular characteristics and may have therapeutic implications.
Collapse
Affiliation(s)
- Ioannis A Voutsadakis
- Algoma District Cancer Program, Sault Area Hospital, Sault Ste. Marie, ON, Canada; .,Section of Internal Medicine, Division of Clinical Sciences, Northern Ontario School of Medicine, Sudbury, ON, Canada
| |
Collapse
|
20
|
Cui M, Zhao Y, Zhang Z, Zhao Y, Han S, Wang R, Ding D, Fang X. IL-8, MSPa, MIF, FGF-9, ANG-2 and AgRP collection were identified for the diagnosis of colorectal cancer based on the support vector machine model. Cell Cycle 2021; 20:781-791. [PMID: 33779485 PMCID: PMC8098075 DOI: 10.1080/15384101.2021.1903208] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 12/08/2020] [Accepted: 03/11/2021] [Indexed: 10/21/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancer, and the early detection of CRC is essential to improve the survival rate of patients. To identify diagnostic markers for colorectal cancer (CRC) by screening differentially expressed proteins (DEPs) in CRC. The DEPs were initially obtained from 12 CRC samples and 12 healthy control samples, and verification analysis was performed in another 34 CRC samples and 34 normal controls. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment with DEPs was analyzed by the R package clusterProfiler (Version 3.2.11), and the DEP-associated protein-protein interaction (PPI) network was created from the STRING database. Additionally, Support Vector Machine (SVM) model prediction and survival analyses were conducted on the key DEPs. Preliminary screening and functional analysis showed that the DEPs mainly overrepresented in pathways such as cytokine-cytokine receptor interaction, chemokine signaling pathway, Rap1, Ras, and MAPK signaling pathways. The key DEPs, including AgRP, ANG-2, Dtk, EOT3, FGF-4, FGF-9, HCC-4, IL-16, IL-8, MIF, MSPa, TECK, TPO, TRAIL R3, and VEGF-D, were used to construct a custom chip. The drug-gene interaction network suggested that TPO was a key drug target. ROC curve showed the SVM diagnostic model with the DEPs IL-8, MSPa, MIF, FGF-9, ANG-2, and AgRP had better diagnostic performance with an AUC of 0.933. Survival analysis showed the expression of FGF9, TPO, TRAIL R3, Dtk, TECK and FGF4 were associated with prognosis. This study revealed the important serum proteins in the pathogenesis of CRC, which might serve as useful and noninvasive predictors for the diagnosis of CRC.
Collapse
Affiliation(s)
- Mingfu Cui
- Department of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin Province, China
| | - Yanan Zhao
- Department of Oncology and Hematology Surgery, China-Japan Union Hospital, Changchun, Jilin Province, China
| | - Zuocong Zhang
- Department of Colorectal Surgery, Jilin Province People’s Hospital, Changchun, Jilin Province, China
| | - Yang Zhao
- Anorectal Surgery, Siping Central People’s Hospital, Jilin University, Siping, Jilin Province, China
| | - Songyun Han
- Emergency Department, Tonghua Central Hospital, Jilin University, Tonghua, Jilin Province, China
| | - Ruijie Wang
- Department of Gastrointestinal Surgery, Shengli Oilfield Central Hospital, Dongying, Jilin Province, China
| | - Dayong Ding
- Department of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin Province, China
| | - Xuedong Fang
- Department of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
21
|
Wyżewski Z, Świtlik W, Mielcarska MB, Gregorczyk-Zboroch KP. The Role of Bcl-xL Protein in Viral Infections. Int J Mol Sci 2021; 22:ijms22041956. [PMID: 33669408 PMCID: PMC7920434 DOI: 10.3390/ijms22041956] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/13/2021] [Accepted: 02/14/2021] [Indexed: 02/06/2023] Open
Abstract
Bcl-xL represents a family of proteins responsible for the regulation of the intrinsic apoptosis pathway. Due to its anti-apoptotic activity, Bcl-xL co-determines the viability of various virally infected cells. Their survival may determine the effectiveness of viral replication and spread, dynamics of systemic infection, and viral pathogenesis. In this paper, we have reviewed the role of Bcl-xL in the context of host infection by eight different RNA and DNA viruses: hepatitis B virus (HBV), hepatitis C virus (HCV), human immunodeficiency virus (HIV), influenza A virus (IAV), Epstein-Barr virus (EBV), human T-lymphotropic virus type-1 (HTLV-1), Maraba virus (MRBV), Schmallenberg virus (SBV) and coronavirus (CoV). We have described an influence of viral infection on the intracellular level of Bcl-xL and discussed the impact of Bcl-xL-dependent cell survival control on infection-accompanying pathogenic events such as tissue damage or oncogenesis. We have also presented anti-viral treatment strategies based on the pharmacological regulation of Bcl-xL expression or activity.
Collapse
Affiliation(s)
- Zbigniew Wyżewski
- Institute of Biological Sciences, Cardinal Stefan Wyszyński University in Warsaw, 01-815 Warsaw, Poland
- Correspondence: ; Tel.: +48 728-208-338
| | - Weronika Świtlik
- Department of Biochemistry and Microbiology, Institute of Biology, Warsaw University of Life Sciences-SGGW, 02-787 Warsaw, Poland;
| | - Matylda Barbara Mielcarska
- Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (M.B.M.); (K.P.G.-Z.)
| | | |
Collapse
|
22
|
Al-Mansoob M, Gupta I, Stefan Rusyniak R, Ouhtit A. KYNU, a novel potential target that underpins CD44-promoted breast tumour cell invasion. J Cell Mol Med 2021; 25:2309-2314. [PMID: 33486887 PMCID: PMC7933956 DOI: 10.1111/jcmm.16296] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 12/29/2020] [Indexed: 12/14/2022] Open
Abstract
Using a validated tetracycline‐off‐inducible CD44 expression system in mouse model, we have previously demonstrated that the hyaluronan (HA) receptor CD44 promotes breast cancer (BC) metastasis to the liver. To unravel the mechanisms that underpin CD44‐promoted BC cell invasion, RNA samples were isolated from two cell models: (a) a tetracycline (Tet)‐Off‐regulated expression system of the CD44s in MCF‐7 cells and; (b) as a complementary approach, the highly metastatic BC cells, MDA‐MB‐231, were cultured in the presence and absence of 50 µg/mL of HA. Kynureninase (KYNU), identified by Microarray analysis, was up‐regulated by 3‐fold upon induction and activation of CD44 by HA; this finding suggests that KYNU is a potential novel transcriptional target of CD44‐downtstream signalling. KYNU is a pyridoxal phosphate (PLP) dependent enzyme involved in the biosynthesis of NAD cofactors from tryptophan that has been associated with the onset and development of BC. This review will attempt to identify and discuss the findings supporting this hypothesis and the mechanisms linking KYNU cell invasion via CD44.
