1
|
Hao J, Zhao Y, Liu X, Xu H, Liu L, Wang H, Lan Y, Ji L. Whole-transcriptome sequencing reveals the effects of acupuncture on early embryos post-IVF-ET in poor ovarian response. J Ovarian Res 2025; 18:91. [PMID: 40307905 PMCID: PMC12044900 DOI: 10.1186/s13048-025-01682-7] [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: 10/03/2024] [Accepted: 04/23/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Poor ovarian response (POR) refers to a pathological condition where the ovaries respond inadequately to gonadotropin stimulation during ovulation induction. The decline in both the quantity and quality of available oocytes makes achieving pregnancy through IVF-ET more challenging. Several studies have demonstrated that acupuncture can improve embryo quality, increase the number of high-quality embryos, and enhance pregnancy outcomes in IVF-ET patients, although the underlying mechanisms remain unclear. Therefore, this study aimed to investigate the molecular mechanisms by which acupuncture influences early embryo development in POR mice after IVF-ET through whole-transcriptome sequencing. METHODS We established POR mice model and performed acupuncture treatment. Fresh denuded oocytes were retrieved and transplanted via IVF-ET into new donor female mice to obtain early-stage embryo tissues. Vaginal smear tests were conducted to monitor estrous cycle changes, while oocyte retrieval, ovarian wet weight, and ovarian index were assessed. Serum concentrations of anti-Müllerian hormone (AMH), follicle-stimulating hormone (FSH), estradiol (E2), and luteinizing hormone (LH) were measured via ELISA. Histopathological changes and apoptosis in the ovaries were evaluated using HE and TUNEL staining. Whole-transcriptome sequencing was employed to establish expression profiles of differentially expressed mRNAs (DEmRNAs), DEmiRNAs, DElncRNAs, and DEcircRNAs. The circ/lncRNA-miRNA-mRNA network was constructed to analyze the biological functions and potential mechanisms of acupuncture on early embryo development post-IVF-ET in POR mice. RESULTS Our results demonstrated that acupuncture improved ovarian function in POR mice, corrected serum hormone imbalances, and alleviated abnormal apoptosis in ovarian granulosa cells. Through whole-transcriptome sequencing, we identified 685 DEmRNAs, 13 DEmiRNAs, 325 DElncRNAs, and 4 DEcircRNAs exhibiting regulatory trends. By constructing the circ/lncRNA-miRNA-mRNA network, we found that miR-291a-3p, miR-294-3p, and miR-295-3p may serve as key targets influencing early embryo development via multiple pathways, including the Toll-like receptor signaling pathway and p53 signaling pathway. CONCLUSION Overall, our study is the first to reveal the molecular mechanisms by which acupuncture improves early embryo development under POR conditions, providing new evidence for the use of acupuncture in assisted reproduction.
Collapse
Affiliation(s)
- Jianheng Hao
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China
- The Second Clinical College, Shanxi University of Chinese Medicine, Jinzhong, 030619, Shanxi, China
| | - Yuemeng Zhao
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China
| | - Xuan Liu
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China
| | - Huichao Xu
- The Second Clinical College, Shanxi University of Chinese Medicine, Jinzhong, 030619, Shanxi, China
| | - Liying Liu
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China
| | - Haijun Wang
- The Second Clinical College, Shanxi University of Chinese Medicine, Jinzhong, 030619, Shanxi, China
| | - Ying Lan
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China.
- Chengdu University of Traditional Chinese Medicine Affiliated Hospital, Chengdu, 610075, Sichuan, China.
| | - Laixi Ji
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China.
- The Second Clinical College, Shanxi University of Chinese Medicine, Jinzhong, 030619, Shanxi, China.
| |
Collapse
|
2
|
Alonso-Olivares H, Marques MM, Prieto-Colomina A, López-Ferreras L, Martínez-García N, Vázquez-Jiménez A, Borrell V, Marin MC, Fernandez-Alonso R. Mouse cortical organoids reveal key functions of p73 isoforms: TAp73 governs the establishment of the archetypical ventricular-like zones while DNp73 is central in the regulation of neural cell fate. Front Cell Dev Biol 2024; 12:1464932. [PMID: 39376628 PMCID: PMC11456701 DOI: 10.3389/fcell.2024.1464932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 09/04/2024] [Indexed: 10/09/2024] Open
Abstract
Introduction Neurogenesis is tightly regulated in space and time, ensuring the correct development and organization of the central nervous system. Critical regulators of brain development and morphogenesis in mice include two members of the p53 family: p53 and p73. However, dissecting the in vivo functions of these factors and their various isoforms in brain development is challenging due to their pleiotropic effects. Understanding their role, particularly in neurogenesis and brain morphogenesis, requires innovative experimental approaches. Methods To address these challenges, we developed an efficient and highly reproducible protocol to generate mouse brain organoids from pluripotent stem cells. These organoids contain neural progenitors and neurons that self-organize into rosette-like structures resembling the ventricular zone of the embryonic forebrain. Using this model, we generated organoids from p73-deficient mouse cells to investigate the roles of p73 and its isoforms (TA and DNp73) during brain development. Results and Discussion Organoids derived from p73-deficient cells exhibited increased neuronal apoptosis and reduced neural progenitor proliferation, linked to compensatory activation of p53. This closely mirrors previous in vivo observations, confirming that p73 plays a pivotal role in brain development. Further dissection of p73 isoforms function revealed a dual role of p73 in regulating brain morphogenesis, whereby TAp73 controls transcriptional programs essential for the establishment of the neurogenic niche structure, while DNp73 is responsible for the precise and timely regulation of neural cell fate. These findings highlight the distinct roles of p73 isoforms in maintaining the balance of neural progenitor cell biology, providing a new understanding of how p73 regulates brain morphogenesis.
Collapse
Affiliation(s)
- Hugo Alonso-Olivares
- Instituto de Biomedicina and Departamento de Biología Molecular, Universidad de León, León, Spain
| | - Margarita M. Marques
- Instituto de Desarrollo Ganadero y Sanidad Animal and Departamento de Producción Animal, Universidad de León, León, Spain
| | - Anna Prieto-Colomina
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, Alicante, Spain
| | - Lorena López-Ferreras
- Instituto de Biomedicina and Departamento de Biología Molecular, Universidad de León, León, Spain
| | - Nicole Martínez-García
- Instituto de Biomedicina and Departamento de Producción Animal, Universidad de León, León, Spain
| | - Alberto Vázquez-Jiménez
- Instituto de Biomedicina and Departamento de Biología Molecular, Universidad de León, León, Spain
| | - Victor Borrell
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, Alicante, Spain
| | - Maria C. Marin
- Instituto de Biomedicina and Departamento de Biología Molecular, Universidad de León, León, Spain
| | - Rosalia Fernandez-Alonso
- Instituto de Biomedicina and Departamento de Biología Molecular, Universidad de León, León, Spain
| |
Collapse
|
3
|
Neagu AN, Whitham D, Bruno P, Arshad A, Seymour L, Morrissiey H, Hukovic AI, Darie CC. Onco-Breastomics: An Eco-Evo-Devo Holistic Approach. Int J Mol Sci 2024; 25:1628. [PMID: 38338903 PMCID: PMC10855488 DOI: 10.3390/ijms25031628] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/21/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
Known as a diverse collection of neoplastic diseases, breast cancer (BC) can be hyperbolically characterized as a dynamic pseudo-organ, a living organism able to build a complex, open, hierarchically organized, self-sustainable, and self-renewable tumor system, a population, a species, a local community, a biocenosis, or an evolving dynamical ecosystem (i.e., immune or metabolic ecosystem) that emphasizes both developmental continuity and spatio-temporal change. Moreover, a cancer cell community, also known as an oncobiota, has been described as non-sexually reproducing species, as well as a migratory or invasive species that expresses intelligent behavior, or an endangered or parasite species that fights to survive, to optimize its features inside the host's ecosystem, or that is able to exploit or to disrupt its host circadian cycle for improving the own proliferation and spreading. BC tumorigenesis has also been compared with the early embryo and placenta development that may suggest new strategies for research and therapy. Furthermore, BC has also been characterized as an environmental disease or as an ecological disorder. Many mechanisms of cancer progression have been explained by principles of ecology, developmental biology, and evolutionary paradigms. Many authors have discussed ecological, developmental, and evolutionary strategies for more successful anti-cancer therapies, or for understanding the ecological, developmental, and evolutionary bases of BC exploitable vulnerabilities. Herein, we used the integrated framework of three well known ecological theories: the Bronfenbrenner's theory of human development, the Vannote's River Continuum Concept (RCC), and the Ecological Evolutionary Developmental Biology (Eco-Evo-Devo) theory, to explain and understand several eco-evo-devo-based principles that govern BC progression. Multi-omics fields, taken together as onco-breastomics, offer better opportunities to integrate, analyze, and interpret large amounts of complex heterogeneous data, such as various and big-omics data obtained by multiple investigative modalities, for understanding the eco-evo-devo-based principles that drive BC progression and treatment. These integrative eco-evo-devo theories can help clinicians better diagnose and treat BC, for example, by using non-invasive biomarkers in liquid-biopsies that have emerged from integrated omics-based data that accurately reflect the biomolecular landscape of the primary tumor in order to avoid mutilating preventive surgery, like bilateral mastectomy. From the perspective of preventive, personalized, and participatory medicine, these hypotheses may help patients to think about this disease as a process governed by natural rules, to understand the possible causes of the disease, and to gain control on their own health.
Collapse
Affiliation(s)
- Anca-Narcisa Neagu
- Laboratory of Animal Histology, Faculty of Biology, “Alexandru Ioan Cuza” University of Iași, Carol I bvd. 20A, 700505 Iasi, Romania
| | - Danielle Whitham
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA; (D.W.); (P.B.); (A.A.); (L.S.); (H.M.); (A.I.H.)
| | - Pathea Bruno
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA; (D.W.); (P.B.); (A.A.); (L.S.); (H.M.); (A.I.H.)
| | - Aneeta Arshad
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA; (D.W.); (P.B.); (A.A.); (L.S.); (H.M.); (A.I.H.)
| | - Logan Seymour
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA; (D.W.); (P.B.); (A.A.); (L.S.); (H.M.); (A.I.H.)
| | - Hailey Morrissiey
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA; (D.W.); (P.B.); (A.A.); (L.S.); (H.M.); (A.I.H.)
| | - Angiolina I. Hukovic
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA; (D.W.); (P.B.); (A.A.); (L.S.); (H.M.); (A.I.H.)
| | - Costel C. Darie
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, NY 13699-5810, USA; (D.W.); (P.B.); (A.A.); (L.S.); (H.M.); (A.I.H.)
| |
Collapse
|
4
|
Xu Y, Yang X, Xiong Q, Han J, Zhu Q. The dual role of p63 in cancer. Front Oncol 2023; 13:1116061. [PMID: 37182132 PMCID: PMC10174455 DOI: 10.3389/fonc.2023.1116061] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 04/13/2023] [Indexed: 05/16/2023] Open
Abstract
The p53 family is made up of three transcription factors: p53, p63, and p73. These proteins are well-known regulators of cell function and play a crucial role in controlling various processes related to cancer progression, including cell division, proliferation, genomic stability, cell cycle arrest, senescence, and apoptosis. In response to extra- or intracellular stress or oncogenic stimulation, all members of the p53 family are mutated in structure or altered in expression levels to affect the signaling network, coordinating many other pivotal cellular processes. P63 exists as two main isoforms (TAp63 and ΔNp63) that have been contrastingly discovered; the TA and ΔN isoforms exhibit distinguished properties by promoting or inhibiting cancer progression. As such, p63 isoforms comprise a fully mysterious and challenging regulatory pathway. Recent studies have revealed the intricate role of p63 in regulating the DNA damage response (DDR) and its impact on diverse cellular processes. In this review, we will highlight the significance of how p63 isoforms respond to DNA damage and cancer stem cells, as well as the dual role of TAp63 and ΔNp63 in cancer.
