1
|
Brockmueller A, Buhrmann C, Moravejolahkami AR, Shakibaei M. Resveratrol and p53: How are they involved in CRC plasticity and apoptosis? J Adv Res 2024; 66:181-195. [PMID: 38190940 PMCID: PMC11674784 DOI: 10.1016/j.jare.2024.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/27/2023] [Accepted: 01/05/2024] [Indexed: 01/10/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC), which is mainly caused by epigenetic and lifestyle factors, is very often associated with functional plasticity during its development. In addition, the malignant plasticity of CRC cells underscores one of their survival abilities to functionally adapt to specific stresses, including inflammation, that occur during carcinogenesis. This leads to the generation of various subsets of cancer cells with phenotypic diversity and promotes epithelial-mesenchymal transition (EMT), formation of cancer cell stem cells (CSCs) and metabolic reprogramming. This can enhance cancer cell differentiation and facilitate tumorigenic potential, drug resistance and metastasis. AIM OF REVIEW The tumor protein p53 acts as one of the central suppressors of carcinogenesis by regulating its target genes, whose proteins are involved in the plasticity of cancer cells, autophagy, cell cycle, apoptosis, DNA repair. The aim of this review is to summarize the latest published research on resveratrol's effect in the prevention of CRC, its regulatory actions, specifically on the p53 pathway, and its treatment options. KEY SCIENTIFIC CONCEPTS OF REVIEW Resveratrol, a naturally occurring polyphenol, is a potent inducer of a variety of tumor-controlling. However, the underlying mechanisms linking the p53 signaling pathway to the functional anti-plasticity effect of resveratrol in CRC are still poorly understood. Therefore, this review discusses novel relationships between anti-cellular plasticity/heterogeneity, pro-apoptosis and modulation of tumor protein p53 signaling in CRC oncogenesis, as one of the crucial mechanisms by which resveratrol prevents malignant phenotypic changes leading to cell migration and drug resistance, thus improving the ongoing treatment of CRC.
Collapse
Affiliation(s)
- Aranka Brockmueller
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-University Munich, Pettenkoferstr. 11, D-80336 Munich, Germany
| | - Constanze Buhrmann
- Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Amir Reza Moravejolahkami
- Department of Clinical Nutrition, School of Nutrition & Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mehdi Shakibaei
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilians-University Munich, Pettenkoferstr. 11, D-80336 Munich, Germany.
| |
Collapse
|
2
|
Mondal A, Das B, Karmakar S, Pani S, Khan S, Gupta P, Das Sarma J. Modulatory Role of Pantropic Cell Signaling Pathways in the Antimigratory and Antiproliferative Action of Triazole Chelated Iridium(III) Complexes in Cervical Cancer Cells. J Med Chem 2024; 67:20559-20570. [PMID: 39527836 DOI: 10.1021/acs.jmedchem.4c01997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
In the current study, the antimigratory and antiproliferative effect of three substituted triazole-chelated iridium(III) complexes Ir-TRN, Ir-TRH, and Ir-TRF were studied with special emphasis on modulation of P53 activity, a cell cycle regulator. ERK2/MAPK, another crucial cell signaling pathway protein, was also shown to play a crucial role in cell migration and proliferation. The complexes increase the ROS generation within the cell, further supporting apoptotic induction by exerting cellular oxidative stress. These metal complexes also affect ER stress by altering ERp29, an ER-resident chaperone, further inducing the process of apoptosis. The iridium(III) complexes restrict cervical cancer cell migration and proliferation by exerting pronounced effects as P53 activators and downregulation of ERK2/MAPK activity in cervical cancer cells. The underpinning mechanism of P53 and ERK2/MAPK activity in cervical cancer cells in the presence of iridium(III) complexes was studied in detail in this study, which paves the way for developing promising avenues for cancer therapeutics.
Collapse
Affiliation(s)
- Anushka Mondal
- Department of Biological Sciences, Indian Institute of Science Education and Research-Kolkata, Mohanpur, West Bengal 741246, India
| | - Bishnu Das
- Department of Chemical Sciences, Indian Institute of Science Education and Research-Kolkata, Mohanpur, West Bengal 741246, India
| | - Souvik Karmakar
- Department of Biological Sciences, Indian Institute of Science Education and Research-Kolkata, Mohanpur, West Bengal 741246, India
| | - Soumili Pani
- Department of Biological Sciences, Indian Institute of Science Education and Research-Kolkata, Mohanpur, West Bengal 741246, India
- Department of Chemical Sciences, Indian Institute of Science Education and Research-Kolkata, Mohanpur, West Bengal 741246, India
| | - Shrabani Khan
- Department of Chemical Sciences, Indian Institute of Science Education and Research-Kolkata, Mohanpur, West Bengal 741246, India
| | - Parna Gupta
- Department of Chemical Sciences, Indian Institute of Science Education and Research-Kolkata, Mohanpur, West Bengal 741246, India
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research-Kolkata, Mohanpur, West Bengal 741246, India
| |
Collapse
|
3
|
Garcia C, Miller-Awe MD, Witkowski MT. Concepts in B cell acute lymphoblastic leukemia pathogenesis. J Leukoc Biol 2024; 116:18-32. [PMID: 38243586 PMCID: PMC11869204 DOI: 10.1093/jleuko/qiae015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/22/2023] [Accepted: 01/08/2024] [Indexed: 01/21/2024] Open
Abstract
B cell acute lymphoblastic leukemia (B-ALL) arises from genetic alterations impacting B cell progenitors, ultimately leading to clinically overt disease. Extensive collaborative efforts in basic and clinical research have significantly improved patient prognoses. Nevertheless, a subset of patients demonstrate resistance to conventional chemotherapeutic approaches and emerging immunotherapeutic interventions. This review highlights the mechanistic underpinnings governing B-ALL transformation. Beginning with exploring normative B cell lymphopoiesis, we delineate the influence of recurrent germline and somatic genetic aberrations on the perturbation of B cell progenitor differentiation and protumorigenic signaling, thereby facilitating the neoplastic transformation underlying B-ALL progression. Additionally, we highlight recent advances in the multifaceted landscape of B-ALL, encompassing metabolic reprogramming, microbiome influences, inflammation, and the discernible impact of socioeconomic and racial disparities on B-ALL transformation and patient survival.
Collapse
Affiliation(s)
- Clarissa Garcia
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, United States
| | - Megan D. Miller-Awe
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, United States
| | - Matthew T. Witkowski
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, United States
| |
Collapse
|
4
|
Chen C, Demirkhanyan L, Gondi CS. The Multifaceted Role of miR-21 in Pancreatic Cancers. Cells 2024; 13:948. [PMID: 38891080 PMCID: PMC11172074 DOI: 10.3390/cells13110948] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/23/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
With the lack of specific signs and symptoms, pancreatic ductal adenocarcinoma (PDAC) is often diagnosed at late metastatic stages, resulting in poor survival outcomes. Among various biomarkers, microRNA-21 (miR-21), a small non-coding RNA, is highly expressed in PDAC. By inhibiting regulatory proteins at the 3' untranslated regions (UTR), miR-21 holds significant roles in PDAC cell proliferation, epithelial-mesenchymal transition, angiogenesis, as well as cancer invasion, metastasis, and resistance therapy. We conducted a systematic search across major databases for articles on miR-21 and pancreatic cancer mainly published within the last decade, focusing on their diagnostic, prognostic, therapeutic, and biological roles. This rigorous approach ensured a comprehensive review of miR-21's multifaceted role in pancreatic cancers. In this review, we explore the current understandings and future directions regarding the regulation, diagnostic, prognostic, and therapeutic potential of targeting miR-21 in PDAC. This exhaustive review discusses the involvement of miR-21 in proliferation, epithelial-mesenchymal transition (EMT), apoptosis modulation, angiogenesis, and its role in therapy resistance. Also discussed in the review is the interplay between various molecular pathways that contribute to tumor progression, with specific reference to pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Clare Chen
- Department of Internal Medicine, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
| | - Lusine Demirkhanyan
- Department of Internal Medicine, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
- Departments of Internal Medicine and Surgery, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
| | - Christopher S. Gondi
- Department of Internal Medicine, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
- Departments of Internal Medicine and Surgery, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
- Departments of Internal Medicine, Surgery, and Health Science Education and Pathology, University of Illinois College of Medicine Peoria, Peoria, IL 61605, USA
- Health Care Engineering Systems Center, The Grainger College of Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
5
|
Chasov V, Davletshin D, Gilyazova E, Mirgayazova R, Kudriaeva A, Khadiullina R, Yuan Y, Bulatov E. Anticancer therapeutic strategies for targeting mutant p53-Y220C. J Biomed Res 2024; 38:222-232. [PMID: 38738269 PMCID: PMC11144932 DOI: 10.7555/jbr.37.20230093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 09/25/2023] [Accepted: 10/07/2023] [Indexed: 05/14/2024] Open
Abstract
The tumor suppressor p53 is a transcription factor with a powerful antitumor activity that is controlled by its negative regulator murine double minute 2 (MDM2, also termed HDM2 in humans) through a feedback mechanism. At the same time, TP53 is the most frequently mutated gene in human cancers. Mutant p53 proteins lose wild-type p53 tumor suppression functions but acquire new oncogenic properties, among which are deregulating cell proliferation, increasing chemoresistance, disrupting tissue architecture, and promoting migration, invasion and metastasis as well as several other pro-oncogenic activities. The oncogenic p53 mutation Y220C creates an extended surface crevice in the DNA-binding domain destabilizing p53 and causing its denaturation and aggregation. This cavity accommodates stabilizing small molecules that have therapeutic values. The development of suitable small-molecule stabilizers is one of the therapeutic strategies for reactivating the Y220C mutant protein. In this review, we summarize approaches that target p53-Y220C, including reactivating this mutation with small molecules that bind Y220C to the hydrophobic pocket and developing immunotherapies as the goal for the near future, which target tumor cells that express the p53-Y220C neoantigen.
Collapse
Affiliation(s)
- Vitaly Chasov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia
| | - Damir Davletshin
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia
| | - Elvina Gilyazova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia
| | - Regina Mirgayazova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia
| | - Anna Kudriaeva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| | - Raniya Khadiullina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia
| | - Youyong Yuan
- Institute of Life Sciences, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Emil Bulatov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| |
Collapse
|
6
|
Göttig L, Jummer S, Staehler L, Groitl P, Karimi M, Blanchette P, Kosulin K, Branton PE, Schreiner S. The human adenovirus PI3K-Akt activator E4orf1 is targeted by the tumor suppressor p53. J Virol 2024; 98:e0170123. [PMID: 38451084 PMCID: PMC11019960 DOI: 10.1128/jvi.01701-23] [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: 11/20/2023] [Accepted: 01/13/2024] [Indexed: 03/08/2024] Open
Abstract
Human adenoviruses (HAdV) are classified as DNA tumor viruses due to their potential to mediate oncogenic transformation in non-permissive mammalian cells and certain human stem cells. To achieve transformation, the viral early proteins of the E1 and E4 regions must block apoptosis and activate proliferation: the former predominantly through modulating the cellular tumor suppressor p53 and the latter by activating cellular pro-survival and pro-metabolism protein cascades, such as the phosphoinositide 3-kinase (PI3K-Akt) pathway, which is activated by HAdV E4orf1. Focusing on HAdV-C5, we show that E4orf1 is necessary and sufficient to stimulate Akt activation through phosphorylation in H1299 cells, which is not only hindered but repressed during HAdV-C5 infection with a loss of E4orf1 function in p53-positive A549 cells. Contrary to other research, E4orf1 localized not only in the common, cytoplasmic PI3K-Akt-containing compartment, but also in distinct nuclear aggregates. We identified a novel inhibitory mechanism, where p53 selectively targeted E4orf1 to destabilize it, also stalling E4orf1-dependent Akt phosphorylation. Co-IP and immunofluorescence studies showed that p53 and E4orf1 interact, and since p53 is bound by the HAdV-C5 E3 ubiquitin ligase complex, we also identified E4orf1 as a novel factor interacting with E1B-55K and E4orf6 during infection; overexpression of E4orf1 led to less-efficient E3 ubiquitin ligase-mediated proteasomal degradation of p53. We hypothesize that p53 specifically subverts the pro-survival function of E4orf1-mediated PI3K-Akt activation to protect the cell from metabolic hyper-activation or even transformation.IMPORTANCEHuman adenoviruses (HAdV) are nearly ubiquitous pathogens comprising numerous subtypes that infect various tissues and organs. Among many encoded proteins that facilitate viral replication and subversion of host cellular processes, the viral E4orf1 protein has emerged as an intriguing yet under-investigated player in the complex interplay between the virus and its host. Nonetheless, E4orf1 has gained attention as a metabolism activator and oncogenic agent, while recent research is showing that E4orf1 may play a more important role in modulating the cellular pathways such as phosphoinositide 3-kinase-Akt-mTOR. Our study reveals a novel and general impact of E4orf1 on host mechanisms, providing a novel basis for innovative antiviral strategies in future therapeutic settings. Ongoing investigations of the cellular pathways modulated by HAdV are of great interest, particularly since adenovirus-based vectors actually serve as vaccine or gene vectors. HAdV constitute an ideal model system to analyze the underlying molecular principles of virus-induced tumorigenesis.