Collapse
Affiliation(s)
- Maryam Al-Mansoob
- Department of Biological & Environmental Sciences, College of Arts and Science, Qatar University, Doha, Qatar
| | - Ishita Gupta
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Radoslaw Stefan Rusyniak
- Department of Biological & Environmental Sciences, College of Arts and Science, Qatar University, Doha, Qatar
| | - Allal Ouhtit
- Department of Biological & Environmental Sciences, College of Arts and Science, Qatar University, Doha, Qatar
| |
Collapse
|
23
|
TRIB2 modulates proteasome function to reduce ubiquitin stability and protect liver cancer cells against oxidative stress. Cell Death Dis 2021; 12:42. [PMID: 33414446 PMCID: PMC7791120 DOI: 10.1038/s41419-020-03299-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 11/26/2020] [Accepted: 11/30/2020] [Indexed: 02/07/2023]
Abstract
The regulation of homeostasis in the Ubiquitin (Ub) proteasome system (UPS) is likely to be important for the development of liver cancer. Tribbles homolog 2 (TRIB2) is known to affect Ub E3 ligases (E3s) in liver cancer. However, whether TRIB2 regulates the UPS in other ways and the relevant mechanisms are still unknown. Here, we reveal that TRIB2 decreased Ub levels largely by stimulating proteasome degradation of Ub. In the proteasome, proteasome 20S subunit beta 5 (PSMB5) was critical for the function of TRIB2, although it did not directly interact with TRIB2. However, poly (rC) binding protein 2 (PCBP2), which was identified by mass spectrometry, directly interacted with both TRIB2 and PSMB5. PCBP2 was a prerequisite for the TRIB2 induction of PSMB5 activity and decreased Ub levels. A significant correlation between TRIB2 and PCBP2 was revealed in liver cancer specimens. Interestingly, TRIB2 suppressed the K48-ubiquitination of PCBP2 to increase its level. Therefore, a model showing that TRIB2 cooperates and stimulates PCBP2 to reduce Ub levels was established. Additionally, the reduction in Ub levels induced by TRIB2 and PCBP2 was dependent on K48-ubiquitination. PCBP2 was one of the possible downstream factors of TRIB2 and their interaction relied on the DQLVPD element of TRIB2 and the KH3 domain of PCBP2. This interaction was necessary to maintain the viability of the liver cancer cells and promote tumor growth. Mechanistically, glutathione peroxidase 4 functioned as one of the terminal effectors of TRIB2 and PCBP2 to protect liver cancer cells from oxidative damage. Taken together, the data indicate that, in addition to affecting E3s, TRIB2 plays a critical role in regulating UPS by modulating PSMB5 activity in proteasome to reduce Ub flux, and that targeting TRIB2 might be helpful in liver cancer treatments by enhancing the oxidative damage induced by therapeutic agents.
Collapse
|
24
|
Makhafola TJ, Mbele M, Yacqub-Usman K, Hendren A, Haigh DB, Blackley Z, Meyer M, Mongan NP, Bates DO, Dlamini Z. Apoptosis in Cancer Cells Is Induced by Alternative Splicing of hnRNPA2/B1 Through Splicing of Bcl-x, a Mechanism that Can Be Stimulated by an Extract of the South African Medicinal Plant, Cotyledon orbiculata. Front Oncol 2020; 10:547392. [PMID: 33163396 PMCID: PMC7580256 DOI: 10.3389/fonc.2020.547392] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 08/21/2020] [Indexed: 01/09/2023] Open
Abstract
Alternative splicing is deregulated in cancer and alternatively spliced products can be linked to cancer hallmarks. Targeting alternative splicing could offer novel effective cancer treatments. We investigated the effects of the crude extract of a South African medicinal plant, Cotyledon orbiculata, on cell survival of colon (HCT116) and esophageal (OE33 and KYSE70) cancer cell lines. Using RNASeq, we discovered that the extract interfered with mRNA regulatory pathways. The extract caused hnRNPA2B1 to splice from the hnRNPB1 to the hnRNPA2 isoform, resulting in a switch in the BCL2L1 gene from Bcl-xL to Bcl-xS causing activation of caspase-3-cleavage and apoptosis. Similar splicing effects were induced by the known anti-cancer splicing modulator pladienolide B. Knockdown of hnRNPB1 using siRNA resulted in decreased cell viability and increased caspase-3-cleavage, and over-expression of hnRNPB1 prevented the effect of C. orbiculata extract on apoptosis and cell survival. The effect of the hnRNPA2/B1 splicing switch by the C. orbiculata extract increased hnRNPA2B1 binding to Bcl-xl/s, BCL2, MDM2, cMYC, CD44, CDK6, and cJUN mRNA. These findings suggest that apoptosis in HCT116, OE33, and KYSE cancer cells is controlled by switched splicing of hnRNPA2B1 and BCL2L1, providing evidence that hnRNPB1 regulates apoptosis. Inhibiting this splicing could have therapeutic potential for colon and esophageal cancers. Targeting hnRNPA2B1 splicing in colon cancer regulates splicing of BCL2L1 to induce apoptosis. This approach could be a useful therapeutic strategy to induce apoptosis and restrain cancer cell proliferation and tumor progression. Here, we found that the extract of Cotyledon orbiculata, a South African medicinal plant, had an anti-proliferative effect in cancer cells, mediated by apoptosis induced by alternative splicing of hnRNPA2B1 and BCL2L1.