Collapse
Affiliation(s)
- Yongfeng Xu
- Abdominal Oncology Ward, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Xiaojuan Yang
- Abdominal Oncology Ward, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Qunli Xiong
- Abdominal Oncology Ward, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Junhong Han
- State Key Laboratory of Biotherapy and Cancer Center, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Qing Zhu, ; Junhong Han,
| | - Qing Zhu
- Abdominal Oncology Ward, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
- *Correspondence: Qing Zhu, ; Junhong Han,
| |
Collapse
|
5
|
Koley T, Chowdhury SR, Kushwaha T, Kumar M, Inampudi KK, Kaur P, Singh TP, Viadiu H, Ethayathulla AS. Deciphering the mechanism of p73 recognition of p53 response elements using the crystal structure of p73-DNA complexes and computational studies. Int J Biol Macromol 2022; 206:40-50. [PMID: 35217090 DOI: 10.1016/j.ijbiomac.2022.02.108] [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: 11/06/2021] [Revised: 02/01/2022] [Accepted: 02/17/2022] [Indexed: 11/05/2022]
Abstract
P73 belongs to p53 family transcription factor activating more than 50% of cell fate p53 target genes involved in cell cycle, apoptosis, DNA damage response alongside neuronal system development and differentiation by binding to 20-bp response elements (REs) having sequence motif (PPPCA/T-T/AGYYY) where P-purines and Y-pyrimidines with each 10-bp separated by minimum 0 to 13-bp spacer. The promiscuous nature of recognizing both cell fate and development genes and the underlying RE selectivity mechanism by p73 is not well understood. Here, we report the molecular details of p73 recognizing the REs using the crystal structure of p73 DNA binding domain (DBD) in complex with 12 base pair DNA sequence 5'-cAGGCATGCCTg-3' and molecular dynamics simulations with six different p53 natural promoter sequences. Each 20-base pair natural promoter forms a different major/minor groove due to the presence of nucleotides A/T, A/C, G/G, T/T and G/T at positions 3, 8, 13, 18 uniquely recognized by p73 key residues Lys138 and Arg268. The loops L1 and L3 bearing these residues influence inter-and intra-dimer interfaces interactions and hence p73 forms a unique tetramer with each natural promoter sequence. Structural features of the DNA and the spacing between half-sites influence p73 tetramerization and its transactivation function.
Collapse
Affiliation(s)
- Tirthankar Koley
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Sanghati Roy Chowdhury
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Tushar Kushwaha
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Manoj Kumar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | | | - Punit Kaur
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Tej Pal Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Héctor Viadiu
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | | |
Collapse
|
6
|
Compagnucci C, Martinus K, Griffin J, Depew MJ. Programmed Cell Death Not as Sledgehammer but as Chisel: Apoptosis in Normal and Abnormal Craniofacial Patterning and Development. Front Cell Dev Biol 2021; 9:717404. [PMID: 34692678 PMCID: PMC8531503 DOI: 10.3389/fcell.2021.717404] [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: 05/30/2021] [Accepted: 07/28/2021] [Indexed: 12/22/2022] Open
Abstract
Coordination of craniofacial development involves an complex, intricate, genetically controlled and tightly regulated spatiotemporal series of reciprocal inductive and responsive interactions among the embryonic cephalic epithelia (both endodermal and ectodermal) and the cephalic mesenchyme — particularly the cranial neural crest (CNC). The coordinated regulation of these interactions is critical both ontogenetically and evolutionarily, and the clinical importance and mechanistic sensitivity to perturbation of this developmental system is reflected by the fact that one-third of all human congenital malformations affect the head and face. Here, we focus on one element of this elaborate process, apoptotic cell death, and its role in normal and abnormal craniofacial development. We highlight four themes in the temporospatial elaboration of craniofacial apoptosis during development, namely its occurrence at (1) positions of epithelial-epithelial apposition, (2) within intra-epithelial morphogenesis, (3) during epithelial compartmentalization, and (4) with CNC metameric organization. Using the genetic perturbation of Satb2, Pbx1/2, Fgf8, and Foxg1 as exemplars, we examine the role of apoptosis in the elaboration of jaw modules, the evolution and elaboration of the lambdoidal junction, the developmental integration at the mandibular arch hinge, and the control of upper jaw identity, patterning and development. Lastly, we posit that apoptosis uniquely acts during craniofacial development to control patterning cues emanating from core organizing centres.
Collapse
Affiliation(s)
- Claudia Compagnucci
- Institute for Cell and Neurobiology, Center for Anatomy, Charité Universitätsmedizin Berlin, CCO, Berlin, Germany.,Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, Rome, Italy.,Department of Craniofacial Development, King's College London, London, United Kingdom
| | - Kira Martinus
- Institute for Cell and Neurobiology, Center for Anatomy, Charité Universitätsmedizin Berlin, CCO, Berlin, Germany
| | - John Griffin
- Department of Craniofacial Development, King's College London, London, United Kingdom.,School of Biological Sciences, University of East Anglia, Norwich, United Kingdom
| | - Michael J Depew
- Institute for Cell and Neurobiology, Center for Anatomy, Charité Universitätsmedizin Berlin, CCO, Berlin, Germany.,Department of Craniofacial Development, King's College London, London, United Kingdom
| |
Collapse
|
7
|
Jaiswal SK, Raj S, DePamphilis ML. Developmental Acquisition of p53 Functions. Genes (Basel) 2021; 12:genes12111675. [PMID: 34828285 PMCID: PMC8622856 DOI: 10.3390/genes12111675] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/14/2021] [Accepted: 10/21/2021] [Indexed: 12/12/2022] Open
Abstract
Remarkably, the p53 transcription factor, referred to as “the guardian of the genome”, is not essential for mammalian development. Moreover, efforts to identify p53-dependent developmental events have produced contradictory conclusions. Given the importance of pluripotent stem cells as models of mammalian development, and their applications in regenerative medicine and disease, resolving these conflicts is essential. Here we attempt to reconcile disparate data into justifiable conclusions predicated on reports that p53-dependent transcription is first detected in late mouse blastocysts, that p53 activity first becomes potentially lethal during gastrulation, and that apoptosis does not depend on p53. Furthermore, p53 does not regulate expression of genes required for pluripotency in embryonic stem cells (ESCs); it contributes to ESC genomic stability and differentiation. Depending on conditions, p53 accelerates initiation of apoptosis in ESCs in response to DNA damage, but cell cycle arrest as well as the rate and extent of apoptosis in ESCs are p53-independent. In embryonic fibroblasts, p53 induces cell cycle arrest to allow repair of DNA damage, and cell senescence to prevent proliferation of cells with extensive damage.
Collapse
Affiliation(s)
- Sushil K. Jaiswal
- National Institute of Child Health and Human Development, Bethesda, MD 20892, USA;
- National Human Genome Research Institute, Bethesda, MD 20892, USA
| | - Sonam Raj
- National Cancer Institute, Bethesda, MD 20892, USA;
| | - Melvin L. DePamphilis
- National Institute of Child Health and Human Development, Bethesda, MD 20892, USA;
- Correspondence:
| |
Collapse
|
8
|
Type 1 Diabetes Mellitus and the First Trimester Placenta: Hyperglycemia-Induced Effects on Trophoblast Proliferation, Cell Cycle Regulators, and Invasion. Int J Mol Sci 2021; 22:ijms222010989. [PMID: 34681648 PMCID: PMC8539403 DOI: 10.3390/ijms222010989] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 02/07/2023] Open
Abstract
Type 1 diabetes mellitus (T1DM) is associated with reduced fetal growth in early pregnancy, but a contributing role of the placenta has remained elusive. Thus, we investigated whether T1DM alters placental development in the first trimester. Using a protein array, the level of 60 cell-cycle-related proteins was determined in human first trimester placental tissue (gestational week 5–11) from control (n = 11) and T1DM pregnancies (n = 12). Primary trophoblasts (gestational week 7–12, n = 32) were incubated in the absence (control) or presence of hyperglycemia (25 mM D-glucose) and hyperosmolarity (5.5 mM D-glucose + 19.5 mM D-mannitol). We quantified the number of viable and dead trophoblasts (CASY Counter) and assessed cell cycle distribution (FACS) and trophoblast invasion using a transwell assay. T1DM was associated with a significant (p < 0.05) downregulation of Ki67 (−26%), chk1 (−25%), and p73 (−26%). The number of viable trophoblasts was reduced under hyperglycemia (−23%) and hyperosmolarity (−18%), whereas trophoblast invasion was increased only under hyperglycemia (+6%). Trophoblast cell death and cell cycle distribution remained unaffected. Collectively, our data demonstrate that hyperglycemia decreases trophoblast proliferation as a potential contributing factor to the reduced placental growth in T1DM in vivo.
Collapse
|
9
|
Fish EW, Tucker SK, Peterson RL, Eberhart JK, Parnell SE. Loss of tumor protein 53 protects against alcohol-induced facial malformations in mice and zebrafish. Alcohol Clin Exp Res 2021; 45:1965-1979. [PMID: 34581462 DOI: 10.1111/acer.14688] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 07/26/2021] [Accepted: 07/30/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Alcohol exposure during the gastrulation stage of development causes the craniofacial and brain malformations that define fetal alcohol syndrome. These malformations, such as a deficient philtrum, are exemplified by a loss of midline tissue and correspond, at least in part, to regionally selective cell death in the embryo. The tumor suppressor protein Tp53 is an important mechanism for cell death, but the role of Tp53 in the consequences of alcohol exposure during the gastrulation stage has yet to be examined. The current studies used mice and zebrafish to test whether genetic loss of Tp53 is a conserved mechanism to protect against the effects of early developmental stage alcohol exposure. METHODS Female mice, heterozygous for a mutation in the Tp53 gene, were mated with Tp53 heterozygous males, and the resulting embryos were exposed during gastrulation on gestational day 7 (GD 7) to alcohol (two maternal injections of 2.9 g/kg, i.p., 4 h apart) or a vehicle control. Zebrafish mutants or heterozygotes for the tp53zdf1 M214K mutation and their wild-type controls were exposed to alcohol (1.5% or 2%) beginning 6 h postfertilization (hpf), the onset of gastrulation. RESULTS Examination of GD 17 mice revealed that eye defects were the most common phenotype among alcohol-exposed fetuses, occurring in nearly 75% of the alcohol-exposed wild-type fetuses. Tp53 gene deletion reduced the incidence of eye defects in both the heterozygous and mutant fetuses (to about 35% and 20% of fetuses, respectively) and completely protected against alcohol-induced facial malformations. Zebrafish (4 days postfertilization) also demonstrated alcohol-induced reductions of eye size and trabeculae length that were less common and less severe in tp53 mutants, indicating a protective effect of tp53 deletion. CONCLUSIONS These results identify an evolutionarily conserved role of Tp53 as a pathogenic mechanism for alcohol-induced teratogenesis.