Collapse
Affiliation(s)
- Lilian Göttig
- Institute of Virology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Simone Jummer
- Institute of Virology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Luisa Staehler
- Institute of Virology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Peter Groitl
- Institute of Virology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Maryam Karimi
- Institute of Virology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Paola Blanchette
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
- Goodman Cancer Research Center, McGill University, Montreal, Quebec, Canada
| | - Karin Kosulin
- Molecular Microbiology, Children’s Cancer Research Institute, Vienna, Austria
| | - Philip E. Branton
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
- Goodman Cancer Research Center, McGill University, Montreal, Quebec, Canada
| | - Sabrina Schreiner
- Institute of Virology, School of Medicine, Technical University of Munich, Munich, Germany
- Institute of Virology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (Resolving Infection Susceptibility; EXC 2155), Freiburg, Germany
- Institute of Virology, Medical Center—University of Freiburg, Freiburg, Germany
| |
Collapse
|
7
|
Darvish L, Bahreyni Toossi MT, Azimian H, Shakeri M, Dolat E, Ahmadizad Firouzjaei A, Rezaie S, Amraee A, Aghaee-Bakhtiari SH. The role of microRNA-induced apoptosis in diverse radioresistant cancers. Cell Signal 2023; 104:110580. [PMID: 36581218 DOI: 10.1016/j.cellsig.2022.110580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 12/07/2022] [Accepted: 12/21/2022] [Indexed: 12/27/2022]
Abstract
Resistance to cancer radiotherapy is one of the biggest concerns for success in treating and preventing recurrent disease. Malignant tumors may develop when they block genetic mutations associated with apoptosis or abnormal expression of apoptosis; Tumor treatment may induce the expression of apoptosis-related genes to promote tumor cell apoptosis. MicroRNAs have been shown to contribute to forecasting prognosis, distinguishing between cancer subtypes, and affecting treatment outcomes in cancer. Constraining these miRNAs may be an attractive treatment strategy to help overcome radiation resistance. The delivery of these future treatments is still challenging due to the excess downstream targets that each miRNA can control. Understanding the role of miRNAs brings us one step closer to attaining patient treatment and improving patient outcomes. This review summarized the current information on the role of microRNA-induced apoptosis in determining the radiosensitivity of various cancers.
Collapse
Affiliation(s)
- Leili Darvish
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Hosein Azimian
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Physics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahsa Shakeri
- Department of Medical Physics and Biomedical Engineering, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Research Center for Molecular and Cellular Imaging, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Dolat
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Ahmadizad Firouzjaei
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samaneh Rezaie
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Azadeh Amraee
- Department of Medical Physics, Faculty of Medicine, School of Medicine, Lorestan University of Medical Sciences, khorramabad, Iran
| | - Seyed Hamid Aghaee-Bakhtiari
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Bioinformatics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
8
|
Luo J, Zhao H, Chen L, Liu M. Multifaceted functions of RPS27a: An unconventional ribosomal protein. J Cell Physiol 2023; 238:485-497. [PMID: 36580426 DOI: 10.1002/jcp.30941] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/28/2022] [Accepted: 12/21/2022] [Indexed: 12/30/2022]
Abstract
The ribosomal protein S27a (RPS27a) is cleaved from the fusion protein ubiquitin-RPS27a (Ub-RPS27a). Generally, Ub and RPS27a are coexpressed as a fusion protein but function independently after Ub is cleaved from RPS27a by a deubiquitinating enzyme. As an RP, RPS27a assembles into ribosomes, but it also functions independently of ribosomes. RPS27a is involved in the development and poor prognosis of various cancers, such as colorectal cancer, liver cancer, chronic myeloid leukemia, and renal carcinoma, and is associated with poor prognosis. Notably, the murine double minute 2/P53 axis is a major pathway through which RPS27a regulates cancer development. Moreover, RPS27a maintains sperm motility, regulates winged aphid indirect flight muscle degeneration, and facilitates plant growth. Additionally, RPS27a is a metalloprotein and mercury (Hg) biomarker. In the present review, we described the origin, structure, and biological functions of RPS27a.
Collapse
Affiliation(s)
- Jingshun Luo
- Key Laboratory of Cardiovascular Diseases of Yunnan Province, Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Central laboratory of Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Hong Zhao
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Nursing College, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Linxi Chen
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Meiqing Liu
- Key Laboratory of Cardiovascular Diseases of Yunnan Province, Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Central laboratory of Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
9
|
Mahmoud GA, Ali HE, Radwan RR. Design of pH-responsive polymeric nanocarrier for targeted delivery of pyrogallol with enhanced antitumor potential in colon cancer. Arch Biochem Biophys 2022; 731:109431. [DOI: 10.1016/j.abb.2022.109431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 10/04/2022] [Accepted: 10/08/2022] [Indexed: 11/25/2022]
|
10
|
Brown DW, Beatty PH, Lewis JD. Molecular Targeting of the Most Functionally Complex Gene in Precision Oncology: p53. Cancers (Basel) 2022; 14:5176. [PMID: 36358595 PMCID: PMC9654076 DOI: 10.3390/cancers14215176] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/16/2022] [Accepted: 10/20/2022] [Indexed: 09/29/2023] Open
Abstract
While chemotherapy is a key treatment strategy for many solid tumors, it is rarely curative, and most tumor cells eventually become resistant. Because of this, there is an unmet need to develop systemic treatments that capitalize on the unique mutational landscape of each patient's tumor. The most frequently mutated protein in cancer, p53, has a role in nearly all cancer subtypes and tumorigenesis stages and therefore is one of the most promising molecular targets for cancer treatment. Unfortunately, drugs targeting p53 have seen little clinical success despite promising preclinical data. Most of these drug compounds target specific aspects of p53 inactivation, such as through inhibiting negative regulation by the mouse double minute (MDM) family of proteins. These treatment strategies fail to address cancer cells' adaptation mechanisms and ignore the impact that p53 loss has on the entire p53 network. However, recent gene therapy successes show that targeting the p53 network and cellular dysfunction caused by p53 inactivation is now possible and may soon translate into successful clinical responses. In this review, we discuss p53 signaling complexities in cancer that have hindered the development and use of p53-targeted drugs. We also describe several current therapeutics reporting promising preclinical and clinical results.
Collapse
Affiliation(s)
- Douglas W. Brown
- Department of Oncology, University of Alberta, Edmonton, AB T6G 2E1, Canada
- Entos Pharmaceuticals, Unit 4550, 10230 Jasper Avenue, Edmonton, AB T5J 4P6, Canada
| | - Perrin H. Beatty
- Entos Pharmaceuticals, Unit 4550, 10230 Jasper Avenue, Edmonton, AB T5J 4P6, Canada
| | - John D. Lewis
- Department of Oncology, University of Alberta, Edmonton, AB T6G 2E1, Canada
- Entos Pharmaceuticals, Unit 4550, 10230 Jasper Avenue, Edmonton, AB T5J 4P6, Canada
| |
Collapse
|
11
|
Manturthi S, Bhattacharya D, Sakhare KR, Narayan KP, Patri SV. Cimetidine-Based Cationic Amphiphiles for In Vitro Gene Delivery Targetable to Colon Cancer. ACS OMEGA 2022; 7:31388-31402. [PMID: 36092589 PMCID: PMC9453813 DOI: 10.1021/acsomega.2c03777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/10/2022] [Indexed: 06/15/2023]
Abstract
Cimetidine, a histamine-2 (H2) receptor antagonist, has been found to have anticancer properties against a number of cancer-type cells. In this report, we have demonstrated that cimetidine can acts as a hydrophilic domain in cationic lipids and targetable to the gastric system by carrying reporter genes and therapeutic genes through in vitro transfection. Two lipids, namely, Toc-Cim and Chol-Cim consisting cimetidine as the main head group and hydrophobic moieties as alpha-tocopherol or cholesterol, respectively, were designed and synthesized. 1,2-Dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) is a well-known co-lipid employed to produce liposomes as uniform vesicles. The liposomes and lipoplexes were structurally and functionally evaluated for global surface charges and hydrodynamic diameters, and results found that both liposome and lipoplex size and surface charges are optimal to screen the transfection potentials. DNA-binding studies were analyzed as complete binding at all formulated N/P ratios. The liposomes and lipoplexes of both the lipids Toc-Cim and Chol-Cim show minimal cytotoxicity even though at higher concentrations. The results of the transfection experiments revealed that tocopherol-based cationic lipids (Toc-Cim) show finer transfection efficacy with optimized N/P ratios (2:1 and 4:1) in the colon cancer cell line. Toc-Cim lipoplexes show higher cellular uptake compare to Chol-Cim in the colon cancer cell line at 2:1 and 4:1 N/P ratios. Toc-Cim and Chol-Cim lipids showed highly compatible serum, examined up to 50% of the serum concentration. To evaluate the apoptotic cell death in CT-26 cells, exposed to Toc-Cim:p53 and Chol-Cim:p53 lipoplexes at 2:1 N/P ratios, superior results showed with Toc-Cim:p53. An effect of TP53 protein expression in CT-26 cell lines assayed by western blot, transfected with Toc-Cim:p53 and Chol-Cim:p53 lipoplexes, demonstrated the superior efficacy of Toc-Cim. All of the findings suggest that Toc-Cim lipid is relatively secure and is an effective transfection agent to colon cancer gene delivery.
Collapse
Affiliation(s)
- Shireesha Manturthi
- Department
of Chemistry, National Institute of Technology
Warangal, Hanamkonda, Telangana 506004, India
| | - Dwaipayan Bhattacharya
- Department
of Biological Science, Bits Pilani-Hyderabad, Hyderabad, Telangana 500078, India
| | - Kalyani Rajesh Sakhare
- Department
of Biological Science, Bits Pilani-Hyderabad, Hyderabad, Telangana 500078, India
| | - Kumar Pranav Narayan
- Department
of Biological Science, Bits Pilani-Hyderabad, Hyderabad, Telangana 500078, India
| | - Srilakshmi V. Patri
- Department
of Chemistry, National Institute of Technology
Warangal, Hanamkonda, Telangana 506004, India
| |
Collapse
|
12
|
Ibrahim EH, Alshahrani MY, Ghramh HA, Alothaid H, Kilany M, Morsy K, El-kott AF, Taha R, El-Mekkawy HI, EL-Shaboury GA, El-Mansi AA, Mohammed ME, Sayed MA, Yahia IS. Origanum majorana harvested from Al-Soda, Saudi Arabia promotes mitotic arrest and apoptosis in colon cancer cells. JOURNAL OF KING SAUD UNIVERSITY - SCIENCE 2022; 34:101878. [DOI: 10.1016/j.jksus.2022.101878] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2025]
|
13
|
MicroRNA-101-3p Suppresses Cancer Cell Growth by Inhibiting the USP47-Induced Deubiquitination of RPL11. Cancers (Basel) 2022; 14:cancers14040964. [PMID: 35205710 PMCID: PMC8870143 DOI: 10.3390/cancers14040964] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/11/2022] [Accepted: 02/13/2022] [Indexed: 12/04/2022] Open
Abstract
Simple Summary An abnormal expression of microRNA is commonly observed in cancer. Since a single miRNA can target numerous genes, it is important to understand the exact mechanism for the regulation of cancer growth by miRNAs. Here, we show that miR-101-3p, which is downregulated in several cancers, regulates RPL11 ubiquitination by targeting USP47, thereby controlling p53 levels by affecting the localization of RPL11 and its interaction with MDM2. Our results provide a novel mechanism for the inhibition of cancer cell growth by miR-101-3p, and suggest that miR-101-3p could be a potential target as an anticancer agent. Abstract MicroRNAs (miRNAs) are a class of small non-coding RNA molecules that regulate a countless number of genes in the cell, and the aberrant expression of miRNA can lead to cancer. Here, we demonstrate that miR-101-3p regulates the RPL11–MDM2–p53 pathway by targeting ubiquitin-specific peptidase 47 (USP47), consequently inhibiting cancer cell proliferation. We confirm that miR-101-3p directly binds to the 3′-UTR region of the USP47 gene and inhibits USP47 expression. In addition, the overexpression of miR-101-3p suppresses cell proliferation in a p53-dependent manner. MiR-101-3p promotes interaction between RPL11 and MDM2 by inducing the translocation of RPL11 from the nucleolus to the nucleoplasm, thus preventing the MDM2-mediated proteasomal degradation of p53. However, these phenomena are restored by the overexpression of USP47, but not by its catalytically inactive form. Indeed, miR-101-3p regulates RPL11 localization and its interaction with MDM2 by inhibiting the USP47-induced deubiquitination of RPL11. Finally, the expression of miR-101-3p is downregulated in lung cancer patients, and the patients with low miR-101-3p expression exhibit a lower survival rate, indicating that miR-101-3p is associated with tumorigenesis. Together, our findings suggest that miR-101-3p functions as a tumor suppressor by targeting USP47 and could be a potential therapeutic target for cancers.