Collapse
Affiliation(s)
- Tshepiso Jan Makhafola
- SA-Medical Research Council (MRC)/UP Precision Prevention & Novel Drug Targets for HIV-Associated Cancers Extramural Unit, Faculty of Health Sciences, Pan African Cancer Research Institute (PACRI), University of Pretoria, Pretoria, South Africa.,Division of Cancer and Stem Cells, Centre for Cancer Sciences, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Mzwandile Mbele
- SA-Medical Research Council (MRC)/UP Precision Prevention & Novel Drug Targets for HIV-Associated Cancers Extramural Unit, Faculty of Health Sciences, Pan African Cancer Research Institute (PACRI), University of Pretoria, Pretoria, South Africa.,Division of Cancer and Stem Cells, Centre for Cancer Sciences, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Kiren Yacqub-Usman
- Division of Cancer and Stem Cells, Centre for Cancer Sciences, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Amy Hendren
- Division of Cancer and Stem Cells, Centre for Cancer Sciences, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Daisy Belle Haigh
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom
| | - Zoe Blackley
- Division of Cancer and Stem Cells, Centre for Cancer Sciences, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Mervin Meyer
- Biolabels Unit, Department of Biotechnology, Department of Science and Technology (DST)/Mintek Nanotechnology Innovation Centre (NIC), University of the Western Cape, Bellville, South Africa
| | - Nigel Patrick Mongan
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom
| | - David Owen Bates
- Division of Cancer and Stem Cells, Centre for Cancer Sciences, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Zodwa Dlamini
- SA-Medical Research Council (MRC)/UP Precision Prevention & Novel Drug Targets for HIV-Associated Cancers Extramural Unit, Faculty of Health Sciences, Pan African Cancer Research Institute (PACRI), University of Pretoria, Pretoria, South Africa
| |
Collapse
|
25
|
Khan AQ, Rashid K, AlAmodi AA, Raza SS, Uddin S. Recent developments in unraveling signaling mechanisms underlying drug resistance due to cancer stem-like cells. Curr Opin Pharmacol 2020; 54:130-141. [PMID: 33166909 DOI: 10.1016/j.coph.2020.09.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 12/20/2022]
Abstract
Resistance in cancer cells to therapeutic measures is challenging and requires a rigorous delineation of the underlying mechanisms. Emerging findings reflect the characteristics of tumor cells to do the reprogramming of signaling machinery in order to overturn the therapeutic responses. Recent evidence shows that the tumor acquires drug resistance due to the presence of cancer stem cells (CSCs). Hence the understanding that how tumor cells reprogram their signaling mechanisms converging towards the stemness of CSCs is imperative for novel and effective therapy. This review outlines the current updates on how CSC-associated signaling pathways and its enhanced stemness trigger the development of drug resistance. Furthermore, we also discussed the strategies with a combinational approach that can simultaneously target both CSC-induced stemness and the resistance-related signaling pathways, which may provide an optimal outcome to overcome the problem of drug resistance in cancer therapy.
Collapse
Affiliation(s)
- Abdul Q Khan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Khalid Rashid
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | | | - Syed Shadab Raza
- Department of Stem Cell Biology and Regenerative Medicine, Era University, Lucknow, India
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar.
| |
Collapse
|
26
|
Pegoraro S, Ros G, Sgubin M, Petrosino S, Zambelli A, Sgarra R, Manfioletti G. Targeting the intrinsically disordered architectural High Mobility Group A (HMGA) oncoproteins in breast cancer: learning from the past to design future strategies. Expert Opin Ther Targets 2020; 24:953-969. [PMID: 32970506 DOI: 10.1080/14728222.2020.1814738] [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: 02/07/2023]
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) is the most difficult breast cancer subtype to treat because of its heterogeneity and lack of specific therapeutic targets. High Mobility Group A (HMGA) proteins are chromatin architectural factors that have multiple oncogenic functions in breast cancer, and they represent promising molecular therapeutic targets for this disease. AREAS COVERED We offer an overview of the strategies that have been exploited to counteract HMGA oncoprotein activities at the transcriptional and post-transcriptional levels. We also present the possibility of targeting cancer-associated factors that lie downstream of HMGA proteins and discuss the contribution of HMGA proteins to chemoresistance. EXPERT OPINION Different strategies have been exploited to counteract HMGA protein activities; these involve interfering with their nucleic acid binding properties and the blocking of HMGA expression. Some approaches have provided promising results. However, some unique characteristics of the HMGA proteins have not been exploited; these include their extensive protein-protein interaction network and their intrinsically disordered status that present the possibility that HMGA proteins could be involved in the formation of proteinaceous membrane-less organelles (PMLO) by liquid-liquid phase separation. These unexplored characteristics could open new pharmacological avenues to counteract the oncogenic contributions of HMGA proteins.
Collapse
Affiliation(s)
- Silvia Pegoraro
- Department of Life Sciences, University of Trieste , Trieste, Italy
| | - Gloria Ros
- Department of Life Sciences, University of Trieste , Trieste, Italy
| | - Michela Sgubin
- Department of Life Sciences, University of Trieste , Trieste, Italy
| | - Sara Petrosino
- Department of Life Sciences, University of Trieste , Trieste, Italy
| | | | - Riccardo Sgarra
- Department of Life Sciences, University of Trieste , Trieste, Italy
| | | |
Collapse
|
27
|
Murray AS, Hyland TE, Sala-Hamrick KE, Mackinder JR, Martin CE, Tanabe LM, Varela FA, List K. The cell-surface anchored serine protease TMPRSS13 promotes breast cancer progression and resistance to chemotherapy. Oncogene 2020; 39:6421-6436. [PMID: 32868877 DOI: 10.1038/s41388-020-01436-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 08/08/2020] [Accepted: 08/17/2020] [Indexed: 12/18/2022]
Abstract
Breast cancer progression is accompanied by increased expression of extracellular and cell-surface proteases capable of degrading the extracellular matrix as well as cleaving and activating downstream targets. The type II transmembrane serine proteases (TTSPs) are a family of cell-surface proteases that play critical roles in numerous types of cancers. Therefore, the aim of this study was to identify novel and uncharacterized TTSPs with differential expression in breast cancer and to determine their potential roles in progression. Systematic in silico data analysis followed by immunohistochemical validation identified increased expression of the TTSP family member, TMPRSS13 (transmembrane protease, serine 13), in invasive ductal carcinoma patient tissue samples compared to normal breast tissue. To test whether loss of TMPRSS13 impacts tumor progression, TMPRSS13 was genetically ablated in the oncogene-induced transgenic MMTV-PymT tumor model. TMPRSS13 deficiency resulted in a significant decrease in overall tumor burden and growth rate, as well as a delayed formation of detectable mammary tumors, thus suggesting a causal relationship between TMPRSS13 expression and the progression of breast cancer. Complementary studies using human breast cancer cell culture models revealed that siRNA-mediated silencing of TMPRSS13 expression decreases proliferation, induces apoptosis, and attenuates invasion. Importantly, targeting TMPRSS13 expression renders aggressive triple-negative breast cancer cell lines highly responsive to chemotherapy. At the molecular level, knockdown of TMPRSS13 in breast cancer cells led to increased protein levels of the tumor-suppressive protease prostasin. TMPRSS13/prostasin co-immunoprecipitation and prostasin zymogen activation experiments identified prostasin as a potential novel target for TMPRSS13. Regulation of prostasin levels may be a mechanism that contributes to the pro-oncogenic properties of TMPRSS13 in breast cancer. TMPRSS13 represents a novel candidate for targeted therapy in combination with standard of care chemotherapy agents in patients with hormone receptor-negative breast cancer or in patients with tumors refractory to endocrine therapy.