Collapse
Affiliation(s)
- Eric W Fish
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Scott K Tucker
- Department of Molecular Biosciences, Waggoner Center for Alcohol and Addiction Research and Institute for Neuroscience, University of Texas, Austin, Texas, USA
| | - Rachel L Peterson
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Johann K Eberhart
- Department of Molecular Biosciences, Waggoner Center for Alcohol and Addiction Research and Institute for Neuroscience, University of Texas, Austin, Texas, USA
| | - Scott E Parnell
- Bowles Center for Alcohol Studies, University of North Carolina, Chapel Hill, North Carolina, USA.,Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA.,Carolina Institute for Developmental Disabilities, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
10
|
López-Ferreras L, Martínez-García N, Maeso-Alonso L, Martín-López M, Díez-Matilla Á, Villoch-Fernandez J, Alonso-Olivares H, Marques MM, Marin MC. Deciphering the Nature of Trp73 Isoforms in Mouse Embryonic Stem Cell Models: Generation of Isoform-Specific Deficient Cell Lines Using the CRISPR/Cas9 Gene Editing System. Cancers (Basel) 2021; 13:cancers13133182. [PMID: 34202306 PMCID: PMC8268375 DOI: 10.3390/cancers13133182] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/13/2021] [Accepted: 06/17/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary The Trp73 gene is involved in the regulation of multiple biological processes such as response to stress, differentiation and tissue architecture. This gene gives rise to structurally different N and C-terminal isoforms which lead to differences in its biological activity in a cell type dependent manner. However, there is a current lack of physiological models to study these isoforms. The aim of this study was to generate specific p73-isoform-deficient mouse embryonic stem cell lines using the CRISPR/Cas9 system. Their special features, self-renewal and pluripotency, make embryonic stem cells a useful research tool that allows the generation of cells from any of the three germ layers carrying specific inactivation of p73-isoforms. Characterization of the generated cell lines indicates that while the individual elimination of TA- or DN-p73 isoform is compatible with pluripotency, it results in alterations of the transcriptional profiles and the pluripotent state of the embryonic stem cells in an isoform-specific manner. Abstract The p53 family has been widely studied for its role in various physiological and pathological processes. Imbalance of p53 family proteins may contribute to developmental abnormalities and pathologies in humans. This family exerts its functions through a profusion of isoforms that are generated by different promoter usage and alternative splicing in a cell type dependent manner. In particular, the Trp73 gene gives rise to TA and DN-p73 isoforms that confer p73 a dual nature. The biological relevance of p73 does not only rely on its tumor suppression effects, but on its pivotal role in several developmental processes. Therefore, the generation of cellular models that allow the study of the individual isoforms in a physiological context is of great biomedical relevance. We generated specific TA and DN-p73-deficient mouse embryonic stem cell lines using the CRISPR/Cas9 gene editing system and validated them as physiological bona fide p73-isoform knockout models. Global gene expression analysis revealed isoform-specific alterations of distinctive transcriptional networks. Elimination of TA or DN-p73 is compatible with pluripotency but prompts naïve pluripotent stem cell transition into the primed state, compromising adequate lineage differentiation, thus suggesting that differential expression of p73 isoforms acts as a rheostat during early cell fate determination.
Collapse
Affiliation(s)
- Lorena López-Ferreras
- Instituto de Biomedicina (IBIOMED), Universidad de León, 24071 León, Spain; (L.L.-F.); (N.M.-G.); (L.M.-A.); (M.M.-L.); (Á.D.-M.); (J.V.-F.); (H.A.-O.)
- Departamento de Biología Molecular, Universidad de León, 24071 León, Spain
| | - Nicole Martínez-García
- Instituto de Biomedicina (IBIOMED), Universidad de León, 24071 León, Spain; (L.L.-F.); (N.M.-G.); (L.M.-A.); (M.M.-L.); (Á.D.-M.); (J.V.-F.); (H.A.-O.)
- Departamento de Producción Animal, Universidad de León, 24071 León, Spain
| | - Laura Maeso-Alonso
- Instituto de Biomedicina (IBIOMED), Universidad de León, 24071 León, Spain; (L.L.-F.); (N.M.-G.); (L.M.-A.); (M.M.-L.); (Á.D.-M.); (J.V.-F.); (H.A.-O.)
- Departamento de Biología Molecular, Universidad de León, 24071 León, Spain
| | - Marta Martín-López
- Instituto de Biomedicina (IBIOMED), Universidad de León, 24071 León, Spain; (L.L.-F.); (N.M.-G.); (L.M.-A.); (M.M.-L.); (Á.D.-M.); (J.V.-F.); (H.A.-O.)
- Biomar Microbial Technologies, Parque Tecnológico de León, Armunia, 24009 León, Spain
| | - Ángela Díez-Matilla
- Instituto de Biomedicina (IBIOMED), Universidad de León, 24071 León, Spain; (L.L.-F.); (N.M.-G.); (L.M.-A.); (M.M.-L.); (Á.D.-M.); (J.V.-F.); (H.A.-O.)
| | - Javier Villoch-Fernandez
- Instituto de Biomedicina (IBIOMED), Universidad de León, 24071 León, Spain; (L.L.-F.); (N.M.-G.); (L.M.-A.); (M.M.-L.); (Á.D.-M.); (J.V.-F.); (H.A.-O.)
- Departamento de Biología Molecular, Universidad de León, 24071 León, Spain
| | - Hugo Alonso-Olivares
- Instituto de Biomedicina (IBIOMED), Universidad de León, 24071 León, Spain; (L.L.-F.); (N.M.-G.); (L.M.-A.); (M.M.-L.); (Á.D.-M.); (J.V.-F.); (H.A.-O.)
- Departamento de Biología Molecular, Universidad de León, 24071 León, Spain
| | - Margarita M. Marques
- Departamento de Producción Animal, Universidad de León, 24071 León, Spain
- Instituto de Desarrollo Ganadero y Sanidad Animal (INDEGSAL), Universidad de León, 24071 León, Spain
- Correspondence: (M.M.M.); (M.C.M.); Tel.: +34-987-291757 (M.M.M.); +34-987-291490 (M.C.M.)
| | - Maria C. Marin
- Instituto de Biomedicina (IBIOMED), Universidad de León, 24071 León, Spain; (L.L.-F.); (N.M.-G.); (L.M.-A.); (M.M.-L.); (Á.D.-M.); (J.V.-F.); (H.A.-O.)
- Departamento de Biología Molecular, Universidad de León, 24071 León, Spain
- Correspondence: (M.M.M.); (M.C.M.); Tel.: +34-987-291757 (M.M.M.); +34-987-291490 (M.C.M.)
| |
Collapse
|
11
|
Tan J, Chen SX, Lei QY, Yi SQ, Wu N, Wang YL, Xiao ZJ, Wu H. Mitochonic acid 5 regulates mitofusin 2 to protect microglia. Neural Regen Res 2021; 16:1813-1820. [PMID: 33510088 PMCID: PMC8328753 DOI: 10.4103/1673-5374.306094] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Microglial apoptosis is associated with neuroinflammation and no effective strategies are currently available to protect microglia against inflammation-induced apoptosis. Mouse microglial BV-2 cells (5 × 106) were incubated with 10 μg/mL lipopolysaccharides for 12 hours to mimic an inflammatory environment. Then the cells were co-cultured with mitochonic acid 5 (MA-5) for another 12 hours. MA-5 improved the survival of lipopolysaccharide-exposed cells. MA-5 decreased the activity of caspase-3, which is associated with apoptosis. MA-5 reduced the number of terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling-positive cells, and increased adenosine triphosphate levels in cells. MA-5 decreased the open state of the mitochondrial permeability transition pore and reduced calcium overload and diffusion of second mitochondria-derived activator of caspase (Smac). MA-5 decreased the expression of apoptosis-related proteins (mitochondrial Smac, cytoplasmic Smac, pro-caspase-3, cleaved-caspase-3, and caspase-9), and increased the levels of anti-apoptotic proteins (Bcl2 and X-linked inhibitor of apoptosis protein), mitochondria-related proteins (mitochondrial fusion protein 2, mitochondrial microtubule-associated proteins 1A/1B light chain 3B II), and autophagy-related proteins (Beclin1, p62 and autophagy related 5). However, MA-5 did not promote mitochondrial homeostasis or decrease microglial apoptosis when Mitofusin 2 expression was silenced. This shows that MA-5 increased Mitofusin 2-related mitophagy, reversed cellular energy production and maintained energy metabolism in BV-2 cells in response to lipopolysaccharide-induced inflammation. These findings indicate that MA-5 may promote the survival of microglial cells via Mitofusin 2-related mitophagy in response to lipopolysaccharide-induced inflammation.
Collapse
Affiliation(s)
- Jian Tan
- Department of Neurology, The First Affiliated Hospital of University of South China, Hengyang, Hunan Province, China
| | - Shuang-Xi Chen
- Department of Neurology, The First Affiliated Hospital of University of South China, Hengyang, Hunan Province, China
| | - Qing-Yun Lei
- Department of Neurology, The First Affiliated Hospital of University of South China, Hengyang, Hunan Province, China
| | - Shan-Qing Yi
- Department of Neurology, The First Affiliated Hospital of University of South China, Hengyang, Hunan Province, China
| | - Na Wu
- Department of Neurology, The First Affiliated Hospital of University of South China, Hengyang, Hunan Province, China
| | - Yi-Lin Wang
- Department of Neurology, The First Affiliated Hospital of University of South China, Hengyang, Hunan Province, China
| | - Zi-Jian Xiao
- Department of Neurology, The First Affiliated Hospital of University of South China, Hengyang, Hunan Province, China
| | - Heng Wu
- Department of Neurology, The First Affiliated Hospital of University of South China, Hengyang, Hunan Province, China
| |
Collapse
|
12
|
Bai X, Tang J. Myrcene Exhibits Antitumor Activity Against Lung Cancer Cells by Inducing Oxidative Stress and Apoptosis Mechanisms. Nat Prod Commun 2020. [DOI: 10.1177/1934578x20961189] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Myrcene, a natural olefinic hydrocarbon, possesses anti-inflammatory, analgesic, antibiotic, and antimutagenic properties, but its anticancer effect has not yet been elucidated. Hence, the present study was framed to investigate the molecular mechanism by which myrcene mediates the anticancer activity of A549 lung adenocarcinoma cells. In vitro, A549 lung cancer cells were cultured either with or without myrcene, and the effects on cellular metabolic activity, levels of reactive oxygen species (ROS), mitochondrial integrity, deoxyribonucleic acid (DNA) damage, and activity of caspases were analyzed. The study demonstrated that compared with control cells, myrcene induces cell death in a dose-dependent manner while inducing ROS levels. Further experiments revealed that the metabolic activity of the A549 lung adenocarcinoma cells was diminished with increased DNA damage and altered cellular integrity. In addition, increased activity of caspase-3 was also evidenced with reduced mitochondrial membrane potential synthesis in the myrcene-treated cells, which demonstrate that lung cancer cells experience signs of toxicity during myrcene treatment through the activation of the apoptosis mechanism via mitochondria-mediated cell death signaling and induction of oxidative stress. The results provide the first report on the evidence of anticancer activity and the possibility of a new drug that could be used for the treatment of lung cancer.
Collapse
Affiliation(s)
- Xudong Bai
- Department of Thoracic Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, P. R. China
| | - Jin Tang
- Department of Ultrasound, Shanxi Provincial Cancer Hospital, Taiyuan, P. R. China
| |
Collapse
|
13
|
Geng C, Wei J, Wu C. Yap-Hippo pathway regulates cerebral hypoxia-reoxygenation injury in neuroblastoma N2a cells via inhibiting ROCK1/F-actin/mitochondrial fission pathways. Acta Neurol Belg 2020; 120:879-892. [PMID: 29796942 DOI: 10.1007/s13760-018-0944-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 05/15/2018] [Indexed: 01/03/2023]
Abstract
Yes-associated protein (Yap), a regulator of cellular apoptosis, has been demonstrated to be involved in cerebral ischemia-reperfusion (IR) injury through poorly defined mechanisms. The present study aimed to explore the role of Yap in regulating cerebral IR injury in vitro, with a focus on mitochondrial fission and ROCK1/F-actin pathways. Our data demonstrated that Yap was actually downregulated in N2a cells after cerebral hypoxia-reoxygenation (HR) injury, and that lower expression of Yap was closely associated with increased cell death. However, the reintroduction of Yap was able to suppress the HR-mediated N2a cells death via blocking the mitochondria-related apoptotic signal. At the molecular levels, Yap overexpression sustained mitochondrial potential, normalized the mitochondrial respiratory function, reduced ROS overproduction, limited HtrA2/Omi release from mitochondria into the nucleus, and suppressed pro-apoptotic proteins activation. Subsequently, functional studies have further illustrated that HR-mediated mitochondrial apoptosis was highly regulated by mitochondrial fission, whereas Yap overexpression was able to attenuate HR-mediated mitochondrial fission and, thus, promote N2a cell survival in the context of HR injury. At last, we demonstrated that Yap handled mitochondrial fission via closing ROCK1/F-actin signaling pathways. Activation of ROCK1/F-actin pathways abrogated the protective role of Yap overexpression on mitochondrial homeostasis and N2a cell survival in the setting of HR injury. Altogether, our data identified Yap as the endogenous defender to relieve HR-mediated nerve damage via antagonizing ROCK1/F-actin/mitochondrial fission pathways.
