1
|
Abdel-Megeed RM, Abdel-Hamid AHZ, Kadry MO. Titanium nanostructure mitigating doxorubicin-induced testicular toxicity in rats via regulating major autophagy signaling pathways. Toxicol Rep 2025; 14:101869. [PMID: 39811821 PMCID: PMC11731616 DOI: 10.1016/j.toxrep.2024.101869] [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: 10/11/2024] [Revised: 12/04/2024] [Accepted: 12/13/2024] [Indexed: 01/16/2025] Open
Abstract
Doxorubicin (DOX) is a powerful antineoplastic FDA-approved anthracycline-derived antibiotic and is considered as the most suitable intervention for solid tumors and hematological cancers therapy. However, its therapeutic application is highly limited due to acute and chronic renal, hematological and testicular toxicity. Oxidative stress, lipid peroxidation and apoptosis in germ cells as well as low sperm count, motility and disturbing steroidogenesis are the principal machineries of DOX-induced testicular toxicity. Nevertheless, the comprehensive molecular pathways responsible for DOX-induced testicular damage are not yet fully understood. The current study aims to clarify the role of autophagy and apoptotic signaling pathways in testicular toxicity induced by DOX in the rat model. The study also investigates the potential role of both titanium dioxide nanoparticles (TiO2NP) loaded with DOX and Lactoferrin in combination with DOX in mitigating testicular toxicity induced by DOX the standard antitumor drug. In the present study, male Wister albino rats were intoxicated with a total cumulative dose of DOX (18 mg/kg) via intra-peritoneal injection and served as positive control group. The other two groups administered either TiO2NP-DOX or lactoferrin-DOX. Furthermore, biochemical and molecular analyses were then performed. DOX intoxication induced testicular toxicity, revealing mineral imbalance as indicated by an increase in both calcium and magnesium concentrations. Administration of either TiO2NP-DOX or lactoferrin-DOX resulted in a significant modulation of disrupted mineral concentrations, with TiO2NP-DOX showing superiority in modulating both magnesium and calcium concentrations. Acid Phosphatase level significantly increased upon DOX-induced testicular damage. Molecular analysis of EGFR and K-RAS gene expression showed significant overexpression, while p53 and JAK-2 gene expression was significantly reduced post-DOX intoxication. Protein expression of both AKT and PI3K significantly increased upon DOX administration. Results showed a remarkable modulation of all disrupted gene and protein expressions upon treatment with TiO2NP-DOX or Lactoferrin-DOX with the superiority of TiO2NP-DOX in modulating these parameters. In conclusion, TiO2NP-DOX could be a promising drug delivery system to improve bioavailability and drug release, as well as reducing DOX's adverse effects particularly on testicular function.
Collapse
Affiliation(s)
- Rehab M. Abdel-Megeed
- Therapeutic Chemistry Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, El Buhouth St., Dokki, Cairo 12622, Egypt
| | - Abdel-Hamid Z. Abdel-Hamid
- Therapeutic Chemistry Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, El Buhouth St., Dokki, Cairo 12622, Egypt
| | - Mai O. Kadry
- Therapeutic Chemistry Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, El Buhouth St., Dokki, Cairo 12622, Egypt
| |
Collapse
|
2
|
Bhardwaj C, Rohilla M, Chopra S, Kaur A, Panigrahi I, Srivastava P. EV-microRNA signatures in pregnant women with idiopathic recurrent pregnancy loss: deciphering microRNAome pathway networks at feto-maternal interface. Front Immunol 2025; 16:1578738. [PMID: 40421018 PMCID: PMC12105548 DOI: 10.3389/fimmu.2025.1578738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 04/21/2025] [Indexed: 05/28/2025] Open
Abstract