Collapse
Affiliation(s)
- Andrew S Murray
- Department of Pharmacology, Wayne State University, Detroit, MI, USA.,Department of Oncology, Wayne State University, Detroit, MI, USA
| | - Thomas E Hyland
- Department of Pharmacology, Wayne State University, Detroit, MI, USA
| | | | - Jacob R Mackinder
- Department of Pharmacology, Wayne State University, Detroit, MI, USA
| | - Carly E Martin
- Department of Pharmacology, Wayne State University, Detroit, MI, USA.,Department of Oncology, Wayne State University, Detroit, MI, USA
| | - Lauren M Tanabe
- Department of Pharmacology, Wayne State University, Detroit, MI, USA
| | - Fausto A Varela
- Department of Pharmacology, Wayne State University, Detroit, MI, USA
| | - Karin List
- Department of Pharmacology, Wayne State University, Detroit, MI, USA. .,Department of Oncology, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
28
|
Barillé-Nion S, Lohard S, Juin PP. Targeting of BCL-2 Family Members during Anticancer Treatment: A Necessary Compromise between Individual Cell and Ecosystemic Responses? Biomolecules 2020; 10:E1109. [PMID: 32722518 PMCID: PMC7464802 DOI: 10.3390/biom10081109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/15/2020] [Accepted: 07/22/2020] [Indexed: 01/07/2023] Open
Abstract
The imbalance between BCL-2 homologues and pro-death counterparts frequently noted in cancer cells endows them with a cell autonomous survival advantage. To eradicate ectopic cells, inhibitors of these homologues (BH3 mimetics) were developed to trigger, during anticancer treatment, full activation of the canonical mitochondrial apoptotic pathway and related caspases. Despite efficiency in some clinical settings, these compounds do not completely fulfill their initial promise. We herein put forth that a growing body of evidence indicates that mitochondrial integrity, controlled by BCL-2 family proteins, and downstream caspases regulate other cell death modes and influence extracellular signaling by committed cells. Moreover, intercellular communications play a key role in spreading therapeutic response across cancer cell populations and in engaging an immune response. We thus advocate that BH3 mimetics administration would be more efficient in the long term if it did not induce apoptosis in all sensitive cells at the same time, but if it could instead allow (or trigger) death signal production by non-terminally committed dying cell populations. The development of such a trade-off strategy requires to unravel the effects of BH3 mimetics not only on each individual cancer cell but also on homotypic and heterotypic cell interactions in dynamic tumor ecosystems.
Collapse
Affiliation(s)
- Sophie Barillé-Nion
- Centre de Recherche en Cancérologie et Immunologie Nantes Angers (CRCINA), INSERMU1232, Université de Nantes, F-44000 Nantes, France; (S.B.-N.); (S.L.)
- SIRIC ILIAD, 44000 Nantes, France
| | - Steven Lohard
- Centre de Recherche en Cancérologie et Immunologie Nantes Angers (CRCINA), INSERMU1232, Université de Nantes, F-44000 Nantes, France; (S.B.-N.); (S.L.)
- Radiation Oncology Branch, National Cancer Institute, Bethesda, MD 20892, USA
| | - Philippe P. Juin
- Centre de Recherche en Cancérologie et Immunologie Nantes Angers (CRCINA), INSERMU1232, Université de Nantes, F-44000 Nantes, France; (S.B.-N.); (S.L.)
- SIRIC ILIAD, 44000 Nantes, France
- Institut de Cancérologie de l’Ouest, 15 Rue André Boquel, 49055 Angers, France
| |
Collapse
|
29
|
Andersen CJ, Dupree L, Murray K, Ragonesi N, McMullen K, Cintrón-Rivera L, Doerr A. Low-Density Lipoproteins, High-Density Lipoproteins (HDL), and HDL-Associated Proteins Differentially Modulate Chronic Myelogenous Leukemia Cell Viability. Lipids 2020; 55:615-626. [PMID: 32558932 DOI: 10.1002/lipd.12254] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 04/17/2020] [Accepted: 05/05/2020] [Indexed: 12/20/2022]
Abstract
Cellular lipid metabolism, lipoprotein interactions, and liver X receptor (LXR) activation have been implicated in the pathophysiology and treatment of cancer, although findings vary across cancer models and by lipoprotein profiles. In this study, we investigated the effects of human-derived low-density lipoproteins (LDL), high-density lipoproteins (HDL), and HDL-associated proteins apolipoprotein A1 (apoA1) and serum amyloid A (SAA) on markers of viability, cholesterol flux, and differentiation in K562 cells-a bone marrow-derived, stem-like erythroleukemia cell model of chronic myelogenous leukemia (CML). We further evaluated whether lipoprotein-mediated effects were altered by concomitant LXR activation. We observed that LDL promoted higher K562 cell viability in a dose- and time-dependent manner and increased cellular cholesterol concentrations, while LXR activation by the agonist TO901317 ablated these effects. LXR activation in the presence of HDL, apoA1 and SAA-rich HDL suppressed K562 cell viability, while robustly inducing mRNA expression of ATP-binding cassette transporter A1 (ABCA1). HDL and its associated proteins additionally suppressed mRNA expression of anti-apoptotic B-cell lymphoma-extra large (BCL-xL), and the erythroid lineage marker 5'-aminolevulinate synthase 2 (ALAS2), while SAA-rich HDL induced mRNA expression of the megakaryocytic lineage marker integrin subunit alpha 2b (ITGA2B). Together, these findings suggest that lipoproteins and LXR may impact the viability and characteristics of CML cells.