Collapse
|
14
|
Butera G, Manfredi M, Fiore A, Brandi J, Pacchiana R, De Giorgis V, Barberis E, Vanella V, Galasso M, Scupoli MT, Marengo E, Cecconi D, Donadelli M. Tumor Suppressor Role of Wild-Type P53-Dependent Secretome and Its Proteomic Identification in PDAC. Biomolecules 2022; 12:305. [PMID: 35204804 PMCID: PMC8869417 DOI: 10.3390/biom12020305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/03/2022] [Accepted: 02/09/2022] [Indexed: 12/10/2022] Open
Abstract
The study of the cancer secretome is gaining even more importance in cancers such as pancreatic ductal adenocarcinoma (PDAC), whose lack of recognizable symptoms and early detection assays make this type of cancer highly lethal. The wild-type p53 protein, frequently mutated in PDAC, prevents tumorigenesis by regulating a plethora of signaling pathways. The importance of the p53 tumor suppressive activity is not only primarily involved within cells to limit tumor cell proliferation but also in the extracellular space. Thus, loss of p53 has a profound impact on the secretome composition of cancer cells and marks the transition to invasiveness. Here, we demonstrate the tumor suppressive role of wild-type p53 on cancer cell secretome, showing the anti-proliferative, apoptotic and chemosensitivity effects of wild-type p53 driven conditioned medium. By using high-resolution SWATH-MS technology, we characterized the secretomes of p53-deficient and p53-expressing PDAC cells. We found a great number of secreted proteins that have known roles in cancer-related processes, 30 of which showed enhanced and 17 reduced secretion in response to p53 silencing. These results are important to advance our understanding on the link between wt-p53 and cancer microenvironment. In conclusion, this approach may detect a secreted signature specifically driven by wild-type p53 in PDAC.
Collapse
Affiliation(s)
- Giovanna Butera
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, 37134 Verona, Italy; (G.B.); (A.F.); (R.P.); (M.G.); (M.T.S.)
| | - Marcello Manfredi
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (M.M.); (V.D.G.); (E.B.); (V.V.)
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy;
- ISALIT, Spin-off at the University of Piemonte Orientale, 28100 Novara, Italy
| | - Alessandra Fiore
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, 37134 Verona, Italy; (G.B.); (A.F.); (R.P.); (M.G.); (M.T.S.)
| | - Jessica Brandi
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (J.B.); (D.C.)
| | - Raffaella Pacchiana
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, 37134 Verona, Italy; (G.B.); (A.F.); (R.P.); (M.G.); (M.T.S.)
| | - Veronica De Giorgis
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (M.M.); (V.D.G.); (E.B.); (V.V.)
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy;
| | - Elettra Barberis
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (M.M.); (V.D.G.); (E.B.); (V.V.)
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy;
- ISALIT, Spin-off at the University of Piemonte Orientale, 28100 Novara, Italy
| | - Virginia Vanella
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (M.M.); (V.D.G.); (E.B.); (V.V.)
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy;
| | - Marilisa Galasso
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, 37134 Verona, Italy; (G.B.); (A.F.); (R.P.); (M.G.); (M.T.S.)
- Department of Medicine, Section of Hematology, University of Verona, 37134 Verona, Italy
| | - Maria Teresa Scupoli
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, 37134 Verona, Italy; (G.B.); (A.F.); (R.P.); (M.G.); (M.T.S.)
- Research Center LURM, Interdepartmental Laboratory of Medical Research, University of Verona, 37134 Verona, Italy
| | - Emilio Marengo
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, 28100 Novara, Italy;
- ISALIT, Spin-off at the University of Piemonte Orientale, 28100 Novara, Italy
- Department of Sciences and Technological Innovation, University of Piemonte Orientale, 28100 Novara, Italy
| | - Daniela Cecconi
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (J.B.); (D.C.)
| | - Massimo Donadelli
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, 37134 Verona, Italy; (G.B.); (A.F.); (R.P.); (M.G.); (M.T.S.)
| |
Collapse
|
15
|
Marques MA, de Andrade GC, Silva JL, de Oliveira GAP. Protein of a thousand faces: The tumor-suppressive and oncogenic responses of p53. Front Mol Biosci 2022; 9:944955. [PMID: 36090037 PMCID: PMC9452956 DOI: 10.3389/fmolb.2022.944955] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/18/2022] [Indexed: 12/30/2022] Open
Abstract
The p53 protein is a pleiotropic regulator working as a tumor suppressor and as an oncogene. Depending on the cellular insult and the mutational status, p53 may trigger opposing activities such as cell death or survival, senescence and cell cycle arrest or proliferative signals, antioxidant or prooxidant activation, glycolysis, or oxidative phosphorylation, among others. By augmenting or repressing specific target genes or directly interacting with cellular partners, p53 accomplishes a particular set of activities. The mechanism in which p53 is activated depends on increased stability through post-translational modifications (PTMs) and the formation of higher-order structures (HOS). The intricate cell death and metabolic p53 response are reviewed in light of gaining stability via PTM and HOS formation in health and disease.
Collapse
Affiliation(s)
- Mayra A. Marques
- *Correspondence: Mayra A. Marques, ; Guilherme A. P. de Oliveira,
| | | | | | | |
Collapse
|
16
|
Rajput PS, Khan SR, Singh P, Chawla PA. Treatment of Small Cell Lung Cancer with Lurbinectedin: A Review. Anticancer Agents Med Chem 2021; 22:812-820. [PMID: 34229593 DOI: 10.2174/1871520621666210706150057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 05/15/2021] [Accepted: 05/23/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Lurbinectedin was approved on June 15, 2020 by Food and Drug Administration with a brand name ZEPZELCA as the first systematic approved therapy for patients having Small Cell Lung Cancer (SCLC). OBJECTIVES In this review, an attempt is made to summarize different aspects of Lurbinectedin, including the pathophysiology, chemistry, chemical synthesis, mechanism of action, adverse reactions, including pharmacokinetics of lurbinectedin. Special attention is given to various reported clinical trials of lurbinectedin. METHODS A comprehensive literature search was conducted in the relevant databases like ScienceDirect, PubMed, ResearchGate and Google Scholar to identify studies. Further upon a thorough study of these reports, significant findings/data were collected and compiled under suitable headings. Important findings related to clinical trials have been tabulated. CONCLUSION Lurbinectedin is known to act by inhibiting the active transcription of encoding genes, thereby bringing about the suppression of tumour related macrophages with an impact on tumour atmosphere. Lurbinectedin has emerged as a potential drug candidate for the treatment of small cell lung cancer (SCLC).
Collapse
Affiliation(s)
- Prince Singh Rajput
- Department of Pharmaceutical Analysis, ISF College of Pharmacy, G.T. Road, Moga-142 001, Punjab, India
| | - Sharib Raza Khan
- Department of Pharmaceutical Analysis, ISF College of Pharmacy, G.T. Road, Moga-142 001, Punjab, India
| | - Preeti Singh
- Department of Pharmaceutical Analysis, ISF College of Pharmacy, G.T. Road, Moga-142 001, Punjab, India
| | - Pooja A Chawla
- Department of Pharmaceutical Analysis, ISF College of Pharmacy, G.T. Road, Moga-142 001, Punjab, India
| |
Collapse
|
17
|
Al-Shun SA, El-Senduny FF, Ismail MA, El-Sayed WM, Badria FA, Youssef MM. Anticancer activity of new cationic arylthiophenes against hepatocellular carcinoma. Life Sci 2021; 269:119028. [PMID: 33444618 DOI: 10.1016/j.lfs.2021.119028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 12/25/2020] [Accepted: 12/31/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the second most common cancer-related death in the world. No effective curative option exists for the treatment of HCC. The available drugs exhibit severe toxic effects and low therapeutic index. AIM This work aimed to examine different monocationic arylthiophene derivatives for possible use as chemotherapeutic agents against HCC. METHODS The IC50 values for the compounds were determined. The mechanism of cytotoxicity was further investigated using different methods. RESULTS Compound 2j proved to retain the highest cytotoxicity in comparison to as a positive control. The selectivity index of compound 2j revealed the safety to normal cells. Moreover, compound 2j was able to inhibit HepG2 cells´ migration and division. The anticancer effect of compound 2j was found to be partially via cell cycle arrest, activation of the tumour suppressor p53 protein, and induction of apoptosis via both intrinsic and extrinsic pathways. Compound 2j has a potential sensitization activity and significantly reduced the IC50 values for the anticancer drugs doxorubicin, cisplatin, and taxol. CONCLUSION The tested arylthiophenes showed a potent cytotoxicity against HepG2 cells and were safe to normal cells. The most active compound 2j was found to be able to inhibit cell division and migration and also to induce apoptosis. Compound 2j also proved to have a sensitization effect on standard anticancer drugs.
Collapse
Affiliation(s)
- Sara A Al-Shun
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Mansoura University, Mansoura 35516, Egypt.
| | - Fardous F El-Senduny
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Mansoura University, Mansoura 35516, Egypt.
| | - Mohamed A Ismail
- Division of Organic Chemistry, Department of Chemistry, Faculty of Science, Mansoura University, Mansoura 35516, Egypt.
| | - Wael M El-Sayed
- Department of Zoology, Faculty of Science, Ain Shams University, Abbassia 11566, Cairo, Egypt.
| | - Farid A Badria
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| | - Magdy M Youssef
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Mansoura University, Mansoura 35516, Egypt.
| |
Collapse
|
18
|
Suttisansanee U, Pitchakarn P, Ting P, Inthachat W, Thiyajai P, Rodthayoy D, Karinchai J, Chantarasuwan B, Nuchuchua O, Temviriyanukul P. Health-promoting bioactivity and in vivo genotoxicity evaluation of a hemiepiphyte fig, Ficus dubia. Food Sci Nutr 2021; 9:2269-2279. [PMID: 33841843 PMCID: PMC8020917 DOI: 10.1002/fsn3.2205] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 01/29/2021] [Accepted: 02/14/2021] [Indexed: 12/20/2022] Open
Abstract
Ficus species have been used as a typical component in food and folk medicine in Asia for centuries. However, little is known regarding the bioactivity and genotoxicity of the recently identified Ficus dubia (FD), an indigenous plant of the tropical evergreen rain forest. FD is unique from other Ficus species because of its highly sought‐after red‐brown latex. Antioxidant properties together with phenolic and flavonoid contents of FD were elucidated. Health‐promoting characteristics were examined by studying the inhibition of enzymes as a drug target for diabetes, hypertension, Alzheimer's disease, and obesity, together with anticancer ability against human colorectal adenocarcinoma, human hepatocellular carcinoma, human ovarian carcinoma, human prostate adenocarcinoma, and human lung carcinoma. Besides, FD genotoxicity was tested using the Drosophila wing spot test. Results showed that both FD root and latex exhibited antioxidant activity due to the presence of phenolics and flavonoids, specifically caffeic acid and cyanidin. The ethanolic fraction of FD root demonstrated a potent antidiabetic mechanism underlying α‐glucosidase inhibitory activity similar to acarbose. This fraction also suppressed lung and ovarian cancer growth, possibly by G1 and G2/M arrest, respectively. All tested fractions lacked mutagenicity in vivo. Results indicated that FD can be developed as novel antidiabetic compounds; however, its bioactive compounds should be further identified.