Background Despite extensive research in the past decade, the exact pathogenesis of recurrent pregnancy loss (RPL) remains unknown. At the time of pregnancy, human placenta releases microRNAs (miRNAs) enclosed in extracellular vesicles (EVs), which enter into maternal circulation and play an important role at feto-maternal interface to sustain a successful pregnancy. Aberrant expression of these miRNAs often results in adverse pregnancy complications. Therefore, studying the expression of these EV-miRNAs in maternal circulation could provide insights into the pathogenesis of RPL. Methods The present study included idiopathic currently pregnant (<22 weeks of gestation) RPL women (n=10) and gestational-age-matched healthy pregnant women as control (n=5). EVs were isolated from plasma samples and characterized for their morphology and cell-surface marker. Total RNA was isolated and subjected to miRNA sequencing on Illumina NovaSeq 6000 platform. Differentially expressed (DE) miRNAs were identified using DESeq package. Target prediction and pathway analysis were done using TargetScan, miRDB, miRTarBase, and DIANA-miRPath v3.0 online tool. Protein-protein interaction was done using STRING, and hub genes were identified using Cytoscape software. Results miRNA sequencing revealed 66 (44 known and 22 novel) significantly DE miRNAs between RPL and healthy pregnant women. Among these, 37 were downregulated and 29 were upregulated, log2|FC| ≥ 1. Network-based analysis showed highest degree for nine miRNAs (hsa-miR-155-5p, hsa-miR-26a-5p, hsa-miR-204-5p, hsa-miR-140-5p, hsa-miR-139-5p, hsa-let-7e-5p, hsa-miR-149-5p, hsa-miR-374a-5p, and hsa-miR-190a-5p). Gene Ontology (GO) and KEGG pathway analysis of target genes showed significant involvement of Hippo, FoxO, TGF-β, and p53 signaling pathways, which play a crucial role in RPL. Top 10 identified hub genes (NFKB1, IL6, JUN, FOS, CXCL8, PTGS2, TGFB1, MMP9, STAT1, and CD4) were significantly enriched in immunological pathways-Th1/Th2/Th17 differentiation, NF-κB pathway, TNF-α signaling, IL-17 signaling pathway, and vascular endothelial growth factor (VEGF) pathway. Conclusion These results suggest that circulating EV-miRNAs in maternal blood could provide clinical insights into the pathogenesis of RPL and dysregulated immunological and molecular pathways at feto-maternal interface.
Collapse
Affiliation(s)
- Chitra Bhardwaj
- Genetic Metabolic Unit, Department of Pediatrics, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Minakshi Rohilla
- Department of Obstetrics & Gynaecology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Seema Chopra
- Department of Obstetrics & Gynaecology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Anupriya Kaur
- Genetic Metabolic Unit, Department of Pediatrics, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Inusha Panigrahi
- Genetic Metabolic Unit, Department of Pediatrics, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Priyanka Srivastava
- Genetic Metabolic Unit, Department of Pediatrics, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| |
Collapse
|
3
|
Jaber S, Eldawra E, Rakotopare J, Simeonova I, Lejour V, Gabriel M, Cañeque T, Volochtchouk V, Licaj M, Fajac A, Rodriguez R, Morillon A, Bardot B, Toledo F. Oncogenic and teratogenic effects of Trp53Y217C, an inflammation-prone mouse model of the human hotspot mutant TP53Y220C. eLife 2025; 13:RP102434. [PMID: 40223808 PMCID: PMC11996178 DOI: 10.7554/elife.102434] [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] [Indexed: 04/15/2025] Open
Abstract
Missense 'hotspot' mutations localized in six p53 codons account for 20% of TP53 mutations in human cancers. Hotspot p53 mutants have lost the tumor suppressive functions of the wildtype protein, but whether and how they may gain additional functions promoting tumorigenesis remain controversial. Here, we generated Trp53Y217C, a mouse model of the human hotspot mutant TP53Y220C. DNA damage responses were lost in Trp53Y217C/Y217C (Trp53YC/YC) cells, and Trp53YC/YC fibroblasts exhibited increased chromosome instability compared to Trp53-/- cells. Furthermore, Trp53YC/YC male mice died earlier than Trp53-/- males, with more aggressive thymic lymphomas. This correlated with an increased expression of inflammation-related genes in Trp53YC/YC thymic cells compared to Trp53-/- cells. Surprisingly, we recovered only one Trp53YC/YC female for 22 Trp53YC/YC males at weaning, a skewed distribution explained by a high frequency of Trp53YC/YC female embryos with exencephaly and the death of most Trp53YC/YC female neonates. Strikingly, however, when we treated pregnant females with the anti-inflammatory drug supformin (LCC-12), we observed a fivefold increase in the proportion of viable Trp53YC/YC weaned females in their progeny. Together, these data suggest that the p53Y217C mutation not only abrogates wildtype p53 functions but also promotes inflammation, with oncogenic effects in males and teratogenic effects in females.