Collapse
Affiliation(s)
| | - Lydia Dupree
- Department of Biology, Fairfield University, Fairfield, CT, 06824, USA
| | - Kristina Murray
- Department of Biology, Fairfield University, Fairfield, CT, 06824, USA
| | - Nicholas Ragonesi
- Department of Biology, Fairfield University, Fairfield, CT, 06824, USA
| | - Kaley McMullen
- Department of Biology, Fairfield University, Fairfield, CT, 06824, USA
| | | | - Adam Doerr
- Department of Biology, Fairfield University, Fairfield, CT, 06824, USA
| |
Collapse
|
30
|
Roles and mechanisms of alternative splicing in cancer - implications for care. Nat Rev Clin Oncol 2020; 17:457-474. [PMID: 32303702 DOI: 10.1038/s41571-020-0350-x] [Citation(s) in RCA: 462] [Impact Index Per Article: 92.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2020] [Indexed: 12/14/2022]
Abstract
Removal of introns from messenger RNA precursors (pre-mRNA splicing) is an essential step for the expression of most eukaryotic genes. Alternative splicing enables the regulated generation of multiple mRNA and protein products from a single gene. Cancer cells have general as well as cancer type-specific and subtype-specific alterations in the splicing process that can have prognostic value and contribute to every hallmark of cancer progression, including cancer immune responses. These splicing alterations are often linked to the occurrence of cancer driver mutations in genes encoding either core components or regulators of the splicing machinery. Of therapeutic relevance, the transcriptomic landscape of cancer cells makes them particularly vulnerable to pharmacological inhibition of splicing. Small-molecule splicing modulators are currently in clinical trials and, in addition to splice site-switching antisense oligonucleotides, offer the promise of novel and personalized approaches to cancer treatment.
Collapse
|
31
|
Li S, Zhao W, Tao Y, Liu C. Fugan Wan alleviates hepatic fibrosis by inhibiting ACE/Ang II/AT-1R signaling pathway and enhancing ACE2/Ang 1-7/Mas signaling pathway in hepatic fibrosis rat models. Am J Transl Res 2020; 12:592-601. [PMID: 32194907 PMCID: PMC7061829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 05/11/2019] [Indexed: 06/10/2023]
Abstract
Hepatic fibrosis is a repair and healing reaction for chronic injuries of liver. This study aimed to investigate protective effects of Fugan Wan (FGW) on hepatic fibrosis and clarify associated mechanisms. Hepatic fibrosis model was established by administrating dimethyl nitrosamine (DMN) to rats. Rats were divided into control, DMN and FGW groups. Haematoxylin and eosin (HE) staining was conducted to evaluate inflammatory response in hepatic fibrosis tissues. Sirius red staining was used to assess collagen disposition. Quantitative real-time PCR (qRT-PCR) was employed to detect antiotensin-converting enzyme homologue 2 (ACE2), Mas, transforming growth factor β1 (TGF-β1) mRNA. Western blot was used to examine collagen I, smooth muscle actin α (α-SMA), angiotensin type 1 receptor (AT-1R), extra-cellular regulated protein kinase (ERK), phosphorylated ERK (p-ERK), c-Jun and phosphorylated-c-Jun (p-c-Jun) expression. The results indicated that FGW significantly reduced inflammatory response of hepatic fibrosis tissues. FGW significantly decreased collagen deposition compared to that of DMN group (P < 0.01). FGW significantly down-regulated α-SMA expression compared to that of DMN group (P < 0.01). FGW significantly decreased AT-1R levels compared to that of DMN group (P < 0.01). Comparing with DMN group, ACE2 and Mas mRNA levels were significantly increased in FGW group (P < 0.01). FGW significantly down-regulated p-c-Jun and p-ERK1/2 compared to DMN group (P < 0.01). GFW significantly inhibited compared to DMN group (P < 0.01). In conclusion, FGW alleviated hepatic fibrosis by inhibiting ACE/Ang II/AT-1R signaling and enhancing ACE2/Ang 1-7/Mas signaling pathway in hepatic fibrosis rat models.
Collapse
Affiliation(s)
- Shu Li
- Department of Gastroenterology, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai 201900, China
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai 201203, China
| | - Wei Zhao
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai 201203, China
| | - Yanyan Tao
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai 201203, China
| | - Chenghai Liu
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai 201203, China
- Shanghai Key Laboratory of Traditional Chinese Clinical MedicineShanghai 201203, China
- Key Laboratory of Liver and Kidney Diseases, Ministry of EducationShanghai 201203, China
| |
Collapse
|
32
|
Bolan PO, Zviran A, Brenan L, Schiffman JS, Dusaj N, Goodale A, Piccioni F, Johannessen CM, Landau DA. Genotype-Fitness Maps of EGFR-Mutant Lung Adenocarcinoma Chart the Evolutionary Landscape of Resistance for Combination Therapy Optimization. Cell Syst 2020; 10:52-65.e7. [PMID: 31668800 PMCID: PMC6981068 DOI: 10.1016/j.cels.2019.10.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 05/21/2019] [Accepted: 09/30/2019] [Indexed: 12/12/2022]
Abstract
Cancer evolution poses a central obstacle to cure, as resistant clones expand under therapeutic selection pressures. Genome sequencing of relapsed disease can nominate genomic alterations conferring resistance but sample collection lags behind, limiting therapeutic innovation. Genome-wide screens offer a complementary approach to chart the compendium of escape genotypes, anticipating clinical resistance. We report genome-wide open reading frame (ORF) resistance screens for first- and third-generation epidermal growth factor receptor (EGFR) inhibitors and a MEK inhibitor. Using serial sampling, dose gradients, and mathematical modeling, we generate genotype-fitness maps across therapeutic contexts and identify alterations that escape therapy. Our data expose varying dose-fitness relationship across genotypes, ranging from complete dose invariance to paradoxical dose dependency where fitness increases in higher doses. We predict fitness with combination therapy and compare these estimates to genome-wide fitness maps of drug combinations, identifying genotypes where combination therapy results in unexpected inferior effectiveness. These data are applied to nominate combination optimization strategies to forestall resistant disease.