Collapse
Affiliation(s)
- Uthaiwan Suttisansanee
- Institute of Nutrition Mahidol University Salaya, Phuttamonthon Nakhon Pathom Thailand.,Food and Nutrition Academic and Research Cluster Institute of Nutrition Mahidol University Salaya, Phuttamonthon Nakhon Pathom Thailand
| | - Pornsiri Pitchakarn
- Department of Biochemistry Faculty of Medicine Chiang Mai University Meung Chiang Mai Thailand
| | - Pisamai Ting
- Institute of Nutrition Mahidol University Salaya, Phuttamonthon Nakhon Pathom Thailand
| | - Woorawee Inthachat
- Institute of Nutrition Mahidol University Salaya, Phuttamonthon Nakhon Pathom Thailand.,Food and Nutrition Academic and Research Cluster Institute of Nutrition Mahidol University Salaya, Phuttamonthon Nakhon Pathom Thailand
| | - Parunya Thiyajai
- Institute of Nutrition Mahidol University Salaya, Phuttamonthon Nakhon Pathom Thailand
| | - Daraphan Rodthayoy
- Department of Biochemistry Faculty of Medicine Chiang Mai University Meung Chiang Mai Thailand
| | - Jirarat Karinchai
- Department of Biochemistry Faculty of Medicine Chiang Mai University Meung Chiang Mai Thailand
| | | | - Onanong Nuchuchua
- National Nanotechnology Center (NANOTEC) National Science and Technology Development Agency (NSTDA) Pathum Thani Thailand
| | - Piya Temviriyanukul
- Institute of Nutrition Mahidol University Salaya, Phuttamonthon Nakhon Pathom Thailand.,Food and Nutrition Academic and Research Cluster Institute of Nutrition Mahidol University Salaya, Phuttamonthon Nakhon Pathom Thailand
| |
Collapse
|
19
|
Liu Y, Leslie PL, Zhang Y. Life and Death Decision-Making by p53 and Implications for Cancer Immunotherapy. Trends Cancer 2020; 7:226-239. [PMID: 33199193 DOI: 10.1016/j.trecan.2020.10.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 10/11/2020] [Accepted: 10/15/2020] [Indexed: 12/20/2022]
Abstract
The tumor-suppressor protein p53 is mutated in approximately half of all cancers, whereas the p53 signaling network is perturbed in almost all cancers. In response to different stress stimuli, p53 selectively activates genes to elicit a cell survival or cell death response. How p53 makes the decision between life and death remains a fascinating question and an exciting field of research. Understanding how this decision is made has major implications for improving cancer treatments, particularly in recently evolved immune checkpoint inhibition therapy. We highlight progress and challenges in understanding the mechanisms governing the p53 life and death decision-making process, and discuss how this decision is relevant to immune system regulation. Finally, we discuss how knowledge of the p53 pro-survival and pro-death decision node can be applied to optimize immune checkpoint inhibitor therapy for cancer treatment.
Collapse
Affiliation(s)
- Yong Liu
- Department of Radiation Oncology and Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7461, USA; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China.
| | - Patrick L Leslie
- Department of Radiation Oncology and Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7461, USA
| | - Yanping Zhang
- Department of Radiation Oncology and Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7461, USA.
| |
Collapse
|
20
|
Role of Bcl-2 Family Proteins in Photodynamic Therapy Mediated Cell Survival and Regulation. Molecules 2020; 25:molecules25225308. [PMID: 33203053 PMCID: PMC7696921 DOI: 10.3390/molecules25225308] [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: 10/05/2020] [Revised: 11/05/2020] [Accepted: 11/06/2020] [Indexed: 12/20/2022] Open
Abstract
Photodynamic therapy (PDT) is a treatment modality that involves three components: combination of a photosensitizer, light and molecular oxygen that leads to localized formation of reactive oxygen species (ROS). The ROS generated from this promising therapeutic modality can be lethal to the cell and leads to consequential destruction of tumor cells. However, sometimes the ROS trigger a stress response survival mechanism that helps the cells to cope with PDT-induced damage, resulting in resistance to the treatment. One preferred mechanism of cell death induced by PDT is apoptosis, and B-cell lymphoma 2 (Bcl-2) family proteins have been described as a major determinant of life or death decision of the death pathways. Apoptosis is a cellular self-destruction mechanism to remove old cells through the biological event of tissue homeostasis. The Bcl-2 family proteins act as a critical mediator of a life–death decision of cells in maintaining tissue homeostasis. There are several reports that show cancer cells developing resistance due to the increased interaction of the pro-survival Bcl-2 family proteins. However, the key mechanisms leading to apoptosis evasion and drug resistance have not been adequately understood. Therefore, it is critical to understand the mechanisms of PDT resistance, as well as the Bcl-2 family proteins, to give more insight into the treatment outcomes. In this review, we describe the role of Bcl-2 gene family proteins’ interaction in response to disease progression and PDT-induced resistance mechanisms.
Collapse
|
21
|
Wang Y, Yang L, Zhang X, Cui W, Liu Y, Sun Q, He Q, Zhao S, Zhang G, Wang Y, Chen S. Epigenetic regulation of ferroptosis by H2B monoubiquitination and p53. EMBO Rep 2019; 20:e47563. [PMID: 31267712 PMCID: PMC6607012 DOI: 10.15252/embr.201847563] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 04/05/2019] [Accepted: 04/23/2019] [Indexed: 02/06/2023] Open
Abstract
Monoubiquitination of histone H2B on lysine 120 (H2Bub1) is an epigenetic mark generally associated with transcriptional activation, yet the global functions of H2Bub1 remain poorly understood. Ferroptosis is a form of non-apoptotic cell death characterized by the iron-dependent overproduction of lipid hydroperoxides, which can be inhibited by the antioxidant activity of the solute carrier family member 11 (SLC7A11/xCT), a component of the cystine/glutamate antiporter. Whether nuclear events participate in the regulation of ferroptosis is largely unknown. Here, we show that the levels of H2Bub1 are decreased during erastin-induced ferroptosis and that loss of H2Bub1 increases the cellular sensitivity to ferroptosis. H2Bub1 epigenetically activates the expression of SLC7A11. Additionally, we show that the tumor suppressor p53 negatively regulates H2Bub1 levels independently of p53's transcription factor activity by promoting the nuclear translocation of the deubiquitinase USP7. Moreover, our studies reveal that p53 decreases H2Bub1 occupancy on the SLC7A11 gene regulatory region and represses the expression of SLC7A11 during erastin treatment. These data not only suggest a noncanonical role of p53 in chromatin regulation but also link p53 to ferroptosis via an H2Bub1-mediated epigenetic pathway. Overall, our work uncovers a previously unappreciated epigenetic mechanism for the regulation of ferroptosis.
Collapse
Affiliation(s)
- Yufei Wang
- Laboratory of Molecular and Cellular BiologySchool of Forensic SciencesCenter for Translational Medicine at The First Affiliated HospitalXi'an Jiao Tong University Health Science CenterXi'anShaanxiChina
| | - Lu Yang
- Laboratory of Molecular and Cellular BiologySchool of Forensic SciencesCenter for Translational Medicine at The First Affiliated HospitalXi'an Jiao Tong University Health Science CenterXi'anShaanxiChina
| | - Xiaojun Zhang
- Department of Science and EducationPeople's Hospital of ZunhuaTangshanHebeiChina
| | - Wen Cui
- School of Forensic Sciences and Laboratory MedicineJining Medical UniversityJiningShandongChina
| | - Yanping Liu
- Laboratory of Molecular and Cellular BiologySchool of Forensic SciencesCenter for Translational Medicine at The First Affiliated HospitalXi'an Jiao Tong University Health Science CenterXi'anShaanxiChina
| | - Qin‐Ru Sun
- Laboratory of Molecular and Cellular BiologySchool of Forensic SciencesCenter for Translational Medicine at The First Affiliated HospitalXi'an Jiao Tong University Health Science CenterXi'anShaanxiChina
| | - Qing He
- Laboratory of Molecular and Cellular BiologySchool of Forensic SciencesCenter for Translational Medicine at The First Affiliated HospitalXi'an Jiao Tong University Health Science CenterXi'anShaanxiChina
| | - Shiyan Zhao
- Community Health Service Center of YaoqiangJinanShandongChina
| | - Guo‐An Zhang
- School of Forensic Sciences and Laboratory MedicineJining Medical UniversityJiningShandongChina
| | - Yequan Wang
- School of Forensic Sciences and Laboratory MedicineJining Medical UniversityJiningShandongChina
| | - Su Chen
- Laboratory of Molecular and Cellular BiologySchool of Forensic SciencesCenter for Translational Medicine at The First Affiliated HospitalXi'an Jiao Tong University Health Science CenterXi'anShaanxiChina
- Department of Science and EducationPeople's Hospital of ZunhuaTangshanHebeiChina
- School of Forensic Sciences and Laboratory MedicineJining Medical UniversityJiningShandongChina
| |
Collapse
|
22
|
Khooei A, Atabaki Pasdar F, Fazel A, Mahmoudi M, Nikravesh MR, Daneshmand Shahbazian S. P53 expression in various types of hydropic placentas (through ploidy analysis as a complementary tool in diagnosis of samples). CASPIAN JOURNAL OF INTERNAL MEDICINE 2019; 10:205-210. [PMID: 31363399 PMCID: PMC6619464 DOI: 10.22088/cjim.10.2.205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 12/17/2018] [Accepted: 12/20/2018] [Indexed: 11/25/2022]
Abstract
BACKGROUND Placentas characterized by hydropic swelling of chorionic villi occur in a spectrum of pathological conditions including hydropic abortion (HA), partial hydatidiform mole (PHM) and complete hydatidiform mole (CHM). The purpose of this study was to investigate whether the expression of p53 tumour suppressor protein could differentiate these various types of hydropic placentas. METHODS p53 immunohistochemical staining was performed in 19 molar (8 PHM and 11 CHM) and 10 non-molar (HA) formalin-fixed, paraffin-embedded tissue samples. Ploidy analysis using flow cytometry was performed as a complementary tool in diagnosis of samples. RESULTS DNA histograms obtained from all samples had confirmed diploidy in HAs and CHMs and triploidy in PHMs. p53 immunoreactivity was assessed in villous cytotrophoblasts, syncytiotrophoblasts and stromal cells. The p53 positive reaction was predominantly observed in the nuclei of cytotrophoblastic cells and rarely in stromal cells, no reaction was seen in syncytiotrophoblasts. The mean percentage of p53 positive cells were 6.10±3.75 for HA, 25.87±13.4 for PHM and 39.83±18.76 for CHM. There was a significant difference in P53 immunoreactivity of cytotrophoblastic cells between CHM and HA (P<0.001), and between PHM and HA (P=0.004). There was no significant difference in immunohistochemical reactivity between CHM and PHM (P=0.068). CONCLUSION This study confirms that p53 immunostaining may be helpful in distinguishing complete and partial hydatidiform mole from hydropic abortion, but not complete hydatidiform mole from partial hydatidiform mole.
Collapse
Affiliation(s)
- Alireza Khooei
- Department of Pathology, Emam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Atabaki Pasdar
- Department of Anatomical Sciences, Faculty of Medicine, Urmia University of Medical Sciences, Urmia
| | - Alireza Fazel
- Department of Anatomical Sciences, Faculty of Medicine, Urmia University of Medical Sciences, Urmia
| | - Mahmoud Mahmoudi
- Immunology Research Center, Bu Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Nikravesh
- Department of Anatomy and Cell Biology, faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shahrzad Daneshmand Shahbazian
- Department of Biomedical Engineering, School of Electrical Engineering, Iran University of Science & Technology (IUST), Tehran, Iran
| |
Collapse
|
23
|
Chen L, Wang H. Nicotine Promotes Human Papillomavirus (HPV)-Immortalized Cervical Epithelial Cells (H8) Proliferation by Activating RPS27a-Mdm2-P53 Pathway In Vitro. Toxicol Sci 2018; 167:408-418. [DOI: 10.1093/toxsci/kfy246] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Lu Chen
- Department of Occupational and Environmental Health, School of Public Health
- Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Jiangxi 330006, Nanchang, People’s Republic of China
| | - Huai Wang
- Department of Occupational and Environmental Health, School of Public Health
- Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Jiangxi 330006, Nanchang, People’s Republic of China
| |
Collapse
|
24
|
The Guardian of the Genome Revisited: p53 Downregulates Genes Required for Telomere Maintenance, DNA Repair, and Centromere Structure. Cancers (Basel) 2018; 10:cancers10050135. [PMID: 29734785 PMCID: PMC5977108 DOI: 10.3390/cancers10050135] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 04/30/2018] [Accepted: 05/02/2018] [Indexed: 02/06/2023] Open
Abstract
The p53 protein has been extensively studied for its capacity to prevent proliferation of cells with a damaged genome. Surprisingly, however, our recent analysis of mice expressing a hyperactive mutant p53 that lacks the C-terminal domain revealed that increased p53 activity may alter genome maintenance. We showed that p53 downregulates genes essential for telomere metabolism, DNA repair, and centromere structure and that a sustained p53 activity leads to phenotypic traits associated with dyskeratosis congenita and Fanconi anemia. This downregulation is largely conserved in human cells, which suggests that our findings could be relevant to better understand processes involved in bone marrow failure as well as aging and tumor suppression.