Collapse
Affiliation(s)
- Sara Jaber
- Genetics of Tumor Suppression, Institut Curie, CNRS UMR3244, Sorbonne University, PSL UniversityParisFrance
| | - Eliana Eldawra
- Genetics of Tumor Suppression, Institut Curie, CNRS UMR3244, Sorbonne University, PSL UniversityParisFrance
| | - Jeanne Rakotopare
- Genetics of Tumor Suppression, Institut Curie, CNRS UMR3244, Sorbonne University, PSL UniversityParisFrance
| | - Iva Simeonova
- Chromatin Dynamics, Institut Curie, CNRS UMR3664, Sorbonne University, PSL UniversityParisFrance
| | - Vincent Lejour
- Genetics of Tumor Suppression, Institut Curie, CNRS UMR3244, Sorbonne University, PSL UniversityParisFrance
| | - Marc Gabriel
- Non Coding RNA, Epigenetic and Genome Fluidity, Institut Curie, CNRS UMR3244, Sorbonne University, PSL UniversityParisFrance
| | - Tatiana Cañeque
- Chemical Biology, Institut Curie, CNRS UMR3666, INSERM U1143, PSL UniversityParisFrance
| | - Vitalina Volochtchouk
- Genetics of Tumor Suppression, Institut Curie, CNRS UMR3244, Sorbonne University, PSL UniversityParisFrance
| | - Monika Licaj
- Genetics of Tumor Suppression, Institut Curie, CNRS UMR3244, Sorbonne University, PSL UniversityParisFrance
| | - Anne Fajac
- Genetics of Tumor Suppression, Institut Curie, CNRS UMR3244, Sorbonne University, PSL UniversityParisFrance
| | - Raphaël Rodriguez
- Chemical Biology, Institut Curie, CNRS UMR3666, INSERM U1143, PSL UniversityParisFrance
| | - Antonin Morillon
- Non Coding RNA, Epigenetic and Genome Fluidity, Institut Curie, CNRS UMR3244, Sorbonne University, PSL UniversityParisFrance
| | - Boris Bardot
- Genetics of Tumor Suppression, Institut Curie, CNRS UMR3244, Sorbonne University, PSL UniversityParisFrance
- Signaling and Neural Crest Development, Institut Curie, CNRS UMR3347, INSERM U1021, Université Paris-Saclay, PSL UniversityOrsayFrance
| | - Franck Toledo
- Genetics of Tumor Suppression, Institut Curie, CNRS UMR3244, Sorbonne University, PSL UniversityParisFrance
- Hematopoietic and Leukemic Development, Centre de Recherche Saint-Antoine, INSERM UMRS938, Sorbonne UniversityParisFrance
| |
Collapse
|
4
|
Cardano M, Buscemi G, Zannini L. Sex Disparities in P53 Regulation and Functions: Novel Insights for Personalized Cancer Therapies. Cells 2025; 14:363. [PMID: 40072091 PMCID: PMC11898824 DOI: 10.3390/cells14050363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 03/15/2025] Open
Abstract
Epidemiological studies have revealed significant sex differences in the incidence of tumors unrelated to reproductive functions, with females demonstrating a lesser risk and a better response to therapy than males. However, the reasons for these disparities are still unknown and cancer therapies are generally sex-unbiased. The tumor-suppressor protein p53 is a transcription factor that can activate the expression of multiple target genes mainly involved in the maintenance of genome stability and tumor prevention. It is encoded by TP53, which is the most-frequently mutated gene in human cancers and therefore constitutes an attractive target for therapy. Recently, evidence of sex differences has emerged in both p53 regulations and functions, possibly providing novel opportunities for personalized cancer medicine. Here, we will review and discuss current knowledge about sexual disparities in p53 pathways, their role in tumorigenesis and cancer progression, and their importance in the therapy choice process, finally highlighting the importance of considering sex contribution in both basic research and clinical practice.