Collapse
Affiliation(s)
| | - Asaf Zviran
- Division of Hematology and Medical Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA; New York Genome Center, New York, NY 10013, USA; Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Lisa Brenan
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Joshua S Schiffman
- Division of Hematology and Medical Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA; New York Genome Center, New York, NY 10013, USA; Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10065, USA
| | | | - Amy Goodale
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | | | - Dan A Landau
- Division of Hematology and Medical Oncology, Department of Medicine and Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA; New York Genome Center, New York, NY 10013, USA; Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
33
|
Baruah T, Hauneihkim K, Kma L. Naringenin sensitizes lung cancer NCI-H23 cells to radiation by downregulation of akt expression and metastasis while promoting apoptosis. Pharmacogn Mag 2020. [DOI: 10.4103/pm.pm_535_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
34
|
Abstract
Oncogenic activation of RAS isoforms leads tumor initiation and progression in many types of cancers and is gaining increasing interest as target for novel therapeutic strategies. In sharp contrast with other types of cancer, the importance of RAS in breast tumorigenesis has long been undermined by the low frequency of its oncogenic mutation in human breast lesions. Nevertheless, a wealth of studies over the last years have revealed how the engagement of RAS function might be mandatory downstream varied oncogenic alterations for the progression, metastatic dissemination, and therapy resistance in breast cancers. We review herein the major studies over the last three decades which have explored the controversial role of RAS proteins and their mutation status in breast tumorigenesis and have contributed to reveal their role as supporting actors, instead of as primary cause, in breast cancer.
Collapse
Affiliation(s)
- Mirco Galiè
- Department of Neuroscience, Biomedicine and Movement, University of Verona, Verona, Italy
| |
Collapse
|
35
|
Improved Electrophoretic Separation to Assist the Monitoring of Bcl-xL Post-Translational Modifications. Int J Mol Sci 2019; 20:ijms20225571. [PMID: 31717257 PMCID: PMC6888115 DOI: 10.3390/ijms20225571] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/25/2019] [Accepted: 11/05/2019] [Indexed: 01/20/2023] Open
Abstract
Bcl-xL is an oncogene of which the survival functions are finely tuned by post-translational modifications (PTM). Within the Bcl-2 family of proteins, Bcl-xL shows unique eligibility to deamidation, a time-related spontaneous reaction. Deamidation is still a largely overlooked PTM due to a lack of easy techniques to monitor Asn→Asp/IsoAsp conversions or Glu→Gln conversions. Being able to detect PTMs is essential to achieve a comprehensive description of all the regulatory mechanisms and functions a protein can carry out. Here, we report a gel composition improving the electrophoretic separation of deamidated forms of Bcl-xL generated either by mutagenesis or by alkaline treatment. Importantly, this new gel formulation proved efficient to provide the long-sought evidence that even doubly-deamidated Bcl-xL remains eligible for regulation by phosphorylation.
Collapse
|
36
|
Saravanakumar K, Hu X, Shanmugam S, Chelliah R, Sekar P, Oh DH, Vijayakumar S, Kathiresan K, Wang MH. Enhanced cancer therapy with pH-dependent and aptamer functionalized doxorubicin loaded polymeric (poly D, L-lactic-co-glycolic acid) nanoparticles. Arch Biochem Biophys 2019; 671:143-151. [PMID: 31283911 DOI: 10.1016/j.abb.2019.07.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 07/02/2019] [Accepted: 07/04/2019] [Indexed: 12/11/2022]
Abstract
Aptamer based drug delivery systems are gaining the importance in anticancer therapy due to their targeted drug delivery efficiency without harming the normal cells. The present work formulated the pH-dependent aptamer functionalized polymer-based drug delivery system against human lung cancer. The prepared aptamer functionalized doxorubicin (DOX) loaded poly (D, L-lactic-co-glycolic acid) (PLGA), poly (N-vinylpyrrolidone) (PVP) nanoparticles (APT-DOX-PLGA-PVP NPs) were spherical in shape with an average size of 87.168 nm. The crystallography and presence of the PLGA (poly (D, L-lactic-co-glycolic acid)) and DOX (doxorubicin) in APT-DOX-PLGA-PVP NPs were indicated by the X-ray diffraction (XRD), Fourier transforms infrared spectroscopy (FTIR), and 1H and 13C nuclear magnetic resonance spectrometer (NMR). The pH-dependent aptamer AS1411 based drug release triggered the cancer cell death was evidenced by cytotoxicity assay, flow cytometry, and fluorescent microscopic imaging. In addition, the cellular uptake of the DOX was determined and the apoptosis-related signaling pathway in the A549 cells was studied by Western blot analysis. Further, the in vivo study revealed that mice treated with APT-DOX-PLGA-PVP NPs were significantly recovered from cancer as evident by mice weight and tumor size followed by the histopathological study. It was reported that the APT-DOX-PLGA-PVP NPs induced the apoptosis through the activation of the apoptosis-related proteins. Hence, the present study revealed that the APT-DOX-PLGA-PVP NPs improved the therapeutic efficiency through the nucleolin receptor endocytosis targeted drug release.
Collapse
Affiliation(s)
- Kandasamy Saravanakumar
- Department of Medical Biotechnology, College of Biomedical Sciences, Kangwon National University, Chuncheon, 200-701, Republic of Korea
| | - Xiaowen Hu
- Department of Medical Biotechnology, College of Biomedical Sciences, Kangwon National University, Chuncheon, 200-701, Republic of Korea
| | - Sabarathinam Shanmugam
- Bioprocess Laboratory, Department of Microbial Biotechnology, Bharathiar University, Coimbatore, 641046, Tamil Nadu, India
| | - Ramachandran Chelliah
- Department of Food Science and Biotechnology College of Biotechnology and Bioscience, Kangwon National University, Chuncheon, Republic of Korea
| | - Ponarulselvam Sekar
- Bioprocess Laboratory, Department of Microbial Biotechnology, Bharathiar University, Coimbatore, 641046, Tamil Nadu, India
| | - Deog-Hwan Oh
- Department of Food Science and Biotechnology College of Biotechnology and Bioscience, Kangwon National University, Chuncheon, Republic of Korea
| | - Sekar Vijayakumar
- Department of Medical Biotechnology, College of Biomedical Sciences, Kangwon National University, Chuncheon, 200-701, Republic of Korea
| | - Kandasamy Kathiresan
- Centre of Advanced Study in Marine Biology, Faculty of Marine Sciences, Annamalai University, Parangipettai, 608 502, Tamil Nadu, India
| | - Myeong-Hyeon Wang
- Department of Medical Biotechnology, College of Biomedical Sciences, Kangwon National University, Chuncheon, 200-701, Republic of Korea.