Collapse
Affiliation(s)
- Chizi Geng
- Physician of Neurology Department, Beijing Luhe Hospital, Capital Medical University, Beijing, China.
| | - Jianchao Wei
- Director of Neurology Department, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Chengsi Wu
- Deputy Director of Eurology Department, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
14
|
Hilliard SA, Li Y, Dixon A, El-Dahr SS. Mdm4 controls ureteric bud branching via regulation of p53 activity. Mech Dev 2020; 163:103616. [PMID: 32464196 DOI: 10.1016/j.mod.2020.103616] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 05/19/2020] [Accepted: 05/21/2020] [Indexed: 02/07/2023]
Abstract
The antagonism between Mdm2 and its close homolog Mdm4 (also known as MdmX) and p53 is vital for embryogenesis and organogenesis. Previously, we demonstrated that targeted disruption of Mdm2 in the Hoxb7+ ureteric bud (Ub) lineage, which gives rise to the renal collecting system, causes renal hypodysplasia culminating in perinatal lethality. In this study, we examine the unique role of Mdm4 in establishing the collecting duct system of the murine kidney. Hoxb7Cre driven loss of Mdm4 in the Ub lineage (UbMdm4-/-) disrupts branching morphogenesis and triggers UB cell apoptosis. UbMdm4-/- kidneys exhibit abnormally dilated Ub tips while the medulla is hypoplastic. These structural alterations result in secondary depletion of nephron progenitors and nascent nephrons. As a result, newborn UbMdm4-/- mice have hypo-dysplastic kidneys. Transcriptional profiling revealed downregulation of the Ret-tyrosine kinase pathway components, Gdnf, Wnt11, Sox8, Etv4 and Cxcr4 in the UbMdm4-/- mice relative to controls. Moreover, the expression levels of the canonical Wnt signaling members Axin2 and Wnt9b are downregulated. Mdm4 deletion upregulated p53 activity and p53-target gene expression including Cdkn1a (p21), Gdf15, Ccng1, PERP, and Fas. Germline loss of p53 in UbMdm4-/- mice largely rescues kidney development and terminal differentiation of the collecting duct. We conclude that Mdm4 plays a unique and vital role in Ub branching morphogenesis and collecting system development.
Collapse
Affiliation(s)
- Sylvia A Hilliard
- Tulane University School of Medicine, Department of Pediatrics, Section of Pediatric Nephrology, New Orleans, LA 70112, United States of America
| | - Yuwen Li
- Tulane University School of Medicine, Department of Pediatrics, Section of Pediatric Nephrology, New Orleans, LA 70112, United States of America
| | - Angelina Dixon
- Tulane University School of Medicine, Department of Pediatrics, Section of Pediatric Nephrology, New Orleans, LA 70112, United States of America
| | - Samir S El-Dahr
- Tulane University School of Medicine, Department of Pediatrics, Section of Pediatric Nephrology, New Orleans, LA 70112, United States of America.
| |
Collapse
|
15
|
Istiqomah MA, Hasibuan PAZ, Sumaiyah S, Yusraini E, Oku H, Basyuni M. Anticancer Effects of Polyisoprenoid From Nypa fruticans Leaves by Controlling Expression of p53, EGFR, PI3K, AKT1, and mTOR Genes in Colon Cancer (WiDr) Cells. Nat Prod Commun 2020. [DOI: 10.1177/1934578x20918412] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The current research seeks to examine the anticancer effect of polyisoprenoids from mangrove palm, Nypa fruticans, leaves in WiDr cells by analyzing the cell cycles of cancer and regulating the expression of p53, epidermal growth factor receptor (EGFR), PI3K, AKT1, and mammalian target of rapamycin (mTOR) genes by using the reverse transcription-polymerase chain reaction (RT-PCR). An inhibited cell cycle analysis was conducted using the flow cytometry, and the upregulation or downregulation of the expression of p53, EGFR, PI3K, AKT1, and mTOR genes was obtained using RT-PCR. The data were then statistically analyzed using one-way analysis of variance by a post hoc test, a parametric statistical analysis using Tukey’s honest significant difference. Polyisoprenoids in N. fruticans extracts worked as chemotheraupetic in the G0-G1 cycle is 79.0%, however, with positive control 5-fluorouracil as 88.1% and are carried out by the specific upregulation of the expression of the p53 gene and the downregulation of the expression of the EGFR, PI3K, AKT1, and mTOR genes. This study can also explain the significant pharmacological properties of the leaves of the species N. fruticans that work specifically in the G0-G1 phase to upregulate the expression of the p53 gene and downregulate the expression of the EGFR, PI3K, AKT1, and mTOR genes. This study also revealed polyisoprenoid (100% dolichol), which blocked the growth and development of WiDr colon cancer cells.
Collapse
Affiliation(s)
| | | | - Sumaiyah Sumaiyah
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| | - Era Yusraini
- Department of Food Science and Technology, Faculty of Agriculture, Universitas Sumatera Utara, Medan, North Sumatra, Indonesia
| | - Hirosuke Oku
- Molecular Biotechnology Group, Tropical Biosphere Research Center, University of the Ryukyus, Nishihara, Okinawa, Japan
| | - Mohammad Basyuni
- Department of Forestry, Faculty of Forestry, Universitas Sumatera Utara, Medan, Indonesia
- Center of Excellence for Mangrove, Universitas Sumatera Utara, Medan, Indonesia
| |
Collapse
|
16
|
Xu T, Guo P, He Y, Pi C, Wang Y, Feng X, Hou Y, Jiang Q, Zhao L, Wei Y. Application of curcumin and its derivatives in tumor multidrug resistance. Phytother Res 2020; 34:2438-2458. [PMID: 32255545 DOI: 10.1002/ptr.6694] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 02/27/2020] [Accepted: 03/22/2020] [Indexed: 12/16/2022]
Abstract
Malignant tumor endangers seriously the health of all mankind. Multidrug resistance (MDR) is one of the main causes of clinical tumor chemotherapy failure. Curcumin (CUR) has not only antitumor activity but also reversing tumor MDR effect. CUR reverses tumor MDR via regulating related signal pathways or corresponding expressed proteins or gene. When combined with chemotherapeutic agents, CUR can be a chemotherapeutic sensitive agent to enhance chemotherapy efficacy and weaken tumor MDR. On the other hand, to improve the MDR reversal effect of CUR, its derivatives have been extensively studied. Therefore, this article mainly focuses on reviewing the application of CUR and its derivatives in MDR and its mechanism of reversing MDR.
Collapse
Affiliation(s)
- Ting Xu
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, People's Republic of China
| | - Pu Guo
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, People's Republic of China
| | - Yingmeng He
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, People's Republic of China
| | - Chao Pi
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, People's Republic of China
| | - Yuanyuan Wang
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, People's Republic of China
| | - Xianhu Feng
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, People's Republic of China
| | - Yi Hou
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, People's Republic of China
| | - Qingsheng Jiang
- School of International Education, Southwest Medical University, Luzhou, Sichuan, People's Republic of China
| | - Ling Zhao
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, People's Republic of China
| | - Yumeng Wei
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, People's Republic of China
| |
Collapse
|
17
|
Ciuffoli V, Lena AM, Gambacurta A, Melino G, Candi E. Myoblasts rely on TAp63 to control basal mitochondria respiration. Aging (Albany NY) 2019; 10:3558-3573. [PMID: 30487319 PMCID: PMC6286837 DOI: 10.18632/aging.101668] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 11/15/2018] [Indexed: 12/15/2022]
Abstract
p53, with its family members p63 and p73, have been shown to promote myoblast differentiation by regulation of the function of the retinoblastoma protein and by direct activation of p21Cip/Waf1 and p57Kip2, promoting cell cycle exit. In previous studies, we have demonstrated that the TAp63γ isoform is the only member of the p53 family that accumulates during in vitro myoblasts differentiation, and that its silencing led to delay in myotube fusion. To better dissect the role of TAp63γ in myoblast physiology, we have generated both sh-p63 and Tet-On inducible TAp63γ clones. Gene array analysis of sh-p63 C2C7 clones showed a significant modulation of genes involved in proliferation and cellular metabolism. Indeed, we found that sh-p63 C2C7 myoblasts present a higher proliferation rate and that, conversely, TAp63γ ectopic expression decreases myoblasts proliferation, indicating that TAp63γ specifically contributes to myoblasts proliferation, independently of p53 and p73. In addition, sh-p63 cells have a defect in mitochondria respiration highlighted by a reduction in spare respiratory capacity and a decrease in complex I, IV protein levels. These results demonstrated that, beside contributing to cell cycle exit, TAp63γ participates to myoblasts metabolism control.
Collapse
Affiliation(s)
- Veronica Ciuffoli
- Department of Experimental Medicine and TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Anna Maria Lena
- Department of Experimental Medicine and TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Alessandra Gambacurta
- Department of Experimental Medicine and TOR, University of Rome "Tor Vergata", Rome, Italy
| | - Gerry Melino
- Department of Experimental Medicine and TOR, University of Rome "Tor Vergata", Rome, Italy.,MRC-Toxicology Unit, University of Cambridge, Cambridge, UK
| | - Eleonora Candi
- Department of Experimental Medicine and TOR, University of Rome "Tor Vergata", Rome, Italy.,IDI-IRCCS, Biochemistry laboratory, Rome, Italy
| |
Collapse
|
18
|
Smirnov A, Cappello A, Lena AM, Anemona L, Mauriello A, Di Daniele N, Annicchiarico-Petruzzelli M, Melino G, Candi E. ZNF185 is a p53 target gene following DNA damage. Aging (Albany NY) 2019; 10:3308-3326. [PMID: 30446632 PMCID: PMC6286825 DOI: 10.18632/aging.101639] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 11/01/2018] [Indexed: 12/17/2022]
Abstract
The transcription factor p53 is a key player in the tumour suppressive DNA damage response and a growing number of target genes involved in these pathways has been identified. p53 has been shown to be implicated in controlling cell motility and its mutant form enhances metastasis by loss of cell directionality, but the p53 role in this context has not yet being investigated. Here, we report that ZNF185, an actin cytoskeleton-associated protein from LIM-family of Zn-finger proteins, is induced following DNA-damage. ChIP-seq analysis, chromatin crosslinking immune-precipitation experiments and luciferase assays demonstrate that ZNF185 is a bona fide p53 target gene. Upon genotoxic stress, caused by DNA-damaging drug etoposide and UVB irradiation, ZNF185 expression is up-regulated and in etoposide-treated cells, ZNF185 depletion does not affect cell proliferation and apoptosis, but interferes with actin cytoskeleton remodelling and cell polarization. Bioinformatic analysis of different types of epithelial cancers from both TCGA and GTEx databases showed a significant decrease in ZNF185 mRNA level compared to normal tissues. These findings are confirmed by tissue micro-array IHC staining. Our data highlight the involvement of ZNF185 and cytoskeleton changes in p53-mediated cellular response to genotoxic stress and indicate ZNF185 as potential biomarker for epithelial cancer diagnosis.