Collapse
|
5
|
Gao L, Jia R. Alternative Splicing: Emerging Roles in Anti-Aging Strategies. Biomolecules 2025; 15:131. [PMID: 39858525 PMCID: PMC11763286 DOI: 10.3390/biom15010131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/21/2024] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
Alternative splicing plays a fundamental role in gene expression and protein complexity. Aberrant splicing impairs cell homeostasis and is closely associated with aging and cellular senescence. Significant changes to alternative splicing, including dysregulated splicing events and the abnormal expression of splicing factors, have been detected during the aging process or in age-related disorders. Here, we highlight the possibility of suppressing aging and cellular senescence by controlling alternative splicing. In this review, we will summarize the latest research progress on alternative splicing in aging and cellular senescence, discuss the roles and regulatory mechanisms of alternative splicing during aging, and then excavate existing and potential approaches to anti-aging by controlling alternative splicing. Novel therapeutic breakthroughs concerning aging and senescence entail a further understanding of regulating alternative splicing mechanically and accurately.
Collapse
Affiliation(s)
| | - Rong Jia
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China;
| |
Collapse
|
6
|
Huang H, Huang C, Li Y, Liang X, Kim N, Xu Y. Methyl Paraben Affects Porcine Oocyte Maturation Through Mitochondrial Dysfunction. Biomolecules 2024; 14:1466. [PMID: 39595642 PMCID: PMC11591637 DOI: 10.3390/biom14111466] [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: 10/10/2024] [Revised: 11/07/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Parabens are widely used in various industries, which are including chemical, pharmaceutical, food, cosmetic, and plastic processing industries. Among these, methyl paraben (MP) serves as an antimicrobial preservative in processed foods, pharmaceuticals, and cosmetics, and it is particularly detected in baby care products. Studies indicate that MP functions as an endocrine-disrupting compound with estrogenic properties, negatively affecting mitochondrial bioenergetics and antioxidant activity in testicular germ cells. However, limited information exists regarding studies on the effects of MP in oocytes. The aim of this study was to investigate the specific mechanism and the toxic effects of MP during oocyte maturation cultured in vitro using a porcine oocyte model. The results indicated that MP (50 μM) inhibited oocyte expansion, significantly reducing the expression of expansion-related genes MAPK1 and ERK1, and decreased the first polar body extrusion significantly as well. ATP levels decreased, reactive oxygen species (ROS) levels remained unchanged, and glutathione (GSH) levels decreased significantly, resulting in an elevated ROS/GSH ratio. The expression of antioxidant genes SOD1 and GPX was significantly decreased. Additionally, a significant decrease in levels of mitochondrial production and biosynthesis protein PGC1α+β, whereas levels of antioxidant-related protein Nrf2 and related gene expression were significantly increased. Autophagy protein LC3B and gene expression significantly decreased, and apoptosis assay indicated a significant increase in levels of caspase3 protein and apoptosis-related genes. These results demonstrated the negative effect of MP on oocyte maturation. In conclusion, our findings indicate that MP disrupts redox balance and induces mitochondrial dysfunction during meiosis in porcine oocytes, resulting in the inhibition of meiotic progression. The present study reveals the mechanism underlying the effects of methyl para-hydroxybenzoate on oocyte maturation.