| |
Collapse
|
37
|
Wu H, Li Y, Hou Q, Zhou R, Li Z, Wu S, Yu J, Jiang M. Single‑cell intratumoral stemness analysis reveals the involvement of cell cycle and DNA damage repair in two different types of esophageal cancer. Oncol Rep 2019; 41:3201-3208. [PMID: 31002369 PMCID: PMC6489016 DOI: 10.3892/or.2019.7117] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 04/05/2019] [Indexed: 12/14/2022] Open
Abstract
Intratumoral heterogeneity, particularly the potential cancer stemness of single cancer cells, has not yet been fully elucidated in human esophageal cancer. Single‑cell transcriptome sequencing of two types of esophageal adenocarcinoma (EAC) and two types of esophageal squamous cell carcinoma (ESCC) tissues was performed, and the intratumoral cancer stemness of the types of esophageal cancer were characterized at the single‑cell level in the present study. By comparing the transcriptomic profiles of single cancer cells with high and low stemness in individual patients, it was revealed that the overexpression of cell cycle‑associated genes in EAC cells was highly correlated with stemness, whereas overexpression of genes involved in the signaling pathways of DNA replication and DNA damage repair was significantly correlated with stemness in ESCC. High expression of these stemness‑associated genes was correlated with poor prognosis of patients. Additionally, poly [ADP‑ribose] polymerase(PARP)4 was identified as a novel cancer stemness‑associated gene in ESCC and its association with survival was validated in a cohort of 121 patients with ESCC. These findings have profound potential implications for the use of cell cycle inhibitors in EAC and PARP inhibitors in ESCC, which may provide novel mechanistic insights into the plasticity of esophageal cancer.
Collapse
Affiliation(s)
- Hongjin Wu
- Cancer Research Institute, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 320000, P.R. China
| | - Ying Li
- Cancer Research Institute, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 320000, P.R. China
| | - Qiang Hou
- Cancer Research Institute, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 320000, P.R. China
| | - Rongjin Zhou
- Cancer Research Institute, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 320000, P.R. China
| | - Ziwei Li
- Cancer Research Institute, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 320000, P.R. China
| | - Shixiu Wu
- Cancer Research Institute, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 320000, P.R. China
| | - Juehua Yu
- Yunnan Institute of Digestive Disease, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Mingfeng Jiang
- Cancer Research Institute, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 320000, P.R. China
| |
Collapse
|
38
|
BCL-2 family isoforms in apoptosis and cancer. Cell Death Dis 2019; 10:177. [PMID: 30792387 PMCID: PMC6384907 DOI: 10.1038/s41419-019-1407-6] [Citation(s) in RCA: 453] [Impact Index Per Article: 75.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Revised: 12/17/2018] [Accepted: 01/29/2019] [Indexed: 12/17/2022]
Abstract
The BCl-2 family has long been identified for its role in apoptosis. Following the initial discovery of BCL-2 in the context of B-cell lymphoma in the 1980s, a number of homologous proteins have since been identified. The members of the Bcl-2 family are designated as such due to their BCL-2 homology (BH) domains and involvement in apoptosis regulation. The BH domains facilitate the family members’ interactions with each other and can indicate pro- or anti-apoptotic function. Traditionally, these proteins are categorised into one of the three subfamilies; anti-apoptotic, BH3-only (pro-apoptotic), and pore-forming or ‘executioner’ (pro-apoptotic) proteins. Each of the BH3-only or anti-apoptotic proteins has a distinct pattern of activation, localisation and response to cell death or survival stimuli. All of these can vary across cell or stress types, or developmental stage, and this can cause the delineation of the roles of BCL-2 family members. Added to this complexity is the presence of relatively uncharacterised isoforms of many of the BCL-2 family members. There is a gap in our knowledge regarding the function of BCL-2 family isoforms. BH domain status is not always predictive or indicative of protein function, and several other important sequences, which can contribute to apoptotic activity have been identified. While therapeutic strategies targeting the BCL-2 family are constantly under development, it is imperative that we understand the molecules, which we are attempting to target. This review, discusses our current knowledge of anti-apoptotic BCL-2 family isoforms. With significant improvements in the potential for splicing therapies, it is important that we begin to understand the distinctions of the BCL-2 family, not limited to just the mechanisms of apoptosis control, but in their roles outside of apoptosis.
Collapse
|
39
|
Surowy HM, Giesen AK, Otte J, Büttner R, Falkenstein D, Friedl H, Meier F, Petzsch P, Wachtmeister T, Westphal D, Wieczorek D, Wruck W, Adjaye J, Rütten A, Redler S. Gene expression profiling in aggressive digital papillary adenocarcinoma sheds light on the architecture of a rare sweat gland carcinoma. Br J Dermatol 2019; 180:1150-1160. [PMID: 30472730 DOI: 10.1111/bjd.17446] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2018] [Indexed: 12/26/2022]
Abstract
BACKGROUND Sweat gland carcinomas are rare cutaneous adnexal malignancies. Aggressive digital papillary adenocarcinoma (ADPA) represents a very rare subentity, thought to arise almost exclusively from the sweat glands of the fingers and toes. The aetiology of sweat gland carcinomas and ADPA is largely unknown. ADPAs are most likely driven by somatic mutations. However, somatic mutation patterns are largely unexplored, creating barriers to the development of effective therapeutic approaches to the treatment of ADPA. OBJECTIVES To investigate the transcriptome profile of ADPA using a sample of eight formalin-fixed, paraffin-embedded tissue samples of ADPA and healthy control tissue. METHODS Transcriptome profiling was performed using the Affymetrix PrimeView Human Gene Expression Microarray and findings were validated via reverse transcription of RNA and real-time quantitative polymerase chain reaction. RESULTS Transcriptome analyses showed increased tumour expression of 2266 genes, with significant involvement of cell cycle, ribosomal and crucial cancer pathways. Our results point to tumour overexpression of FGFR2 (P = 0·001). CONCLUSIONS The results indicate the involvement of crucial oncogenic driver pathways, highlighting cell cycle and ribosomal pathways in the aetiology of ADPA. Suggested tumour overexpression of FGFR2 raises the hope that targeting the fibroblast growth factor (FGF)/FGF receptor axis might be a promising treatment for ADPA and probably for the overall group of sweat gland carcinomas.