Collapse
|
25
|
Simabuco FM, Morale MG, Pavan IC, Morelli AP, Silva FR, Tamura RE. p53 and metabolism: from mechanism to therapeutics. Oncotarget 2018; 9:23780-23823. [PMID: 29805774 PMCID: PMC5955117 DOI: 10.18632/oncotarget.25267] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 04/06/2018] [Indexed: 11/25/2022] Open
Abstract
The tumor cell changes itself and its microenvironment to adapt to different situations, including action of drugs and other agents targeting tumor control. Therefore, metabolism plays an important role in the activation of survival mechanisms to keep the cell proliferative potential. The Warburg effect directs the cellular metabolism towards an aerobic glycolytic pathway, despite the fact that it generates less adenosine triphosphate than oxidative phosphorylation; because it creates the building blocks necessary for cell proliferation. The transcription factor p53 is the master tumor suppressor; it binds to more than 4,000 sites in the genome and regulates the expression of more than 500 genes. Among these genes are important regulators of metabolism, affecting glucose, lipids and amino acids metabolism, oxidative phosphorylation, reactive oxygen species (ROS) generation and growth factors signaling. Wild-type and mutant p53 may have opposing effects in the expression of these metabolic genes. Therefore, depending on the p53 status of the cell, drugs that target metabolism may have different outcomes and metabolism may modulate drug resistance. Conversely, induction of p53 expression may regulate differently the tumor cell metabolism, inducing senescence, autophagy and apoptosis, which are dependent on the regulation of the PI3K/AKT/mTOR pathway and/or ROS induction. The interplay between p53 and metabolism is essential in the decision of cell fate and for cancer therapeutics.
Collapse
Affiliation(s)
- Fernando M. Simabuco
- Laboratory of Functional Properties in Foods, School of Applied Sciences (FCA), Universidade de Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Mirian G. Morale
- Center for Translational Investigation in Oncology/LIM24, Instituto do Câncer do Estado de São Paulo (ICESP), São Paulo, Brazil
- Department of Radiology and Oncology, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Isadora C.B. Pavan
- Laboratory of Functional Properties in Foods, School of Applied Sciences (FCA), Universidade de Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Ana P. Morelli
- Laboratory of Functional Properties in Foods, School of Applied Sciences (FCA), Universidade de Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Fernando R. Silva
- Laboratory of Functional Properties in Foods, School of Applied Sciences (FCA), Universidade de Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Rodrigo E. Tamura
- Center for Translational Investigation in Oncology/LIM24, Instituto do Câncer do Estado de São Paulo (ICESP), São Paulo, Brazil
- Department of Radiology and Oncology, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
26
|
Kopnin B. Genetic Events Responsible for Colorectal Tumorigenesis: Achievements and Challenges. TUMORI JOURNAL 2018; 79:235-43. [PMID: 8249174 DOI: 10.1177/030089169307900401] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Colorectal carcinogenesis is a multistep process that is accompanied by accumulation of changes in proto-oncogenes and tumor-suppressor genes. APC/MCC, RAS, DCC, p53 mutations and/or allelic losses, hyperexpression of c-MYC and RB genes, as well as other genomic alterations appear at characteristic stages of tumor development and are observed in most neoplasms. However, consideration of each of these abnormalities leaves many unanswered questions. The striking data on recurrent amplification of the RB tumor-suppressor gene as well as suppressive activities of protein kinase C and activated RAS genes, at least in some colon carcinoma cell lines, suggest the unusual effects of some signalling pathways in colonic epithelial cells. The results obtained to date indicate that distinct sets of genetic changes may underlie the development of colorectal tumors.
Collapse
Affiliation(s)
- B Kopnin
- Institute of Carcinogenesis, Cancer Research Center, Moscow
| |
Collapse
|
27
|
Kawasaki K, Fujii M, Sato T. Gastroenteropancreatic neuroendocrine neoplasms: genes, therapies and models. Dis Model Mech 2018; 11:11/2/dmm029595. [PMID: 29590641 PMCID: PMC5894937 DOI: 10.1242/dmm.029595] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) refer to a group of heterogeneous cancers of neuroendocrine cell phenotype that mainly fall into one of two subtypes: gastroenteropancreatic neuroendocrine tumors (GEP-NETs; well differentiated) or gastroenteropancreatic neuroendocrine carcinomas (GEP-NECs; poorly differentiated). Although originally defined as orphan cancers, their steadily increasing incidence highlights the need to better understand their etiology. Accumulating epidemiological and clinical data have shed light on the pathological characteristics of these diseases. However, the relatively low number of patients has hampered conducting large-scale clinical trials and hence the development of novel treatment strategies. To overcome this limitation, tractable disease models that faithfully reflect clinical features of these diseases are needed. In this Review, we summarize the current understanding of the genetics and biology of these diseases based on conventional disease models, such as genetically engineered mouse models (GEMMs) and cell lines, and discuss the phenotypic differences between the models and affected humans. We also highlight the emerging disease models derived from human clinical samples, including patient-derived xenograft models and organoids, which may provide biological and therapeutic insights into GEP-NENs.
Collapse
Affiliation(s)
- Kenta Kawasaki
- Department of Gastroenterology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Masayuki Fujii
- Department of Gastroenterology, Keio University School of Medicine, Tokyo 160-8582, Japan.,Department of Surgical Oncology, The University of Tokyo, Tokyo 113-8654, Japan
| | - Toshiro Sato
- Department of Gastroenterology, Keio University School of Medicine, Tokyo 160-8582, Japan
| |
Collapse
|
28
|
Cheng L, Deng H, Ma D, Zhai B, Zhang Q, Li L, Xi Z. Branch-PCR constructed TP53 gene nanovector for potential cancer therapy. Chem Commun (Camb) 2018; 54:9687-9690. [DOI: 10.1039/c8cc05066j] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
A novel and efficient branch PCR strategy can be used to construct a TP53 gene nanovector for effective cancer therapy.
Collapse
Affiliation(s)
- Longhuai Cheng
- State Key Laboratory of Elemento-Organic Chemistry and Department of Chemical Biology
- National Pesticide Engineering Research Center (Tianjin)
- Collaborative Innovation Center of Chemical Science and Engineering, (Tianjin)
- College of Chemistry
- Nankai University
| | - Huiting Deng
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy
- Tianjin Key Laboratory of Molecular Drug Research
- Nankai University
- Tianjin
- China
| | - Dejun Ma
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy
- Tianjin Key Laboratory of Molecular Drug Research
- Nankai University
- Tianjin
- China
| | - Beibei Zhai
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy
- Tianjin Key Laboratory of Molecular Drug Research
- Nankai University
- Tianjin
- China
| | - Qiangzhe Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy
- Tianjin Key Laboratory of Molecular Drug Research
- Nankai University
- Tianjin
- China
| | - Luyuan Li
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy
- Tianjin Key Laboratory of Molecular Drug Research
- Nankai University
- Tianjin
- China
| | - Zhen Xi
- State Key Laboratory of Elemento-Organic Chemistry and Department of Chemical Biology
- National Pesticide Engineering Research Center (Tianjin)
- Collaborative Innovation Center of Chemical Science and Engineering, (Tianjin)
- College of Chemistry
- Nankai University
| |
Collapse
|
29
|
Aubrey BJ, Kelly GL, Janic A, Herold MJ, Strasser A. How does p53 induce apoptosis and how does this relate to p53-mediated tumour suppression? Cell Death Differ 2017; 25:104-113. [PMID: 29149101 DOI: 10.1038/cdd.2017.169] [Citation(s) in RCA: 901] [Impact Index Per Article: 112.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 09/05/2017] [Accepted: 09/08/2017] [Indexed: 01/10/2023] Open
Abstract
The tumour suppressor gene TP53 is mutated in ~50% of human cancers. In addition to its function in tumour suppression, p53 also plays a major role in the response of malignant as well as nontransformed cells to many anticancer therapeutics, particularly those that cause DNA damage. P53 forms a homotetrameric transcription factor that is reported to directly regulate ~500 target genes, thereby controlling a broad range of cellular processes, including cell cycle arrest, cell senescence, DNA repair, metabolic adaptation and cell death. For a long time, induction of apoptotic death in nascent neoplastic cells was regarded as the principal mechanism by which p53 prevents tumour development. This concept has, however, recently been challenged by the findings that in striking contrast to Trp53-deficient mice, gene-targeted mice that lack the critical effectors of p53-induced apoptosis do not develop tumours spontaneously. Remarkably, even mice lacking all mediators critical for p53-induced apoptosis, G1/S boundary cell cycle arrest and cell senescence do not develop any tumours spontaneously. In this review we discuss current understanding of the mechanisms by which p53 induces cell death and how this affects p53-mediated tumour suppression and the response of malignant cells to anticancer therapy.
Collapse
Affiliation(s)
- Brandon J Aubrey
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Gemma L Kelly
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Ana Janic
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Marco J Herold
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
30
|
P-3F, a microtubule polymerization inhibitor enhances P53 stability through the change in localization of RPS27a. Int J Biochem Cell Biol 2017; 92:53-62. [DOI: 10.1016/j.biocel.2017.09.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 09/05/2017] [Accepted: 09/15/2017] [Indexed: 02/02/2023]
|
31
|
Wang H, Feng J, Zhou T, Wei L, Zhou J. Involvement of RPL11 in the enhancement of P53 stability by a podophyllum derivative, a topoisomerase II inhibitor. Cell Biol Int 2017; 42:121-129. [DOI: 10.1002/cbin.10877] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 09/24/2017] [Indexed: 02/03/2023]
Affiliation(s)
- Huai Wang
- School of Public Health; Nanchang University; 461 Ba Yi Avenue Nanchang, Jiangxi 330006 P. R. China
- Jiangxi Provincial Key Laboratory of Preventive Medicine; Nanchang University; 461 Ba Yi Avenue Nanchang, Jiangxi 330006 P. R. China
| | - Jiang Feng
- School of Public Health; Nanchang University; 461 Ba Yi Avenue Nanchang, Jiangxi 330006 P. R. China
- Jiangxi Provincial Key Laboratory of Preventive Medicine; Nanchang University; 461 Ba Yi Avenue Nanchang, Jiangxi 330006 P. R. China
| | - Tong Zhou
- School of Public Health; Nanchang University; 461 Ba Yi Avenue Nanchang, Jiangxi 330006 P. R. China
- Jiangxi Provincial Key Laboratory of Preventive Medicine; Nanchang University; 461 Ba Yi Avenue Nanchang, Jiangxi 330006 P. R. China
| | - Lijun Wei
- School of Public Health; Nanchang University; 461 Ba Yi Avenue Nanchang, Jiangxi 330006 P. R. China
- Jiangxi Provincial Key Laboratory of Preventive Medicine; Nanchang University; 461 Ba Yi Avenue Nanchang, Jiangxi 330006 P. R. China
| | - Jianming Zhou
- School of Public Health; Nanchang University; 461 Ba Yi Avenue Nanchang, Jiangxi 330006 P. R. China
- Jiangxi Provincial Key Laboratory of Preventive Medicine; Nanchang University; 461 Ba Yi Avenue Nanchang, Jiangxi 330006 P. R. China
| |
Collapse
|
32
|
Chen J, Wang A, Chen Q. SirT3 and p53 Deacetylation in Aging and Cancer. J Cell Physiol 2017; 232:2308-2311. [PMID: 27791271 DOI: 10.1002/jcp.25669] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 10/27/2016] [Indexed: 12/23/2022]
Affiliation(s)
- Jijun Chen
- Research and Development; Allonger LLC, Columbia Maryland
- Mei Chen Biotechnology Co. Ltd.; Qingdao, Shan Dong China
| | - Aiqin Wang
- The Affiliated Hospital of Qingdao University; Qingdao, Shan Dong China
| | - Qingqi Chen
- The Affiliated Hospital of Qingdao University; Qingdao, Shan Dong China
| |
Collapse
|
33
|
Le M, Mothersill CE, Seymour CB, Rainbow AJ, McNeill FE. An Observed Effect of p53 Status on the Bystander Response to Radiation-Induced Cellular Photon Emission. Radiat Res 2017; 187:169-185. [PMID: 28118118 DOI: 10.1667/rr14342.1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
In this study, we investigated the potential influence of p53 on ultraviolet (UV) signal generation and response of bystander cells to the UV signals generated by beta-irradiated cells. Five cell lines of various p53 status (HaCaT, mutated; SW48, wild-type; HT29, mutated; HCT116+/+, wild-type; HCT116-/-, null) were irradiated with beta particles from tritium. Signal generation (photon emission at 340 ± 5 nm) was quantified from irradiated cells using a photomultiplier tube. Bystander response (clonogenic survival) was assessed by placing reporter cell flasks directly superior to irradiated signal-emitting cells. All cell lines emitted significant quantities of UV after tritium exposure. The magnitudes of HaCaT and HT29 photon emission at 340 nm were similar to each other while they were significantly different from the stronger signals emitted from SW48, HCT116+/+ and HCT116-/- cells. In regard to the bystander responses, HaCaT, HCT116+/+ and SW48 cells demonstrated significant reductions in survival as a result of exposure to emission signals. HCT116-/- and HT29 cells did not exhibit any changes in survival and thus were considered to be lacking the mechanisms or functions required to elicit a response. The survival response was found not to correlate with the observed signal strength for all experimental permutations; this may be attributed to varying emission spectra from cell line to cell line or differences in response sensitivity. Overall, these results suggest that the UV-mediated bystander response is influenced by the p53 status of the cell line. Wild-type p53 cells (HCT116+/+ and SW48) demonstrated significant responses to UV signals whereas the p53-null cell line (HCT116-/-) lacked any response. The two mutated p53 cell lines exhibited contrasting responses, which may be explained by unique modulation of functions by different point mutations. The reduced response (cell death) exhibited by p53-mutated cells compared to p53 wild-type cells suggests a possible role of the assessed p53 mutations in radiation-induced cancer susceptibility and reduced efficacy of radiation-directed therapy.