Collapse
Affiliation(s)
- Artem Smirnov
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome 00133, Italy
| | - Angela Cappello
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome 00133, Italy
| | - Anna Maria Lena
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome 00133, Italy
| | - Lucia Anemona
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome 00133, Italy
| | - Alessandro Mauriello
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome 00133, Italy
| | - Nicola Di Daniele
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome 00133, Italy
| | | | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome 00133, Italy.,MRC-Toxicology Unit, University of Cambridge, Cambridge, UK
| | - Eleonora Candi
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome 00133, Italy.,Istituto Dermopatico dell'Immacolata-IRCCS, Rome 00163, Italy
| |
Collapse
|
19
|
Frezza V, Fierro C, Gatti E, Peschiaroli A, Lena AM, Petruzzelli MA, Candi E, Anemona L, Mauriello A, Pelicci PG, Melino G, Bernassola F. ΔNp63 promotes IGF1 signalling through IRS1 in squamous cell carcinoma. Aging (Albany NY) 2019; 10:4224-4240. [PMID: 30594912 PMCID: PMC6326668 DOI: 10.18632/aging.101725] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 12/12/2018] [Indexed: 02/07/2023]
Abstract
Accumulating evidence has proved that deregulation of ΔNp63 expression plays an oncogenic role in head and neck squamous cell carcinomas (HNSCCs). Besides p63, the type 1-insulin-like growth factor (IGF) signalling pathway has been implicated in HNSCC development and progression. Most insulin/IGF1 signalling converges intracellularly onto the protein adaptor insulin receptor substrate-1 (IRS-1) that transmits signals from the receptor to downstream effectors, including the PI3K/AKT and the MAPK kinase pathways, which, ultimately, promote proliferation, invasion, and cell survival. Here we report that p63 directly controls IRS1 transcription and cellular abundance and fosters the PI3K/AKT and MAPK downstream signalling pathways. Inactivation of ΔNp63 expression indeed reduces tumour cell responsiveness to IGF1 stimulation, and inhibits the growth potential of HNSCC cells. In addition, a positive correlation was observed between p63 and IRS1 expression in human HNSCC tissue arrays and in publicly available gene expression data. Our findings indicate that aberrant expression of ΔNp63 in HNSSC may act as an oncogenic stimulus by altering the IGF signalling pathway.
Collapse
Affiliation(s)
- Valentina Frezza
- Department of Experimental Medicine, TOR University of Rome "Tor Vergata", Rome 00133, Italy
| | - Claudia Fierro
- Department of Experimental Medicine, TOR University of Rome "Tor Vergata", Rome 00133, Italy
| | - Elena Gatti
- Department of Experimental Oncology European Institute of Oncology, Milan 20139, Italy
| | - Angelo Peschiaroli
- National Research Council of Italy Institute of Translational Pharmacology (IFT-CNR), Rome 00133, Italy
| | - Anna Maria Lena
- Department of Experimental Medicine, TOR University of Rome "Tor Vergata", Rome 00133, Italy
| | | | - Eleonora Candi
- Department of Experimental Medicine, TOR University of Rome "Tor Vergata", Rome 00133, Italy.,Istituto Dermopatico dell'Immacolata, IRCCS,, Rome 00163, Italy
| | - Lucia Anemona
- Department of Experimental Medicine, TOR University of Rome "Tor Vergata", Rome 00133, Italy
| | - Alessandro Mauriello
- Department of Experimental Medicine, TOR University of Rome "Tor Vergata", Rome 00133, Italy
| | - Pier Giuseppe Pelicci
- Department of Experimental Oncology European Institute of Oncology, Milan 20139, Italy
| | - Gerry Melino
- Department of Experimental Medicine, TOR University of Rome "Tor Vergata", Rome 00133, Italy.,Medical Research Council, Toxicology Unit, University of Cambridge, Leicester LE1 9HN, UK
| | - Francesca Bernassola
- Department of Experimental Medicine, TOR University of Rome "Tor Vergata", Rome 00133, Italy
| |
Collapse
|
20
|
The Emerging Landscape of p53 Isoforms in Physiology, Cancer and Degenerative Diseases. Int J Mol Sci 2019; 20:ijms20246257. [PMID: 31835844 PMCID: PMC6941119 DOI: 10.3390/ijms20246257] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/26/2019] [Accepted: 12/09/2019] [Indexed: 12/13/2022] Open
Abstract
p53, first described four decades ago, is now established as a master regulator of cellular stress response, the “guardian of the genome”. p53 contributes to biological robustness by behaving in a cellular-context dependent manner, influenced by several factors (e.g., cell type, active signalling pathways, the type, extent and intensity of cellular damage, cell cycle stage, nutrient availability, immune function). The p53 isoforms regulate gene transcription and protein expression in response to the stimuli so that the cell response is precisely tuned to the cell signals and cell context. Twelve isoforms of p53 have been described in humans. In this review, we explore the interactions between p53 isoforms and other proteins contributing to their established cellular functions, which can be both tumour-suppressive and oncogenic in nature. Evidence of p53 isoform in human cancers is largely based on RT-qPCR expression studies, usually investigating a particular type of isoform. Beyond p53 isoform functions in cancer, it is implicated in neurodegeneration, embryological development, progeroid phenotype, inflammatory pathology, infections and tissue regeneration, which are described in this review.
Collapse
|
21
|
Massa V, Avagliano L, Grazioli P, De Castro SCP, Parodi C, Savery D, Vergani P, Cuttin S, Doi P, Bulfamante G, Copp AJ, Greene NDE. Dynamic acetylation profile during mammalian neurulation. Birth Defects Res 2019; 112:205-211. [PMID: 31758757 PMCID: PMC7004172 DOI: 10.1002/bdr2.1618] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/25/2019] [Accepted: 11/03/2019] [Indexed: 01/17/2023]
Abstract
BACKGROUND Neural tube defects (NTDs) result from failure of neural tube closure during embryogenesis. These severe birth defects of the central nervous system include anencephaly and spina bifida, and affect 0.5-2 per 1,000 pregnancies worldwide in humans. It has been demonstrated that acetylation plays a pivotal role during neural tube closure, as animal models for defective histone acetyltransferase proteins display NTDs. Acetylation represents an important component of the complex network of posttranslational regulatory interactions, suggesting a possible fundamental role during primary neurulation events. This study aimed to assess protein acetylation contribution to early patterning of the central nervous system both in human and murine specimens. METHODS We used both human and mouse (Cited2 -/- ) samples to analyze the dynamic acetylation of proteins during embryo development through immunohistochemistry, western blot analysis and quantitative polymerase chain reaction. RESULTS We report the dynamic profile of histone and protein acetylation status during neural tube closure. We also report a rescue effect in an animal model by chemical p53 inhibition. CONCLUSIONS Our data suggest that the p53-acetylation equilibrium may play a role in primary neurulation in mammals.
Collapse
Affiliation(s)
- Valentina Massa
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Laura Avagliano
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Paolo Grazioli
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Sandra C P De Castro
- UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Chiara Parodi
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Dawn Savery
- UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Patrizia Vergani
- Department of Obstetrics and Gynaecology, Foundation MBBM, University of Milano-Bicocca, Monza, Italy
| | - Serena Cuttin
- Department of Pathology, San Gerardo Hospital, University of Milano-Bicocca, Monza, Italy
| | - Patrizia Doi
- Department of Health Sciences, University of Milan, Milan, Italy
| | | | - Andrew J Copp
- UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Nicholas D E Greene
- UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| |
Collapse
|
22
|
Miller MW. p53-Mediated Activities in NS-5 Neural Stem Cells: Effects of Ethanol. Alcohol Clin Exp Res 2019; 43:655-667. [PMID: 30748015 DOI: 10.1111/acer.13976] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 02/05/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Transforming growth factor (TGF) β1 and ethanol (EtOH) powerfully inhibit the proliferation, DNA repair, and survival of neural stem cells (NSCs). The present study tests the hypothesis that the EtOH-induced DNA damage response is mediated through p53 pathways and influenced by growth factor signals. METHODS Cultures of nonimmortalized NSCs, NS-5 cells, were transfected with p53 siRNA, exposed to either the mitogenic fibroblast growth factor (FGF) 2 or antimitogenic TGFβ1, and to EtOH. Stage-specific cellular and genomic responses were examined. RESULTS p53 status, EtOH exposure, and growth factor significantly affected the expression of transcripts related to the DNA damage response (including those coding for excision repair proteins), mitotic promoters, and regulators of cell death via the tumor necrosis factor pathway. There were significant compensatory increases in p53 family members, p63 and p73, notably in regard to the regulation of cell cycle restriction and apoptosis. Treatment with p53 siRNA potentiated EtOH- and TGFβ1-induced changes in the numbers of proliferating NSCs and increased the proportion of NSCs expressing the apoptotic marker annexin V. CONCLUSIONS Thus, it appears that EtOH and TGFβ1 affect proliferation, DNA repair, and survival of NSCs via p53-mediated activities.
Collapse
Affiliation(s)
- Michael W Miller
- Department of Neuroscience and Physiology, State University of New York-Upstate Medical University, Syracuse, New York.,Touro College of Osteopathic Medicine, Middletown, New York.,Research Service, Veterans Affairs Medical Center, Syracuse, New York
| |
Collapse
|
23
|
Marques MM, Villoch-Fernandez J, Maeso-Alonso L, Fuertes-Alvarez S, Marin MC. The Trp73 Mutant Mice: A Ciliopathy Model That Uncouples Ciliogenesis From Planar Cell Polarity. Front Genet 2019; 10:154. [PMID: 30930930 PMCID: PMC6428764 DOI: 10.3389/fgene.2019.00154] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 02/13/2019] [Indexed: 12/21/2022] Open
Abstract
p73 transcription factor belongs to one of the most important gene families in vertebrate biology, the p53-family. Trp73 gene, like the other family members, generates multiple isoforms named TA and DNp73, with different and, sometimes, antagonist functions. Although p73 shares many biological functions with p53, it also plays distinct roles during development. Trp73 null mice (p73KO from now on) show multiple phenotypes as gastrointestinal and cranial hemorrhages, rhinitis and severe central nervous system defects. Several groups, including ours, have revisited the apparently unrelated phenotypes observed in total p73KO and revealed a novel p73 function in the organization of ciliated epithelia in brain and trachea, but also an essential role as regulator of ependymal planar cell polarity. Unlike p73KO or TAp73KO mice, tumor-prone Trp53−/− mice (p53KO) do not present ependymal ciliary or planar cell polarity defects, indicating that regulation of ciliogenesis and PCP is a p73-specific function. Thus, loss of ciliary biogenesis and epithelial organization might be a common underlying cause of the diverse p73KO-phenotypes, highlighting Trp73 role as an architect of the epithelial tissue. In this review we would like to discuss the data regarding p73 role as regulator of ependymal cell ciliogenesis and PCP, supporting the view of the Trp73-mutant mice as a model that uncouples ciliogenesis from PCP and a possible model of human congenital hydrocephalus.