Collapse
Affiliation(s)
- H M Surowy
- Heinrich-Heine-University, Medical Faculty, Institute of Human Genetics, Düsseldorf, Germany
| | - A K Giesen
- Heinrich-Heine-University, Medical Faculty, Institute of Human Genetics, Düsseldorf, Germany
| | - J Otte
- Institute for Stem Cell Research and Regenerative Medicine, Düsseldorf, Germany
| | - R Büttner
- Pathology, Institute for Pathology, University Hospital Cologne, Cologne, Germany
| | - D Falkenstein
- Heinrich-Heine-University, Medical Faculty, Institute of Human Genetics, Düsseldorf, Germany
| | - H Friedl
- Klinikum Darmstadt, Department of Dermatology, Darmstadt, Germany
| | - F Meier
- Department of Dermatology, Carl Gustav Carus Medical Center, TU Dresden, Dresden, Germany.,National Center for Tumour Diseases (NCT), Partner Site Dresden, Dresden, Germany
| | - P Petzsch
- Biological and Medical Research Center (BMFZ), Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - T Wachtmeister
- Biological and Medical Research Center (BMFZ), Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - D Westphal
- Department of Dermatology, Carl Gustav Carus Medical Center, TU Dresden, Dresden, Germany.,National Center for Tumour Diseases (NCT), Partner Site Dresden, Dresden, Germany
| | - D Wieczorek
- Heinrich-Heine-University, Medical Faculty, Institute of Human Genetics, Düsseldorf, Germany
| | - W Wruck
- Institute for Stem Cell Research and Regenerative Medicine, Düsseldorf, Germany
| | - J Adjaye
- Institute for Stem Cell Research and Regenerative Medicine, Düsseldorf, Germany
| | - A Rütten
- Dermatopathology, Bodensee, Siemensstrasse 6/1, 88048, Friedrichshafen, Germany
| | - S Redler
- Heinrich-Heine-University, Medical Faculty, Institute of Human Genetics, Düsseldorf, Germany
| |
Collapse
|
40
|
Louault K, Bonneaud TL, Séveno C, Gomez-Bougie P, Nguyen F, Gautier F, Bourgeois N, Loussouarn D, Kerdraon O, Barillé-Nion S, Jézéquel P, Campone M, Amiot M, Juin PP, Souazé F. Interactions between cancer-associated fibroblasts and tumor cells promote MCL-1 dependency in estrogen receptor-positive breast cancers. Oncogene 2019; 38:3261-3273. [PMID: 30631150 PMCID: PMC6756023 DOI: 10.1038/s41388-018-0635-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 11/22/2018] [Accepted: 11/23/2018] [Indexed: 12/24/2022]
Abstract
Selective inhibition of BCL-2 is expected to enhance therapeutic vulnerability in luminal estrogen receptor-positive breast cancers. We show here that the BCL-2 dependency of luminal tumor cells is nevertheless mitigated by breast cancer-associated fibroblasts (bCAFs) in a manner that defines MCL-1 as another critical therapeutic target. bCAFs favor MCL-1 expression and apoptotic resistance in luminal cancer cells in a IL-6 dependent manner while their own, robust, survival also relies on MCL-1. Studies based on ex vivo cultures of human luminal breast cancer tissues further argue that the contribution of stroma-derived signals to MCL-1 expression shapes BCL-2 dependency. Thus, MCL-1 inhibitors are beneficial for targeted apoptosis of breast tumor ecosystems, even in a subtype where MCL-1 dependency is not intrinsically driven by oncogenic pathways.
Collapse
Affiliation(s)
- K Louault
- CRCINA, Team 8, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,SIRIC ILIAD, Angers, Nantes, France
| | - T L Bonneaud
- CRCINA, Team 8, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,SIRIC ILIAD, Angers, Nantes, France
| | - C Séveno
- CRCINA, Team 8, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,SIRIC ILIAD, Angers, Nantes, France
| | - P Gomez-Bougie
- SIRIC ILIAD, Angers, Nantes, France.,CRCINA, Team 10, INSERM, Université d'Angers, Université de Nantes, Nantes, France
| | - F Nguyen
- CRCINA, Team 8, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,ONIRIS, Nantes Atlantic College of Veterinary Medicine Food Science and Engineering, Animal Cancers, Nantes, France
| | - F Gautier
- CRCINA, Team 8, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,SIRIC ILIAD, Angers, Nantes, France.,ICO René Gauducheau, Saint Herblain, France
| | - N Bourgeois
- CRCINA, Team 8, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,SIRIC ILIAD, Angers, Nantes, France
| | - D Loussouarn
- Service d'Anatomie Pathologique, CHU Nantes, Nantes, France
| | - O Kerdraon
- SIRIC ILIAD, Angers, Nantes, France.,ICO René Gauducheau, Saint Herblain, France
| | - S Barillé-Nion
- CRCINA, Team 8, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,SIRIC ILIAD, Angers, Nantes, France
| | - P Jézéquel
- CRCINA, Team 8, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,SIRIC ILIAD, Angers, Nantes, France.,ICO René Gauducheau, Saint Herblain, France
| | - M Campone
- CRCINA, Team 8, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,SIRIC ILIAD, Angers, Nantes, France.,ICO René Gauducheau, Saint Herblain, France
| | - M Amiot
- SIRIC ILIAD, Angers, Nantes, France.,CRCINA, Team 10, INSERM, Université d'Angers, Université de Nantes, Nantes, France
| | - P P Juin
- CRCINA, Team 8, INSERM, Université d'Angers, Université de Nantes, Nantes, France. .,SIRIC ILIAD, Angers, Nantes, France. .,ICO René Gauducheau, Saint Herblain, France. .,CNRS GDR3697 Micronit, Tours, France.
| | - F Souazé
- CRCINA, Team 8, INSERM, Université d'Angers, Université de Nantes, Nantes, France. .,SIRIC ILIAD, Angers, Nantes, France. .,CNRS GDR3697 Micronit, Tours, France.
| |
Collapse
|
41
|
Pharmacological targeting of mitochondria in cancer stem cells: An ancient organelle at the crossroad of novel anti-cancer therapies. Pharmacol Res 2019; 139:298-313. [DOI: 10.1016/j.phrs.2018.11.020] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 11/13/2018] [Accepted: 11/13/2018] [Indexed: 02/07/2023]
|