Collapse
Affiliation(s)
- M Le
- a Radiation Sciences Graduate Program and Departments of
| | | | | | | | - F E McNeill
- c Physics and Astronomy, McMaster University, Hamilton Ontario, L8S 4L8, Canada
| |
Collapse
|
34
|
Miller M, Shirole N, Tian R, Pal D, Sordella R. The Evolution of TP53 Mutations: From Loss-of-Function to Separation-of-Function Mutants. JOURNAL OF CANCER BIOLOGY & RESEARCH 2016; 4:1091. [PMID: 28191499 PMCID: PMC5298884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
As the most mutated gene in cancer, it is no surprise that TP53 has been the center of cancer biology discourse since its discovery in the late 1970s. Although early demonstrations of p53's role in the modulation of cell proliferation and survival solidified its classification as a tumor suppressor and transcription factor, our conceptualization of p53 is ever-evolving. Here, we present novel evidence of the role of alternative splicing isoforms, truncating/separation-of-function mutations, and hotspot silent mutations in the regulation of p53's activities.
Collapse
Affiliation(s)
| | - Nitin Shirole
- Cancer Center, Cold Spring Harbor Laboratory, USA
- Graduate Program in Genetics, Stony Brook University, USA
| | - Ruxiao Tian
- Cancer Center, Cold Spring Harbor Laboratory, USA
| | - Debjani Pal
- Cancer Center, Cold Spring Harbor Laboratory, USA
- Graduate Program in Molecular and Cellular Biology, Stony Brook University, USA
| | - Raffaella Sordella
- Cancer Center, Cold Spring Harbor Laboratory, USA
- Graduate Program in Genetics, Stony Brook University, USA
| |
Collapse
|
35
|
Rahmanian N, Hosseinimehr SJ, Khalaj A. The paradox role of caspase cascade in ionizing radiation therapy. J Biomed Sci 2016; 23:88. [PMID: 27923354 PMCID: PMC5142153 DOI: 10.1186/s12929-016-0306-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 11/30/2016] [Indexed: 12/19/2022] Open
Abstract
Radiotherapy alone or in combination with chemotherapy/surgery is widely used for treatment of cancers. It reduces tumor growth and prevents metastasis. While ionizing radiation activates caspase cascade resulted in apoptosis in cancer cells, it also stimulates tumor cell re-population that leads to reduce the effectiveness of the radiation therapy. This review describes the mechanisms for paradox role of caspase cascade in cancer therapy and discusses the logical and practical strategies for improvement the therapeutic index of radiotherapy through enhancement of radiosensitivity and decreasing the rate of tumor recurrence.
Collapse
Affiliation(s)
- Najmeh Rahmanian
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ali Khalaj
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
36
|
Purushotham G, Padma Y, Nabiha Y, Venkata Raju RR. In vitro evaluation of anti-proliferative, anti-inflammatory and pro-apoptotic activities of the methanolic extracts of Andrographis nallamalayana Ellis on A375 and B16F10 melanoma cell lines. 3 Biotech 2016; 6:212. [PMID: 28330284 PMCID: PMC5045834 DOI: 10.1007/s13205-016-0529-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 09/19/2016] [Indexed: 02/07/2023] Open
Abstract
Natural plant products have been widely used in controlling cancer with fewer or no side effects and the use of plant extracts as complementary to synthetic medicine is gaining increased popularity. Members of Andrographis plants possess important medicinal properties. In the present study, anti-cancerous properties of Andrographis nallamalayana (AN) were tested on A375 and B16F10 skin melanoma cancer cell lines. The leaf extracts of AN significantly reduced the cell viability and cell survival of skin cancer cell lines, achieved by MTT assay and clonogenic assays, respectively. Further, TUNEL assays revealed that AN extracts induces the apoptosis. Western blot analysis revealed that AN leaf extracts reduced the expression of Bcl-2, an anti-apoptotic protein and induced the expression of proapoptotic molecules such as Bcl-2 associated death promoter protein (BAD), Bcl-2 associated X protein (BAX) and cleaved caspase-3. Moreover, the qRT-PCR and western blot analysis demonstrated the reduced expression of G2/M phase proteins cdk1, cyclin B1 and increased expression of p53, cyclin-dependent kinase 1 inhibitor, p21. Further, immunofluorescence analysis revealed that AN reduced the NF-κB nuclear translocation, luciferase reporter assays demonstrated reporter gene activation. qRT-PCR assays showed that AN significantly reduced the expression of NF-κB target genes. The results concluded that the extracts of AN exhibited significant anti-proliferative, anti-inflammatory and pro-apoptotic activities on melanoma skin cancer cell lines.
Collapse
|
37
|
Olivos DJ, Mayo LD. Emerging Non-Canonical Functions and Regulation by p53: p53 and Stemness. Int J Mol Sci 2016; 17:ijms17121982. [PMID: 27898034 PMCID: PMC5187782 DOI: 10.3390/ijms17121982] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 11/10/2016] [Accepted: 11/15/2016] [Indexed: 01/15/2023] Open
Abstract
Since its discovery nearly 40 years ago, p53 has ascended to the forefront of investigated genes and proteins across diverse research disciplines and is recognized most exclusively for its role in cancer as a tumor suppressor. Levine and Oren (2009) reviewed the evolution of p53 detailing the significant discoveries of each decade since its first report in 1979. In this review, we will highlight the emerging non-canonical functions and regulation of p53 in stem cells. We will focus on general themes shared among p53's functions in non-malignant stem cells and cancer stem-like cells (CSCs) and the influence of p53 on the microenvironment and CSC niche. We will also examine p53 gain of function (GOF) roles in stemness. Mutant p53 (mutp53) GOFs that lead to survival, drug resistance and colonization are reviewed in the context of the acquisition of advantageous transformation processes, such as differentiation and dedifferentiation, epithelial-to-mesenchymal transition (EMT) and stem cell senescence and quiescence. Finally, we will conclude with therapeutic strategies that restore wild-type p53 (wtp53) function in cancer and CSCs, including RING finger E3 ligases and CSC maintenance. The mechanisms by which wtp53 and mutp53 influence stemness in non-malignant stem cells and CSCs or tumor-initiating cells (TICs) are poorly understood thus far. Further elucidation of p53's effects on stemness could lead to novel therapeutic strategies in cancer research.
Collapse
Affiliation(s)
- David J Olivos
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
- Department of Pediatrics, Herman B Wells Center for Pediatrics Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Lindsey D Mayo
- Department of Pediatrics, Herman B Wells Center for Pediatrics Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
38
|
Subbaramaiah K, Brown KA, Zahid H, Balmus G, Weiss RS, Herbert BS, Dannenberg AJ. Hsp90 and PKM2 Drive the Expression of Aromatase in Li-Fraumeni Syndrome Breast Adipose Stromal Cells. J Biol Chem 2016; 291:16011-23. [PMID: 27467582 PMCID: PMC4965552 DOI: 10.1074/jbc.m115.698902] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 05/23/2016] [Indexed: 11/06/2022] Open
Abstract
Li-Fraumeni syndrome (LFS) patients harbor germ line mutations in the TP53 gene and are at increased risk of hormone receptor-positive breast cancers. Recently, elevated levels of aromatase, the rate-limiting enzyme for estrogen biosynthesis, were found in the breast tissue of LFS patients. Although p53 down-regulates aromatase expression, the underlying mechanisms are incompletely understood. In the present study, we found that LFS stromal cells expressed higher levels of Hsp90 ATPase activity and aromatase compared with wild-type stromal cells. Inhibition of Hsp90 ATPase suppressed aromatase expression. Silencing Aha1 (activator of Hsp90 ATPase 1), a co-chaperone of Hsp90 required for its ATPase activity, led to both inhibition of Hsp90 ATPase activity and reduced aromatase expression. In comparison with wild-type stromal cells, increased levels of the Hsp90 client proteins, HIF-1α, and PKM2 were found in LFS stromal cells. A complex comprised of HIF-1α and PKM2 was recruited to the aromatase promoter II in LFS stromal cells. Silencing either HIF-1α or PKM2 suppressed aromatase expression in LFS stromal cells. CP-31398, a p53 rescue compound, suppressed levels of Aha1, Hsp90 ATPase activity, levels of PKM2 and HIF-1α, and aromatase expression in LFS stromal cells. Consistent with these in vitro findings, levels of Hsp90 ATPase activity, Aha1, HIF-1α, PKM2, and aromatase were increased in the mammary glands of p53 null versus wild-type mice. PKM2 and HIF-1α were shown to co-localize in the nucleus of stromal cells of LFS breast tissue. Taken together, our results show that the Aha1-Hsp90-PKM2/HIF-1α axis mediates the induction of aromatase in LFS.
Collapse
Affiliation(s)
- Kotha Subbaramaiah
- From the Department of Medicine, Weill Cornell Medical College, New York, New York 10065,
| | - Kristy A Brown
- the Metabolism and Cancer Laboratory, Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia, Monash University, Clayton, Victoria 3800, Australia
| | - Heba Zahid
- the Metabolism and Cancer Laboratory, Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia, Monash University, Clayton, Victoria 3800, Australia, the Faculty of Applied Medical Science, Taibah University, Medina, Saudi Arabia
| | - Gabriel Balmus
- the Department of Biomedical Sciences, Cornell University, Ithaca, New York 14853, and
| | - Robert S Weiss
- the Department of Biomedical Sciences, Cornell University, Ithaca, New York 14853, and
| | - Brittney-Shea Herbert
- the Department of Medical and Molecular Genetics, Indiana University Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Andrew J Dannenberg
- From the Department of Medicine, Weill Cornell Medical College, New York, New York 10065
| |
Collapse
|
39
|
Abstract
Inactivation of the p53 tumor suppressor gene is one of the commonest genetic changes identified in human breast cancer. In this review, the structure and function of the p53 gene and its protein products will be discussed, with particular reference to p53 alterations that contribute to carcinogenesis. The frequency and pattern of p53 alterations in breast cancer will be outlined, laboratory methods for their detection briefly summarized, and the potential use of p53 as a prognostic and predictive marker discussed.
Collapse
|
40
|
Luo SW, Wang WN, Sun ZM, Xie FX, Kong JR, Liu Y, Cheng CH. Molecular cloning, characterization and expression analysis of (B-cell lymphoma-2 associated X protein) Bax in the orange-spotted grouper (Epinephelus coioides) after the Vibrio alginolyticus challenge. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 60:66-79. [PMID: 26905633 DOI: 10.1016/j.dci.2016.02.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 02/13/2016] [Accepted: 02/15/2016] [Indexed: 06/05/2023]
Abstract
Bax is a pro-apoptotic member of Bcl-2 like superfamily, playing an important role in regulating the apoptosis. In this study, the full-length Bax (EcBax) was obtained, containing a 5'UTR of 64 bp, an ORF of 579 bp and a 3'UTR of 1021 bp. The EcBax gene encoded a polypeptide of 192 amino acids with an estimated molecular mass of 21.55 KDa and a predicted isoelectric point (pI) of 6.75. The deduced amino acid sequence analysis showed that EcBax comprised the conserved residues and the characteristic domains known to the critical function of Bax. qRT-PCR analysis revealed that EcBax mRNA was broadly expressed in all of the examined tissues, while the highest expression level was observed in blood, followed by the expression in liver, gill, spleen, kidney, heart, muscle and intestine. A sharp increase of EcBax expression was observed in the vibrio challenge group by comparing with those in the control. Subcellular localization analysis revealed that EcBax was predominantly localized in the cytoplasm. EcBax exerted a regulatory role in modulating the mitochondrial membrane potential, promoting the cytochrome c release, and then activating the downstream caspase signaling. Moreover, the overexpression of EcBax can decrease the cell viability and antagonize NF-kB, AP-1, Stat3 promoter activity in Hela cells. These results indicate that EcBax containing the conserved domain of pro-apoptotic member of Bcl-2 family may disrupt the mammalian signaling and play a regulative role in the apoptotic process.