Collapse
Affiliation(s)
- Margarita M Marques
- Departamento de Producción Animal, Laboratorio de Diferenciación Celular y Diseño de Modelos Celulares, Instituto de Desarrollo Ganadero y Sanidad Animal, Universidad de León, León, Spain
| | - Javier Villoch-Fernandez
- Departamento de Biología Molecular, Laboratorio de Diferenciación Celular y Diseño de Modelos Celulares, Instituto de Biomedicina, Universidad de León, León, Spain
| | - Laura Maeso-Alonso
- Departamento de Biología Molecular, Laboratorio de Diferenciación Celular y Diseño de Modelos Celulares, Instituto de Biomedicina, Universidad de León, León, Spain
| | - Sandra Fuertes-Alvarez
- Departamento de Biología Molecular, Laboratorio de Diferenciación Celular y Diseño de Modelos Celulares, Instituto de Biomedicina, Universidad de León, León, Spain
| | - Maria C Marin
- Departamento de Biología Molecular, Laboratorio de Diferenciación Celular y Diseño de Modelos Celulares, Instituto de Biomedicina, Universidad de León, León, Spain
| |
Collapse
|
24
|
Sheng J, Li H, Dai Q, Lu C, Xu M, Zhang J, Feng J. DUSP1 recuses diabetic nephropathy via repressing JNK-Mff-mitochondrial fission pathways. J Cell Physiol 2019; 234:3043-3057. [PMID: 30191967 DOI: 10.1002/jcp.27124] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 07/05/2018] [Indexed: 12/21/2022]
Abstract
Excessive mitochondrial fission has been identified as the pathogenesis of diabetic nephropathy (DN), although the upstream regulatory signal for mitochondrial fission activation in the setting of DN remains unknown. In the current study, we found that dual-specificity protein phosphatase-1 (DUSP1) was actually downregulated by chronic hyperglycemia stimulus. Lower DUSP1 expression was associated with glucose metabolism disorder, renal dysfunction, kidney hypertrophy, renal fibrosis, and glomerular apoptosis. At the molecular level, defective DUSP1 expression activated JNK pathway, and the latter selectively opened mitochondrial fission by modulating mitochondrial fission factor (Mff) phosphorylation. Excessive Mff-related mitochondrial fission evoked mitochondrial oxidative stress, promoted mPTP opening, exacerbated proapoptotic protein leakage into the cytoplasm, and finally initiated mitochondria-dependent cellular apoptosis in the setting of diabetes. However, overexpression of DUSP1 interrupted Mff-related mitochondrial fission, reducing hyperglycemia-mediated mitochondrial damage and thus improving renal function. Overall, we have shown that DUSP1 functions as a novel malefactor in diabetic renal damage that mediates via modifying Mff-related mitochondrial fission. Thus, finding strategies to regulate the balance of the DUSP1-JNK-Mff signaling pathway and mitochondrial homeostasis may be a therapeutic target for treating diabetic nephropathy in clinical practice.
Collapse
Affiliation(s)
- Junqin Sheng
- Department of Nephrology, Xuhui District Central Hospital of Shanghai, Shanghai, China
| | - Hongyan Li
- Department of Nephrology, Huadu District People's Hospital, Southern Medical University, Guangzhou, China
| | - Qin Dai
- Department of Nephrology, Xuhui District Central Hospital of Shanghai, Shanghai, China
| | - Chang Lu
- Department of Nephrology, Xuhui District Central Hospital of Shanghai, Shanghai, China
| | - Min Xu
- Department of Nephrology, Xuhui District Central Hospital of Shanghai, Shanghai, China
| | - Jisheng Zhang
- Department of Nephrology, Xuhui District Central Hospital of Shanghai, Shanghai, China
| | - Jianxun Feng
- Department of Nephrology, Xuhui District Central Hospital of Shanghai, Shanghai, China
| |
Collapse
|
25
|
Chen Z, Wang C, Yu N, Si L, Zhu L, Zeng A, Liu Z, Wang X. INF2 regulates oxidative stress-induced apoptosis in epidermal HaCaT cells by modulating the HIF1 signaling pathway. Biomed Pharmacother 2019; 111:151-161. [DOI: 10.1016/j.biopha.2018.12.046] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 12/10/2018] [Accepted: 12/12/2018] [Indexed: 12/15/2022] Open
|
26
|
Atypical GATA transcription factor TRPS1 represses gene expression by recruiting CHD4/NuRD(MTA2) and suppresses cell migration and invasion by repressing TP63 expression. Oncogenesis 2018; 7:96. [PMID: 30563971 PMCID: PMC6299095 DOI: 10.1038/s41389-018-0108-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 10/30/2018] [Accepted: 11/26/2018] [Indexed: 01/10/2023] Open
Abstract
Transcriptional repressor GATA binding 1 (TRPS1), an atypical GATA transcription factor, functions as a transcriptional repressor and is also implicated in human cancers. However, the underlying mechanism of TRPS1 contributing to malignancy remains obscure. In the current study, we report that TRPS1 recognizes both gene proximal and distal transcription start site (TSS) sequences to repress gene expression. Co-IP mass spectrometry and biochemical studies showed that TRPS1 binds to CHD4/NuRD(MTA2). Genome-wide and molecular studies revealed that CHD4/NuRD(MTA2) is required for TRPS1 transcriptional repression. Mechanically, TRPS1 and CHD4/NuRD(MTA2) form precision-guided transcriptional repression machinery in which TRPS1 guides the machinery to specific target sites by recognizing GATA elements, and CHD4/NuRD(MTA2) represses the transcription of target genes. Furthermore, TP63 was identified and validated to be a direct target of TRPS1-CHD4/NuRD(MTA2) complex, which represses TP63 expression by involving decommission of TP63 enhancer in the described precision-guided manner, leading to a reduction of the ΔNp63 level and contributing to migration and invasion of cancer cells.
Collapse
|
27
|
Rotblat B, Agostini M, Niklison-Chirou MV, Amelio I, Willis AE, Melino G. Sustained protein synthesis and reduced eEF2K levels in TAp73 -\- mice brain: a possible compensatory mechanism. Cell Cycle 2018; 17:2637-2643. [PMID: 30507330 DOI: 10.1080/15384101.2018.1553341] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The transcription factor p73 is a member of the p53 family, of which the transactivation domain containing isoform (TAp73) plays key roles in brain development and neuronal stem cells. TAp73 also facilitates homoeostasis and prevents oxidative damage in vivo by inducing the expression of its target genes. Recently, we found that in addition to its role in regulation of transcription, TAp73 also affects mRNA translation. In cultured cells, acute TAp73 depletion activates eEF2K, which phosphorylates eEF2 reducing mRNA translation elongation. As a consequence, there is a reduction in global proteins synthesis rates and reprogramming of the translatome, leading to a selective decrease in the translation of rRNA processing factors. Given the dramatic effects of Tap73 depletion in vitro it was important to determine whether similar effects were observed in vivo. Here, we report the surprising finding that in brains of TAp73 KO mice there is a reduced level of eEF2K, which allows protein synthesis rates to be maintained suggesting a compensation model. These data provide new insights to the role of TAp73 in translation regulation and the eEF2K pathway in the brain.
Collapse
Affiliation(s)
- Barak Rotblat
- a MRC Toxicology Unit , University of Cambridge , Rome , UK.,b Department of Life Sciences , Ben Gurion University in the Negev , Beer Sheva , Israel
| | - Massimiliano Agostini
- a MRC Toxicology Unit , University of Cambridge , Rome , UK.,c Department of Experimental Medicine and Surgery, IDI-IRCCS , University of Rome Tor Vergata , Rome , Italy
| | - Maria Victoria Niklison-Chirou
- a MRC Toxicology Unit , University of Cambridge , Rome , UK.,d Blizard Institute of Cell and Molecular Science, Barts and the London School of Medicine and Dentistry , Queen Mary University of London , London , UK
| | - Ivano Amelio
- a MRC Toxicology Unit , University of Cambridge , Rome , UK
| | - Anne E Willis
- a MRC Toxicology Unit , University of Cambridge , Rome , UK
| | - Gerry Melino
- a MRC Toxicology Unit , University of Cambridge , Rome , UK.,c Department of Experimental Medicine and Surgery, IDI-IRCCS , University of Rome Tor Vergata , Rome , Italy
| |
Collapse
|
28
|
Zhang W, Liu K, Pei Y, Ma J, Tan J, Zhao J. Mst1 regulates non-small cell lung cancer A549 cell apoptosis by inducing mitochondrial damage via ROCK1/F‑actin pathways. Int J Oncol 2018; 53:2409-2422. [PMID: 30320378 PMCID: PMC6203146 DOI: 10.3892/ijo.2018.4586] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 09/14/2018] [Indexed: 02/06/2023] Open
Abstract
Mammalian STE20-like kinase 1 (Mst1) is well recognized as a major tumor suppressor in cancer development, growth, metabolic reprogramming, metastasis, cell death and recurrence. However, the roles of Mst1 in non-small cell lung cancer (NSCLC) A549 cell phenotypic alterations remain to be elucidated. The present study aimed to explore the functional role and underlying mechanisms of Mst1 with regards to A549 cell proliferation, migration and apoptosis; this study focused on mitochondrial homeostasis and Rho-associated coiled-coil containing protein kinase 1 (ROCK1)/F‑actin pathways. The results demonstrated that Mst1 was downregulated in A549 cells compared with in a normal pulmonary epithelial cell line. Subsequently, overexpression of Mst1 in A549 cells reduced cell viability and promoted cell apoptosis. Furthermore, overexpression of Mst1 suppressed A549 cell proliferation and migration. At the molecular level, the reintroduction of Mst1 in A549 cells led to activation of mitochondrial apoptosis, as evidenced by a reduction in mitochondrial potential, overproduction of ROS, cytochrome c release from the mitochondria into the nucleus, and upregulation of pro-apoptotic protein expression. In addition, Mst1 overexpression was closely associated with impaired mitochondrial respiratory function and suppressed cellular energy metabolism. Functional studies illustrated that Mst1 overexpression activated ROCK1/F-actin pathways, which highly regulate mitochondrial function. Inhibition of ROCK1/F-actin pathways in A549 cells sustained mitochondrial homeostasis, alleviated caspase-9-dependent mitochondrial apoptosis, enhanced cancer cell migration and increased cell proliferation. In conclusion, these data firmly established the regulatory role of Mst1 in NSCLC A549 cell survival via the modulation of ROCK1/F-actin pathways, which may provide opportunities for novel treatment modalities in clinical practice.
Collapse
Affiliation(s)
- Weiqiang Zhang
- Department of Thoracic Surgery, Army General Hospital of PLA, Beijing 100700, P. R. China
| | - Keiqiang Liu
- Department of Thoracic Surgery, Army General Hospital of PLA, Beijing 100700, P. R. China
| | - Yingxin Pei
- Department of Thoracic Surgery, Army General Hospital of PLA, Beijing 100700, P. R. China
| | - Jingbo Ma
- Department of Thoracic Surgery, Army General Hospital of PLA, Beijing 100700, P. R. China
| | - Jiang Tan
- Department of Thoracic Surgery, Army General Hospital of PLA, Beijing 100700, P. R. China
| | - Jing Zhao
- Department of Thoracic Surgery, Army General Hospital of PLA, Beijing 100700, P. R. China
| |
Collapse
|
29
|
Wei R, Cao J, Yao S. Matrine promotes liver cancer cell apoptosis by inhibiting mitophagy and PINK1/Parkin pathways. Cell Stress Chaperones 2018; 23:1295-1309. [PMID: 30209783 PMCID: PMC6237690 DOI: 10.1007/s12192-018-0937-7] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 08/23/2018] [Accepted: 09/02/2018] [Indexed: 02/05/2023] Open
Abstract
Matrine is a natural alkaloid isolated from the root and stem of the legume plant Sophora. Its anti-proliferative and pro-apoptotic effects on several types of cancer have been well-documented. However, the role of matrine in regulating mitochondrial homeostasis, particularly mitophagy in liver cancer apoptosis, remains uncertain. The aim of our study was to explore whether matrine promotes liver cancer cell apoptosis by modifying mitophagy. HepG2 cells were used in the study and treated with different doses of matrine. Cell viability and apoptosis were determined by MTT assay, TUNEL staining, western blotting, and LDH release assay. Mitophagy was monitored by immunofluorescence assay and western blotting. Mitochondrial function was assessed by immunofluorescence assay, ELISA, and western blotting. The results of our study indicated that matrine treatment dose-dependently reduced cell viability and increased the apoptotic rate of HepG2 cells. Functional studies demonstrated that matrine treatment induced mitochondrial dysfunction and activated mitochondrial apoptosis by inhibiting protective mitophagy. Re-activation of mitophagy abolished the pro-apoptotic effects of matrine on HepG2 cells. Molecular investigations further confirmed that matrine regulated mitophagy via the PINK1/Parkin pathways. Matrine blocked the PINK1/Parkin pathways and repressed mitophagy, whereas activation of the PINK1/Parkin pathways increased mitophagy activity and promoted HepG2 cell survival in the presence of matrine. Together, our data indicated that matrine promoted HepG2 cell apoptosis through a novel mechanism that acted via inhibiting mitophagy and the PINK1/Parkin pathways. This finding provides new insight into the molecular mechanism of matrine for treating liver cancer and offers a potential target to repress liver cancer progression by modulating mitophagy and the PINK1/Parkin pathways.