Collapse
Affiliation(s)
- Sheng-Wei Luo
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, College of Life Science, South China Normal University, Guangzhou 510631, PR China
| | - Wei-Na Wang
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, College of Life Science, South China Normal University, Guangzhou 510631, PR China.
| | - Zuo-Ming Sun
- Division of Immunology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | - Fu-Xing Xie
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, College of Life Science, South China Normal University, Guangzhou 510631, PR China
| | - Jing-Rong Kong
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, College of Life Science, South China Normal University, Guangzhou 510631, PR China
| | - Yuan Liu
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, College of Life Science, South China Normal University, Guangzhou 510631, PR China
| | - Chang-Hong Cheng
- Guangzhou Key Laboratory of Subtropical Biodiversity and Biomonitoring, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, College of Life Science, South China Normal University, Guangzhou 510631, PR China
| |
Collapse
|
41
|
Perz E, Kuhn JG. Review : p53 in the pathogenesis, diagnosis, and treatment of cancer. J Oncol Pharm Pract 2016. [DOI: 10.1177/107815529800400201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Objective. The cellular functions of p53, the conse quences of the loss of p53 function, and the potential impact of p53 in oncology are reviewed within the framework of an overview of the molecular basis of cancer and cell cycle control. Data Sources. A MEDLINE search of articles from 1976 to the present was conducted using the terms p53 protein and p53 gene. The search was restricted to the English language. Oncology and molecular biology textbooks were used as additional references. Data Extraction. We reviewed the literature to discuss the cellular function of p53, the mechanisms of p53 inactivation, the cellular consequences of the loss of p53 function, the role of p53 loss in tumori genesis, and the potential applications of this knowl edge. Data Synthesis. p53 mutations are found in ~ 50% of human cancers. Knowledge of p53 functions and defects provides the basis for potential applica tions in the areas of cancer epidemiology, cancer diagnosis, and determination of prognosis. An under standing of the functions and defects of p53 also presents a host of opportunities for the design of novel cancer therapies. Therapeutic approaches be ing studied include the restoration of p53 by gene therapy, the alteration of mutant p53 expression by antisense therapy, and the use of p53 mutations as a target for directing therapy to cancer cells; some of these approaches are already under phase I investiga tion. As knowledge of p53 unfolds, additional thera peutic approaches will certainly be developed. The story of p53 illustrates that the manipulation of mo lecular interactions is a new frontier in therapeutics and offers an additional role for oncology pharmacy specialists.
Collapse
Affiliation(s)
- Elizabeth Perz
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, College of Pharmacy, The University of Texas at Austin, Austin, Texas
| | - John G. Kuhn
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, Department of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, College of Pharmacy, The University of Texas at Austin, Austin, Texas
| |
Collapse
|
42
|
Valdèz JC, Rachid M, Gobbato N, Perdigòn G. Apoptosis Study in Thymic Involution in Tumour-Bearing Mice. Int J Immunopathol Pharmacol 2016. [DOI: 10.1177/039463209801100201] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
By using a colorectal carcinoma induced by s.c. injection of 1,2-dimethylhydrazine and a transplantable fibrosarcoma developed in inbred BALB/c mice 6–8 weeks old we found that tumour development was accompanied by a remarkable thymic involution. Mice bearing small fibrosarcoma or carcinoma (0,05 cm3) had thymuses and spleens with the same weight as those of normal mice. Thymic atrophy and splenomegalia developed in mice bearing large fibrosarcoma (5,00 cm3) and large carcinoma (0,20 cm3). Thymic involution was not the result of an increase in spontaneous cellular apoptosis. However, an increased susceptibility to apoptosis induced by etoposide was observed in thymocytes from mice bearing large carcinomas or large fibrosarcomas, as compared to the same cells derived from normal or small tumour-bearing mice. Dexamethasone induced comparable levels of apoptosis in thymocytes from all groups (normal mice, mice bearing small and large carcinoma and mice bearing small and large fibrosarcoma); doxorrubicin had no significant effect in any group.
Collapse
Affiliation(s)
- J. C. Valdèz
- Instituto de Microbiologìa “Dr. Luis C. Verna”, Catedra de Inmunologia, Facultad de Bioquimica, Quimica y Farmacia, Universidad Nacional de Tucumàn, San Miguel de Tucumàn, Centro de Referencia para Lactobacilos (CERELA), Republica Argentina
| | - M. Rachid
- Instituto de Microbiologìa “Dr. Luis C. Verna”, Catedra de Inmunologia, Facultad de Bioquimica, Quimica y Farmacia, Universidad Nacional de Tucumàn, San Miguel de Tucumàn, Centro de Referencia para Lactobacilos (CERELA), Republica Argentina
| | - N. Gobbato
- Instituto de Microbiologìa “Dr. Luis C. Verna”, Catedra de Inmunologia, Facultad de Bioquimica, Quimica y Farmacia, Universidad Nacional de Tucumàn, San Miguel de Tucumàn, Centro de Referencia para Lactobacilos (CERELA), Republica Argentina
| | - G. Perdigòn
- Instituto de Microbiologìa “Dr. Luis C. Verna”, Catedra de Inmunologia, Facultad de Bioquimica, Quimica y Farmacia, Universidad Nacional de Tucumàn, San Miguel de Tucumàn, Centro de Referencia para Lactobacilos (CERELA), Republica Argentina
| |
Collapse
|
43
|
Powell E, Shao J, Yuan Y, Chen HC, Cai S, Echeverria GV, Mistry N, Decker KF, Schlosberg C, Do KA, Edwards JR, Liang H, Piwnica-Worms D, Piwnica-Worms H. p53 deficiency linked to B cell translocation gene 2 (BTG2) loss enhances metastatic potential by promoting tumor growth in primary and metastatic sites in patient-derived xenograft (PDX) models of triple-negative breast cancer. Breast Cancer Res 2016; 18:13. [PMID: 26818199 PMCID: PMC4728775 DOI: 10.1186/s13058-016-0673-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 01/12/2016] [Indexed: 11/10/2022] Open
Abstract
Background Despite advances in early diagnosis and treatment of cancer patients, metastasis remains the major cause of mortality. TP53 is one of the most frequently mutated genes in human cancer, and these alterations can occur during the early stages of oncogenesis or as later events as tumors progress to more aggressive forms. Previous studies have suggested that p53 plays a role in cellular pathways that govern metastasis. To investigate how p53 deficiency contributes to late-stage tumor growth and metastasis, we developed paired isogenic patient-derived xenograft (PDX) models of triple-negative breast cancer (TNBC) differing only in p53 status for longitudinal analysis. Methods Patient-derived isogenic human tumor lines differing only in p53 status were implanted into mouse mammary glands. Tumor growth and metastasis were monitored with bioluminescence imaging, and circulating tumor cells (CTCs) were quantified by flow cytometry. RNA-Seq was performed on p53-deficient and p53 wild-type tumors, and functional validation of a lead candidate gene was performed in vivo. Results Isogenic p53 wild-type and p53-deficient tumors metastasized out of mammary glands and colonized distant sites with similar frequency. However, p53-deficient tumors metastasized earlier than p53 wild-type tumors and grew faster in both primary and metastatic sites as a result of increased proliferation and decreased apoptosis. In addition, greater numbers of CTCs were detected in the blood of mice engrafted with p53-deficient tumors. However, when normalized to tumor mass, the number of CTCs isolated from mice bearing parental and p53-deficient tumors was not significantly different. Gene expression profiling followed by functional validation identified B cell translocation gene 2 (BTG2), a downstream effector of p53, as a negative regulator of tumor growth both at primary and metastatic sites. BTG2 expression status correlated with survival of TNBC patients. Conclusions Using paired isogenic PDX-derived metastatic TNBC cells, loss of p53 promoted tumor growth and consequently increased tumor cell shedding into the blood, thus enhancing metastasis. Loss of BTG2 expression in p53-deficient tumors contributed to this metastatic potential by enhancing tumor growth in primary and metastatic sites. Furthermore, clinical data support conclusions generated from PDX models and indicate that BTG2 expression is a candidate prognostic biomarker for TNBC. Electronic supplementary material The online version of this article (doi:10.1186/s13058-016-0673-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Emily Powell
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA. .,Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Jiansu Shao
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Yuan Yuan
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Hsiang-Chun Chen
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Shirong Cai
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Gloria V Echeverria
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Nipun Mistry
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Keith F Decker
- Center for Pharmacogenomics, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| | - Christopher Schlosberg
- Center for Pharmacogenomics, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| | - Kim-Anh Do
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - John R Edwards
- Center for Pharmacogenomics, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| | - Han Liang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - David Piwnica-Worms
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA. .,Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Helen Piwnica-Worms
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
44
|
Velletri T, Xie N, Wang Y, Huang Y, Yang Q, Chen X, Chen Q, Shou P, Gan Y, Cao G, Melino G, Shi Y. P53 functional abnormality in mesenchymal stem cells promotes osteosarcoma development. Cell Death Dis 2016; 7:e2015. [PMID: 26775693 PMCID: PMC4816167 DOI: 10.1038/cddis.2015.367] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 11/13/2015] [Accepted: 11/13/2015] [Indexed: 02/07/2023]
Abstract
It has been shown that p53 has a critical role in the differentiation and functionality of various multipotent progenitor cells. P53 mutations can lead to genome instability and subsequent functional alterations and aberrant transformation of mesenchymal stem cells (MSCs). The significance of p53 in safeguarding our body from developing osteosarcoma (OS) is well recognized. During bone remodeling, p53 has a key role in negatively regulating key factors orchestrating the early stages of osteogenic differentiation of MSCs. Interestingly, changes in the p53 status can compromise bone homeostasis and affect the tumor microenvironment. This review aims to provide a unique opportunity to study the p53 function in MSCs and OS. In the context of loss of function of p53, we provide a model for two sources of OS: MSCs as progenitor cells of osteoblasts and bone tumor microenvironment components. Standing at the bone remodeling point of view, in this review we will first explain the determinant function of p53 in OS development. We will then summarize the role of p53 in monitoring MSC fidelity and in regulating MSC differentiation programs during osteogenesis. Finally, we will discuss the importance of loss of p53 function in tissue microenvironment. We expect that the information provided herein could lead to better understanding and treatment of OS.
Collapse
Affiliation(s)
- T Velletri
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University, School of Medicine, 320 Yueyang Road, Shanghai 200031, China
| | - N Xie
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University, School of Medicine, 320 Yueyang Road, Shanghai 200031, China.,Biochemistry Laboratory IDI-IRCC, Department of Experimental Medicine and Surgery, University of Rome Torvergata, Rome 00133, Italy
| | - Y Wang
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University, School of Medicine, 320 Yueyang Road, Shanghai 200031, China
| | - Y Huang
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University, School of Medicine, 320 Yueyang Road, Shanghai 200031, China
| | - Q Yang
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University, School of Medicine, 320 Yueyang Road, Shanghai 200031, China
| | - X Chen
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University, School of Medicine, 320 Yueyang Road, Shanghai 200031, China
| | - Q Chen
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University, School of Medicine, 320 Yueyang Road, Shanghai 200031, China
| | - P Shou
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University, School of Medicine, 320 Yueyang Road, Shanghai 200031, China
| | - Y Gan
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University, School of Medicine, 320 Yueyang Road, Shanghai 200031, China
| | - G Cao
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University, School of Medicine, 320 Yueyang Road, Shanghai 200031, China
| | - G Melino
- Biochemistry Laboratory IDI-IRCC, Department of Experimental Medicine and Surgery, University of Rome Torvergata, Rome 00133, Italy.,Medical Research Council, Toxicology Unit, Leicester University, Leicester LE1 9HN, UK
| | - Y Shi
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University, School of Medicine, 320 Yueyang Road, Shanghai 200031, China.,Soochow Institutes for Translational Medicine, Soochow University, Suzhou, China
| |
Collapse
|
45
|
Abstract
Cancer cells continue to challenge scientists and oncologists due to the phenomenon of resistance. Moreover, recurrence, as seen in many treated patients, shows that currently-used anti-cancer drugs are unable to prevent the development of new cancer cells harboring new mutations. The purpose of this paper is to try to answer some of the questions regarding why cancer arises and why evolution would naturally lead to the development of cancer. Providing answers to these questions may shed new light on cancer development and potential causes of cancer. This work demonstrates that (1) cancer hallmarks are a series of events that can be organized in three consecutive stages; (2) cancer may develop when cells seek immortality; (3) heterogeneity in tumors may be explained by cancer cells not following universal laws for division; (4) evolution may not have selected for cancer; (5) currently-used anti-cancer drugs, with telomerase and poly adenosine diphosphate ribose polymerase inhibition given as examples, show that we may not be on the right track, as these drugs are probably targeting molecular symptoms of tumors but not their cause; and (6) after an attempt to define the cause of cancer, the potentials of immunotherapy are discussed. Future anti-cancer drugs should be able to shrink the original tumor(s) and most importantly prevent the rise of new cancer cells in treated patients. In order to achieve this goal, new drugs must target the cause of cancer. Therefore, future research must focus on identifying potential causes of cancer common to all types of cancers. Finally, while immunotherapy holds great prospects for future cancer cure and prevention, global action is needed to reduce harmful substances known to contribute to the development of cancer in the environment.
Collapse
Affiliation(s)
- Adouda Adjiri
- Physics Department, Faculty of Sciences, Sétif-1 University, 19000 Sétif, Algeria
| |
Collapse
|
46
|
Zhang M, Zhu X, Zhang Y, Cai Y, Chen J, Sivaprakasam S, Gurav A, Pi W, Makala L, Wu J, Pace B, Tuan-Lo D, Ganapathy V, Singh N, Li H. RCAD/Ufl1, a Ufm1 E3 ligase, is essential for hematopoietic stem cell function and murine hematopoiesis. Cell Death Differ 2015; 22:1922-34. [PMID: 25952549 PMCID: PMC4816109 DOI: 10.1038/cdd.2015.51] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 03/22/2015] [Accepted: 03/24/2015] [Indexed: 12/18/2022] Open
Abstract
The Ufm1 conjugation system is a novel ubiquitin-like modification system, consisting of Ufm1, Uba5 (E1), Ufc1 (E2) and poorly characterized E3 ligase(s). RCAD/Ufl1 (also known as KIAA0776, NLBP and Maxer) was reported to function as a Ufm1 E3 ligase in ufmylation (Ufm1-mediated conjugation) of DDRGK1 and ASC1 proteins. It has also been implicated in estrogen receptor signaling, unfolded protein response (UPR) and neurodegeneration, yet its physiological function remains completely unknown. In this study, we report that RCAD/Ufl1 is essential for embryonic development, hematopoietic stem cell (HSC) survival and erythroid differentiation. Both germ-line and somatic deletion of RCAD/Ufl1 impaired hematopoietic development, resulting in severe anemia, cytopenia and ultimately animal death. Depletion of RCAD/Ufl1 caused elevated endoplasmic reticulum stress and evoked UPR in bone marrow cells. In addition, loss of RCAD/Ufl1 blocked autophagic degradation, increased mitochondrial mass and reactive oxygen species, and led to DNA damage response, p53 activation and enhanced cell death of HSCs. Collectively, our study provides the first genetic evidence for the indispensable role of RCAD/Ufl1 in murine hematopoiesis and development. The finding of RCAD/Ufl1 as a key regulator of cellular stress response sheds a light into the role of a novel protein network including RCAD/Ufl1 and its associated proteins in regulating cellular homeostasis.
Collapse
Affiliation(s)
- M Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
- Department of Biochemistry & Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, USA
| | - X Zhu
- Department of Biochemistry & Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, USA
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Y Zhang
- Department of Biochemistry & Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, USA
| | - Y Cai
- Department of Biochemistry & Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, USA
- Department of Biology, College of Life Sciences, Anhui Normal University, Wuhu, Anhui, China
| | - J Chen
- Department of Biochemistry & Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, USA
| | - S Sivaprakasam
- Department of Biochemistry & Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, USA
| | - A Gurav
- Department of Biochemistry & Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, USA
| | - W Pi
- Department of Biochemistry & Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, USA
| | - L Makala
- Department of Pediatrics, Georgia Regents University, Augusta, GA, USA
| | - J Wu
- Department of Periodontics, College of Dentistry, Cancer Center, University of Illinois at Chicago, Chicago, IL, USA
| | - B Pace
- Department of Biochemistry & Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, USA
- Department of Pediatrics, Georgia Regents University, Augusta, GA, USA
| | - D Tuan-Lo
- Department of Biochemistry & Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, USA
| | - V Ganapathy
- Department of Biochemistry & Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, USA
| | - N Singh
- Department of Biochemistry & Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, USA
| | - H Li
- Department of Biochemistry & Molecular Biology, Cancer Center, Georgia Regents University, Augusta, GA, USA
| |
Collapse
|
47
|
Applegate CC, Lane MA. Role of retinoids in the prevention and treatment of colorectal cancer. World J Gastrointest Oncol 2015; 7:184-203. [PMID: 26483874 PMCID: PMC4606174 DOI: 10.4251/wjgo.v7.i10.184] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 07/10/2015] [Accepted: 09/16/2015] [Indexed: 02/05/2023] Open
Abstract
Vitamin A and its derivatives, retinoids, have been widely studied for their use as cancer chemotherapeutic agents. With respect to colorectal cancer (CRC), several critical mutations dysregulate pathways implicated in progression and metastasis, resulting in aberrant Wnt/β-catenin signaling, gain-of-function mutations in K-ras and phosphatidylinositol-3-kinase/Akt, cyclooxygenase-2 over-expression, reduction of peroxisome proliferator-activated receptor γ activation, and loss of p53 function. Dysregulation leads to increased cellular proliferation and invasion and decreased cell-cell interaction and differentiation. Retinoids affect these pathways by various mechanisms, many involving retinoic acid receptors (RAR). RAR bind to all-trans-retinoic acid (ATRA) to induce the transcription of genes responsible for cellular differentiation. Although most research concerning the chemotherapeutic efficacy of retinoids focuses on the ability of ATRA to decrease cancer cell proliferation, increase differentiation, or promote apoptosis; as CRC progresses, RAR expression is often lost, rendering treatment of CRCs with ATRA ineffective. Our laboratory focuses on the ability of dietary vitamin A to decrease CRC cell proliferation and invasion via RAR-independent pathways. This review discusses our research and others concerning the ability of retinoids to ameliorate the defective signaling pathways listed above and decrease tumor cell proliferation and invasion through both RAR-dependent and RAR-independent mechanisms.
Collapse
|
48
|
Wu ZC, Wang X, Wei JC, Li BB, Shao DH, Li YM, Liu K, Shi YY, Zhou B, Qiu YF, Ma ZY. Antiviral activity of doxycycline against vesicular stomatitis virus in vitro. FEMS Microbiol Lett 2015; 362:fnv195. [PMID: 26459887 DOI: 10.1093/femsle/fnv195] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2015] [Indexed: 12/17/2022] Open
Abstract
Doxycycline (Dox) is a tetracycline derivative with broad-spectrum antimicrobial activities that is used as an effector substance in inducible gene-expression systems. We investigated the antiviral activity of Dox against vesicular stomatitis virus (VSV) infection in cultured H1299 cells. Dox at concentrations of 1.0-2.0 μg ml(-1) significantly inhibited VSV replication and the VSV-induced cytopathic effect in dose-dependent manners, suggesting that Dox may have broader activity in inhibiting viral replication, in addition to its well-defined bacteriostatic activity. Dox exerted its antiviral effect at the early-mid stage of VSV infection, suggesting that it did not interfere with VSV infectivity, adsorption, or entry into target cells. These results indicate that Dox can inhibit VSV infection and may therefore have potential applications for the treatment of viral infections.
Collapse
Affiliation(s)
- Zhuan-Chang Wu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 518, Ziyue road, Shanghai 200241, PR China
| | - Xin Wang
- College of Life Science, Linyi University, Shuangling Road, Linyi City, Shandong 276005, China
| | - Jian-Chao Wei
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 518, Ziyue road, Shanghai 200241, PR China
| | - Bei-Bei Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 518, Ziyue road, Shanghai 200241, PR China
| | - Dong-Hua Shao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 518, Ziyue road, Shanghai 200241, PR China
| | - Yu-Ming Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 518, Ziyue road, Shanghai 200241, PR China
| | - Ke Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 518, Ziyue road, Shanghai 200241, PR China
| | - Yuan-Yuan Shi
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 518, Ziyue road, Shanghai 200241, PR China
| | - Bin Zhou
- Key Laboratory of Animal Diseases Diagnosis and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Ya-Feng Qiu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 518, Ziyue road, Shanghai 200241, PR China
| | - Zhi-Yong Ma
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, No. 518, Ziyue road, Shanghai 200241, PR China
| |
Collapse
|
49
|
BAC transgenic mice provide evidence that p53 expression is highly regulated in vivo. Cell Death Dis 2015; 6:e1878. [PMID: 26379189 PMCID: PMC4650433 DOI: 10.1038/cddis.2015.224] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 05/11/2015] [Accepted: 05/20/2015] [Indexed: 12/17/2022]
Abstract
p53 is an important tumor suppressor and stress response mediator. Proper control of p53 level and activity is tightly associated with its function. Posttranslational modifications and the interactions with Mdm2 and Mdm4 are major mechanisms controlling p53 activity and stability. As p53 protein is short-lived and hardly detectable in unstressed situations, less is known on its basal level expression and the corresponding controlling mechanisms in vivo. In addition, it also remains obscure how p53 expression might contribute to its functional regulation. In this study, we established bacterial artificial chromosome transgenic E.coli β-galactosidase Z gene reporter mice to monitor p53 expression in mouse tissues and identify important regulatory elements critical for the expression in vivo. We revealed preferentially high level of p53 reporter expressions in the proliferating, but not the differentiated compartments of the majority of tissues during development and tissue homeostasis. In addition, tumors as well as regenerating tissues in the p53 reporter mice also expressed high level of β-gal. Furthermore, both the enhancer box sequence (CANNTG) in the p53 promoter and the 3′ terminal untranslated region element were critical in mediating the high-level expression of the reporter. We also provided evidence that cellular myelocytomatosis oncogene was a critical player regulating p53 mRNA expression in proliferating cells and tissues. Finally, we found robust p53 activation preferentially in the proliferating compartment of mouse tissues upon DNA damage and the proliferating cells exhibited an enhanced p53 response as compared with cells in a quiescent state. Together, these results suggested a highly regulated expression pattern of p53 in the proliferating compartment controlled by both transcriptional and posttranscriptional mechanisms, and such regulated p53 expression may impose functional significance upon stress by setting up a precautionary mode in defense of cellular transformation and tumorigenesis.
Collapse
|
50
|
Rajmani RS, Gandham RK, Gupta SK, Sahoo AP, Singh PK, Saxena S, Kumar R, Chaturvedi U, Tiwari AK. Administration of IκB-kinase inhibitor PS1145 enhances apoptosis in DMBA-induced tumor in male Wistar rats. Cell Biol Int 2015; 39:1317-28. [DOI: 10.1002/cbin.10510] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 06/30/2015] [Indexed: 11/11/2022]
Affiliation(s)
- R. S. Rajmani
- Molecular Biology Laboratory; Indian Veterinary Research Institute; Izatnagar, Bareilly Uttar Pradesh India
| | - Ravi Kumar Gandham
- Molecular Biology Laboratory; Indian Veterinary Research Institute; Izatnagar, Bareilly Uttar Pradesh India
| | - Shishir Kumar Gupta
- Molecular Biology Laboratory; Indian Veterinary Research Institute; Izatnagar, Bareilly Uttar Pradesh India
| | - Aditya P. Sahoo
- Molecular Biology Laboratory; Indian Veterinary Research Institute; Izatnagar, Bareilly Uttar Pradesh India
| | - Prafull Kumar Singh
- Molecular Biology Laboratory; Indian Veterinary Research Institute; Izatnagar, Bareilly Uttar Pradesh India
| | - Shikha Saxena
- Molecular Biology Laboratory; Indian Veterinary Research Institute; Izatnagar, Bareilly Uttar Pradesh India
| | - Rajiv Kumar
- Molecular Biology Laboratory; Indian Veterinary Research Institute; Izatnagar, Bareilly Uttar Pradesh India
| | - Uttara Chaturvedi
- Molecular Biology Laboratory; Indian Veterinary Research Institute; Izatnagar, Bareilly Uttar Pradesh India
| | - Ashok K. Tiwari
- Molecular Biology Laboratory; Indian Veterinary Research Institute; Izatnagar, Bareilly Uttar Pradesh India
| |
Collapse
|