Collapse
Affiliation(s)
- Runjie Wei
- Peking University China-Japan Friendship School of Clinical Medicine, No. 2 Yinghua East Road, Chaoyang District, 100029, Beijing, China
| | - Jian Cao
- School of Biological Science and Medical Engineering, Beihang University, No. 37 Xueyuan Road, Haidian District, 100191, Beijing, China
| | - Shukun Yao
- Peking University China-Japan Friendship School of Clinical Medicine, No. 2 Yinghua East Road, Chaoyang District, 100029, Beijing, China.
- Department of Gastroenterology, China-Japan Friendship Hospital, No. 2 Yinghua East Road, Chaoyang District, 100029, Beijing, China.
| |
Collapse
|
30
|
Carbone M, Amelio I, Affar EB, Brugarolas J, Cannon-Albright LA, Cantley LC, Cavenee WK, Chen Z, Croce CM, Andrea AD, Gandara D, Giorgi C, Jia W, Lan Q, Mak TW, Manley JL, Mikoshiba K, Onuchic JN, Pass HI, Pinton P, Prives C, Rothman N, Sebti SM, Turkson J, Wu X, Yang H, Yu H, Melino G. Consensus report of the 8 and 9th Weinman Symposia on Gene x Environment Interaction in carcinogenesis: novel opportunities for precision medicine. Cell Death Differ 2018; 25:1885-1904. [PMID: 30323273 PMCID: PMC6219489 DOI: 10.1038/s41418-018-0213-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Accepted: 08/06/2018] [Indexed: 12/13/2022] Open
Abstract
The relative contribution of intrinsic genetic factors and extrinsic environmental ones to cancer aetiology and natural history is a lengthy and debated issue. Gene-environment interactions (G x E) arise when the combined presence of both a germline genetic variant and a known environmental factor modulates the risk of disease more than either one alone. A panel of experts discussed our current understanding of cancer aetiology, known examples of G × E interactions in cancer, and the expanded concept of G × E interactions to include somatic cancer mutations and iatrogenic environmental factors such as anti-cancer treatment. Specific genetic polymorphisms and genetic mutations increase susceptibility to certain carcinogens and may be targeted in the near future for prevention and treatment of cancer patients with novel molecularly based therapies. There was general consensus that a better understanding of the complexity and numerosity of G × E interactions, supported by adequate technological, epidemiological, modelling and statistical resources, will further promote our understanding of cancer and lead to novel preventive and therapeutic approaches.
Collapse
Affiliation(s)
| | | | - El Bachir Affar
- Department of Medicine, Maisonneuve-Rosemont Hospital Research Center, University of Montréal, Montréal, Quebec, H1T 2M4, Canada
| | - James Brugarolas
- Department of Internal Medicine, Hematology-Oncology Division, Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Lisa A Cannon-Albright
- Genetic Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Huntsman Cancer Institute, Salt Lake City, UT, USA
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT, USA
| | - Lewis C Cantley
- Meyer Cancer Center, Weill Cornell Medical College, 413 E. 69(th) Street, New York, NY, 10021, USA
| | - Webster K Cavenee
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA, 92093, USA
| | - Zhijian Chen
- Department of Molecular Biology and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Carlo M Croce
- Department of Molecular Virology, Immunology, and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Alan D' Andrea
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - David Gandara
- Thoracic Oncology, UC Davis, Sacramento, CA, 96817, USA
| | - Carlotta Giorgi
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Wei Jia
- Hawaii Cancer Center, Honolulu, HI, USA
| | - Qing Lan
- Occupational & Environmental Epidemiology Branch Division of Cancer Epidemiology & Genetics National Cancer Institute NIH, Bethesda, MD, USA
| | - Tak Wah Mak
- The Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Centre, Toronto, ON, M5G 2M9, Canada
| | - James L Manley
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Katsuhiko Mikoshiba
- Laboratory for Developmental Neurobiology, RIKEN Brain Science Institute, Wako, Saitama, 351-0198, Japan
| | - Jose N Onuchic
- Center for Theoretical Biological Physics, Rice University, Houston, TX, 77005, USA
| | - Harvey I Pass
- Division of General Thoracic Surgery, Department of Cardiothoracic Surgery, NYU Langone Medical Center, New York, NY, USA
| | - Paolo Pinton
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Carol Prives
- Department of Biological Sciences, Columbia University, New York, New York, 10027, USA
| | - Nathaniel Rothman
- Occupational & Environmental Epidemiology Branch Division of Cancer Epidemiology & Genetics National Cancer Institute NIH, Bethesda, MD, USA
| | - Said M Sebti
- Drug Discovery Department, Moffitt Cancer Center, and Department of Oncologic Sciences, University of South Florida, Tampa, FL, 33612, USA
| | | | - Xifeng Wu
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Gerry Melino
- MRC Toxicology Unit, Leicester, UK.
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
31
|
Xu P, Zhang G, Sha L, Hou S. RETRACTED: DUSP1 alleviates cerebral ischaemia reperfusion injury via inactivating JNK-Mff pathways and repressing mitochondrial fission. Life Sci 2018; 210:251-262. [PMID: 30138595 DOI: 10.1016/j.lfs.2018.08.049] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 08/20/2018] [Accepted: 08/20/2018] [Indexed: 01/17/2023]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal). This article has been retracted at the request of the Editor-in-Chief. The article titled “DUSP1 alleviates cerebral ischaemia reperfusion injury via inactivating JNK-Mff pathways and repressing mitochondrial fission” is a near duplicate of a previously published manuscript titled “DUSP1 alleviates cardiac ischemia/reperfusion injury by suppressing the Mff-required mitochondrial fission and Bnip3-related mitophagy via the JNK pathways. Redox Biology. 2018;14:576-587.”
Collapse
Affiliation(s)
- Peng Xu
- The Fourth Department of Geronotology, Jinan Military General Hospital, 25 Shifan Road, Tianqiao District, Jinan City, Shandong Province 250031, China
| | - Guofeng Zhang
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, 127 West Chang Le Road, Xi'an 710032, China
| | - Longgui Sha
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai 201399, China
| | - Shuangxing Hou
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai 201399, China.
| |
Collapse
|
32
|
He L, Gu K. Tanshinone IIA regulates colorectal cancer apoptosis via attenuation of Parkin‑mediated mitophagy by suppressing AMPK/Skp2 pathways. Mol Med Rep 2018; 18:1692-1703. [PMID: 29845197 DOI: 10.3892/mmr.2018.9087] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Accepted: 04/04/2018] [Indexed: 11/05/2022] Open
Abstract
Mitophagy is important for cancer development. Notably, the role of Parkin‑mediated mitophagy in colorectal cancer (CRC) mortality has not been fully determined. Therefore, the present study aimed to investigate the effect of Parkin‑mediated mitophagy on CRC apoptosis. In addition, the present study investigated the therapeutic effects of Tanshinone IIA (Tan IIA) on the regulation of CRC cell death via mitophagy. Cellular apoptosis was measured following Tan IIA treatment. In addition, mitophagy activity was evaluated by immunofluorescence and western blotting. The results of the present study revealed that Tan IIA may enhance CRC cell death. In addition, the results demonstrated that Tan IIA enhanced mitochondrial apoptosis, as demonstrated by reduced mitochondrial membrane potential, elevated mitochondrial permeability transition pore opening, and increased oxidative stress, mitochondrial energy disorder and proapoptotic factor expression. Furthermore, the results of the present study demonstrated that Tan IIA induced mitochondrial apoptosis via inhibition of mitophagy. In addition, it was revealed that mitophagy could suppress mitochondrial apoptosis. Functional assays revealed that Tan IIA suppressed the adenosine monophosphate‑activated protein kinase (AMPK) pathway, resulting in the inactivation of S‑phase kinase associated protein 2 (Skp2). Furthermore, reduced levels of Skp2 failed to activate Parkin, thus resulting in inhibition of mitophagy. Conversely, reactivation of AMPK and overexpression of Skp2 rescued mitophagy activity and thus attenuated the Tan IIA‑induced apoptosis of CRC cells. In conclusion, the results of the present study demonstrated the beneficial role of mitophagy in CRC cell survival and suggested that Tan IIA may be an effective therapeutic agent, which suppresses mitophagy activity and enhances CRC apoptosis.
Collapse
Affiliation(s)
- Lili He
- Department of Infectious Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, P.R. China
| | - Kebo Gu
- Hematology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, P.R. China
| |
Collapse
|
33
|
Li M, Yang X, Wang S. PTEN enhances nasal epithelial cell resistance to TNFα‑induced inflammatory injury by limiting mitophagy via repression of the TLR4‑JNK‑Bnip3 pathway. Mol Med Rep 2018; 18:2973-2986. [PMID: 30015897 DOI: 10.3892/mmr.2018.9264] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 05/23/2018] [Indexed: 11/05/2022] Open
Abstract
Nasal epithelial cell inflammatory injury is associated with chronic obstructive pulmonary disease development. However, the mechanism by which inflammation triggers nasal epithelial cell damage remains unclear. In the present study, tumor necrosis factor (TNF)α was used to induce an inflammatory injury and explore the underlying pathogenesis for nasal epithelial cell apoptosis in vitro, with a focus on mitochondrial homeostasis. Then, cellular apoptosis was detected via a terminal deoxynucleotidyl‑transferase‑mediated dUTP nick end labeling assay and western blotting. Mitochondrial function was evaluated via JC‑1 staining, mPTP opening measurement and western blotting. The results demonstrated that TNFα treatment induced nasal epithelial cell apoptosis, proliferation arrest and migration inhibition via downregulating phosphatase and tensin homolog (PTEN) levels. Increased PTEN expression was associated with reduce Toll‑like receptor (TLR)4‑c‑Jun kinase (JNK)‑Bcl2‑interacting protein 3 (Bnip3) pathway signaling, leading to reductions in mitophagy activity. Excessive mitophagy resulted in ATP deficiencies, mitochondrial dysfunction, caspase‑9 activation and cellular apoptosis. By contrast, PTEN overexpression in nasal epithelial cells alleviated the mitochondrial damage and cellular apoptosis via inhibiting the TLR4‑JNK‑Bnip3 pathway, favoring the survival of nasal epithelial cells under inflammatory injury. Therefore, this data uncovered a potential molecular basis for nasal epithelial cell apoptosis in response to inflammatory injury, and PTEN was identified as the endogenous defender of nasal epithelial cell survival via controlling lethal mitophagy by inhibiting the TLR4‑JNK‑Bnip3 pathway, suggesting that this pathway may be a potential target for clinically treating chronic nasal and sinus inflammatory injury.
Collapse
Affiliation(s)
- Meng Li
- Department of Chinese Medicine, Children's Hospital Affiliated to Capital Institute of Pediatrics, Beijing 100020, P.R. China
| | - Xiang Yang
- Department of Cardiac Surgery, Children's Hospital Affiliated to Capital Institute of Pediatrics, Beijing 100020, P.R. China
| | - Shouchuan Wang
- Department of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
34
|
Melatonin therapy for diabetic cardiomyopathy: A mechanism involving Syk-mitochondrial complex I-SERCA pathway. Cell Signal 2018; 47:88-100. [DOI: 10.1016/j.cellsig.2018.03.012] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 03/18/2018] [Accepted: 03/23/2018] [Indexed: 12/22/2022]
|
35
|
Wang H, Zhao X, Ni C, Dai Y, Guo Y. Zearalenone regulates endometrial stromal cell apoptosis and migration via the promotion of mitochondrial fission by activation of the JNK/Drp1 pathway. Mol Med Rep 2018; 17:7797-7806. [PMID: 29620184 DOI: 10.3892/mmr.2018.8823] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Accepted: 12/12/2017] [Indexed: 11/05/2022] Open
Abstract
Increased endometrial stromal cell (ESC) survival and migration is responsible for the development and progression of endometriosis. However, little is known about the mechanisms underlying ESC survival and migration, and limited therapeutic strategies that are able to reverse these abnormalities are available. The present study investigated the effects of zearalenone (ZEA) on ESC survival and migration, particularly focusing on mitochondrial fission and the c‑Jun N‑terminal kinase (JNK)/dynamin‑related protein 1 (Drp1) pathway. The results revealed that ZEA induced ESC apoptosis in a dose‑dependent manner. Furthermore, ZEA treatment triggered excessive mitochondrial fission resulting in structural and functional mitochondrial damage, leading to the collapse of the mitochondrial membrane potential and subsequent leakage of cytochrome c into the cytoplasm. This triggered the mitochondrial pathway of apoptosis. Additionally, ZEA‑induced mitochondrial fission decreased ESC migration through F‑actin/G‑actin homeostasis dysregulation. ZEA also increased JNK phosphorylation and subsequently Drp1 phosphorylation at the serine 616 position, resulting in Drp1 activation. JNK/Drp1 pathway inhibition abolished the inhibitory effects of ZEA on ESC survival and migration. In summary, the present study demonstrated that ZEA reduced ESC survival and migration through the stimulation of mitochondrial fission by activation of the JNK/Drp1 pathway.
Collapse
Affiliation(s)
- Huixiang Wang
- Department of Gynecology and Obstetrics, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, P.R. China
| | - Xiaoli Zhao
- Department of Pathology, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, P.R. China
| | - Chengxiang Ni
- Department of Gynecology and Obstetrics, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, P.R. China
| | - Yuyang Dai
- Department of National Institute for Drug Clinical Trial, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, P.R. China
| | - Yan Guo
- Department of Gynecology and Obstetrics, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, P.R. China
| |
Collapse
|
36
|
Zhou H, Wang J, Zhu P, Zhu H, Toan S, Hu S, Ren J, Chen Y. NR4A1 aggravates the cardiac microvascular ischemia reperfusion injury through suppressing FUNDC1-mediated mitophagy and promoting Mff-required mitochondrial fission by CK2α. Basic Res Cardiol 2018; 113:23. [DOI: 10.1007/s00395-018-0682-1] [Citation(s) in RCA: 259] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Revised: 04/09/2018] [Accepted: 04/30/2018] [Indexed: 12/22/2022]
|
37
|
Wang X, Song Q. Mst1 regulates post-infarction cardiac injury through the JNK-Drp1-mitochondrial fission pathway. Cell Mol Biol Lett 2018; 23:21. [PMID: 29760744 PMCID: PMC5941482 DOI: 10.1186/s11658-018-0085-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 04/20/2018] [Indexed: 12/13/2022] Open
Abstract
Background Post-infarction cardiac injury is closely associated with cardiac remodeling and heart dysfunction. Mammalian STE20-like kinase 1 (Mst1), a regulator of cellular apoptosis, is involved in cardiac remodeling in post-infarction heart, but the mechanisms remain poorly defined. We aimed to explore the role of Mst1 in regulating chronic post-infarction cardiac injury, with a focus on mitochondrial homoeostasis. Methods Wild-type (WT) and Mst1-knockout mice were as the cardiac myocardial infarction model. Cardiac fibrosis, myocardial inflammation response, heart dysfunction and cardiomyocyte death were measured in vivo using immunohistochemistry, immunofluorescence, western blot, qPCR and TUNEL assays. Cardiomyocytes were isolated from WT and Mst1-knockout mice, and a chronic hypoxia model was used to induce damage. Mitochondrial function was determined via JC1 staining, ROS measurement, cyt-c leakage detection and mitochondrial apoptotic pathways analysis. Mitochondrial fission was observed using immunofluorescence. A pathway activator and inhibitor were applied to establish the signaling pathways involved in regulating mitochondrial homeostasis. Results Our study demonstrated that Mst1 expression was significantly upregulated in the heart post-infarction. Activated Mst1 induced cardiac fibrosis, an excessive inflammatory response, and cardiomyocyte death, whereas the genetic ablation of Mst1 protected the myocardium against chronic post-infarction injury. Function assays showed that upregulation of Mst1 activity contributed to JNK pathway activation, which led to Drp1 migration from the cytoplasm onto the surface of the mitochondria, indicative of mitochondrial fission activation. Excessive mitochondrial fission caused mitochondrial fragmentation, resulting in mitochondrial potential collapse, ROS overproduction, mitochondrial pro-apoptotic leakage into the cytoplasm, and the initiation of caspase-9-mediated mitochondrial apoptosis. By contrast, Mst1 deletion helped to maintain mitochondrial structure and function, sending pro-survival signals to the cardiomyocytes. Conclusions Our results identify Mst1 as a malefactor in the development of post-infarction cardiac injury and that it acts through the JNK-Drp1-mitochondrial fission pathway.
Collapse
Affiliation(s)
- Xisong Wang
- Department of Critical Care Medicine, the Chinese PLA General Hospital, Beijing, China
| | - Qing Song
- Department of Critical Care Medicine, the Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
38
|
Pan L, Zhou L, Yin W, Bai J, Liu R. miR-125a induces apoptosis, metabolism disorder and migrationimpairment in pancreatic cancer cells by targeting Mfn2-related mitochondrial fission. Int J Oncol 2018; 53:124-136. [PMID: 29749475 PMCID: PMC5958665 DOI: 10.3892/ijo.2018.4380] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 04/02/2018] [Indexed: 12/13/2022] Open
Abstract
Mitochondrial fission is important for the development and progression of pancreatic cancer (PC). However, little is known regarding its role in pancreatic cancer apoptosis, metabolism and migration. In the current study, the mechanism by which mitochondrial fission modifies the biological characteristics of PC was explored. MicroRNA-125a (miR-125a) had the ability to inhibit mitochondrial fission and contributed to cellular survival. Suppressed mitochondrial fission led to a reduction in mitochondrial debris, preserved the mitochondrial membrane potential, inhibited mitochondrial permeability transition pore opening, ablated cytochrome c leakage into the cytoplasm and reduced the pro-apoptotic protein contents, finally blocking mitochondria related apoptosis pathways. Furthermore, defective mitochondrial fission induced by miR-125a enhanced mitochondria-dependent energy metabolism by promoting activity of electron transport chain complexes. Furthermore, suppressed mitochondrial fission also contributed to PANC-1 cell migration by preserving the F-actin balance. Furthermore, mitofusin 2 (Mfn2), the key defender of mitochondrial fission, is involved in inhibition of miR125a-mediated mitochondrial fission. Low contents of miR-125a upregulated Mfn2 transcription and expression, leading to inactivation of mitochondrial fission. Ultimately, the current study determined that miR-125a and Mfn2 are regulated by hypoxia-inducible factor 1 (HIF1). Knockdown of HIF1 reversed miR-125a expression, and therefore, inhibited Mfn2 expression, leading to activation of mitochondrial fission. Collectively, the present study demonstrated mitochondrial fission as a tumor suppression process that is regulated by the HIF/miR-125a/Mfn2 pathways, acting to restrict PANC-1 cell survival, energy metabolism and migration, with potential implications for novel approaches for PC therapy.
Collapse
Affiliation(s)
- Lichao Pan
- The Second Department of Hepatobiliary Surgery, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Lin Zhou
- Department of Hepatobiliary Surgery, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Weijia Yin
- Department of Biochemistry, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Jia Bai
- Department of Hepatobiliary Surgery, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Rong Liu
- The Second Department of Hepatobiliary Surgery, Chinese PLA General Hospital, Beijing 100853, P.R. China
| |
Collapse
|
39
|
Abstract
Most human cancers harbor mutations in the gene encoding p53. As a result, research on p53 in the past few decades has focused primarily on its role as a tumor suppressor. One consequence of this focus is that the functions of p53 in development have largely been ignored. However, recent advances, such as the genomic profiling of embryonic stem cells, have uncovered the significance and mechanisms of p53 functions in mammalian cell differentiation and development. As we review here, these recent findings reveal roles that complement the well-established roles for p53 in tumor suppression.
Collapse
Affiliation(s)
- Abhinav K Jain
- Department of Epigenetics and Molecular Carcinogenesis, Center for Stem Cell and Development Biology, Center for Cancer Epigenetics, The University of Texas MD Anderson UT Health Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michelle Craig Barton
- Department of Epigenetics and Molecular Carcinogenesis, Center for Stem Cell and Development Biology, Center for Cancer Epigenetics, The University of Texas MD Anderson UT Health Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
40
|
Lei Q, Tan J, Yi S, Wu N, Wang Y, Wu H. Mitochonic acid 5 activates the MAPK-ERK-yap signaling pathways to protect mouse microglial BV-2 cells against TNFα-induced apoptosis via increased Bnip3-related mitophagy. Cell Mol Biol Lett 2018; 23:14. [PMID: 29636771 PMCID: PMC5887257 DOI: 10.1186/s11658-018-0081-5] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 03/27/2018] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND The regulation of microglial function via mitochondrial homeostasis is important in the development of neuroinflammation. The underlying mechanism for this regulatory function remains unclear. In this study, we investigated the protective role of mitochonic acid 5 (MA-5) in microglial mitochondrial apoptosis following TNFα-induced inflammatory injury. METHODS TNFα was used to induce inflammatory injury in mouse microglial BV-2 cells with and without prior MA-5 treatment. Cellular apoptosis was assessed using the MTT and TUNEL assays. Mitochondrial functions were evaluated via mitochondrial membrane potential JC-1 staining, ROS flow cytometry analysis, mPTP opening assessment, and immunofluorescence of cyt-c. Mitophagy was examined using western blots and immunofluorescence. The pathways analysis was carried out using western blots and immunofluorescence with a pathway blocker. RESULTS Our results demonstrated that TNFα induced apoptosis in the microglial BV-2 cell line by activating the caspase-9-dependent mitochondrial apoptotic pathway. Mechanistically, inflammation reduced mitochondrial potential, induced ROS production, and contributed to the leakage of mitochondrial pro-apoptotic factors into the cytoplasm. The inflammatory response reduced cellular energy metabolism and increased oxidative stress. By contrast, treatment with MA-5 reduced mitochondrial apoptosis via upregulation of mitophagy. Increased mitophagy degraded damaged mitochondria, disrupting mitochondrial apoptosis, neutralizing ROS overproduction, and improving cellular energy production. We also identified that MA-5 regulated mitophagy via Bnip3 through the MAPK-ERK-Yap signaling pathway. Inhibiting this signaling pathway or knocking down Bnip3 expression prevented MA-5 from having beneficial effects on mitochondrial homeostasis and increased microglial apoptosis. CONCLUSIONS After TNFα-induced inflammatory injury, MA-5 affects microglial mitochondrial homeostasis in a manner mediated via the amplification of protective, Bnip3-related mitophagy, which is mediated via the MAPK-ERK-Yap signaling pathway.
Collapse
Affiliation(s)
- Qingyun Lei
- Department of Neurology, First Hospital Affiliated to University of South China, Hunan, China
| | - Jian Tan
- Department of Neurology, First Hospital Affiliated to University of South China, Hunan, China
| | - Shangqing Yi
- Department of Neurology, First Hospital Affiliated to University of South China, Hunan, China
| | - Na Wu
- Department of Neurology, First Hospital Affiliated to University of South China, Hunan, China
| | - Yilin Wang
- Department of Neurology, First Hospital Affiliated to University of South China, Hunan, China
| | - Heng Wu
- Department of Neurology, First Hospital Affiliated to University of South China, Hunan, China
| |
Collapse
|