Collapse
Affiliation(s)
- Huimei Huang
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, South China Institute of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529000, China; (H.H.); (C.H.); (Y.L.)
| | - Chuman Huang
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, South China Institute of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529000, China; (H.H.); (C.H.); (Y.L.)
| | - Yinghua Li
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, South China Institute of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529000, China; (H.H.); (C.H.); (Y.L.)
| | - Xingwei Liang
- College of Animal Science & Technology, Guangxi University, Nanning 530004, China;
| | - Namhyung Kim
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, South China Institute of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529000, China; (H.H.); (C.H.); (Y.L.)
| | - Yongnan Xu
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, South China Institute of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529000, China; (H.H.); (C.H.); (Y.L.)
| |
Collapse
|
7
|
Ming WH, Wen L, Hu WJ, Qiao RF, Zhou Y, Su BW, Bao YN, Gao P, Luan ZL. The crosstalk of Wnt/β-catenin signaling and p53 in acute kidney injury and chronic kidney disease. Kidney Res Clin Pract 2024; 43:724-738. [PMID: 39558651 PMCID: PMC11615452 DOI: 10.23876/j.krcp.23.344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 02/06/2024] [Accepted: 02/22/2024] [Indexed: 11/20/2024] Open
Abstract
Wnt/β-catenin is a signaling pathway associated with embryonic development, organ formation, cancer, and fibrosis. Its activation can repair kidney damage during acute kidney injury (AKI) and accelerate the occurrence of renal fibrosis after chronic kidney disease (CKD). Interestingly, p53 has also been found as a key modulator in AKI and CKD in recent years. Meantime, some studies have found crosstalk between Wnt/β-catenin signaling pathways and p53, but more evidence is required on whether they have synergistic effects in renal disease progression. This article reviews the role and therapeutic targets of Wnt/β-catenin and p53 in AKI and CKD and proposes for the first time that Wnt/β-catenin and p53 have a synergistic effect in the treatment of renal injury.
Collapse
Affiliation(s)
- Wen-Hua Ming
- Department of Allergy, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Lin Wen
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Wen-Juan Hu
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Rong-Fang Qiao
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Yang Zhou
- Beijing Institute of Medical Device Testing, Beijing, China
| | - Bo-Wei Su
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Ya-Nan Bao
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Ping Gao
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Zhi-Lin Luan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| |
Collapse
|
8
|
Ma JW, Xiong ZY, Cai XC, Li X, Ren SY, An SQ, Zhang ZY, Zhang YZ. Ningxin-Tongyu-Zishen formula alleviates the senescence of granulosa cells on D-galactose-induced premature ovarian insufficiency mice. Aging (Albany NY) 2024; 16:4541-4562. [PMID: 38428403 PMCID: PMC10968698 DOI: 10.18632/aging.205607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 01/19/2024] [Indexed: 03/03/2024]
Abstract
Ningxin-Tongyu-Zishen formula (NTZF) is a clinical experience formula for the treatment of premature ovarian insufficiency (POI) in traditional Chinese medicine (TCM), and the potential mechanism is unknown. For in vivo experiments, POI mouse models (C57BL/6 mice), were constructed by subcutaneous injection of D-galactose (D-gal, 200 mg/kg). After treatment of NTZF (10.14, 20.27, 40.54 g/kg;) or estradiol valerate (0.15 mg/kg), ovarian function, oxidative stress (OS) and protein expression of Sirt1/p53 were evaluated. For in vitro experiments, H2O2 (200 μM) was used to treat KGN to construct ovarian granulosa cells (OGCs) cell senescence model. Pretreatment with NTZF (1.06 mg/mL) or p53 inhibitor (Pifithrin-α, 1 μM) was performed before induction of senescence, and further evaluated the cell senescence, OS, mRNA and protein expression of Sirt1/p53. In vivo, NTZF improved ovarian function, alleviated OS and Sirt1/p53 signaling abnormalities in POI mice. In vitro experiments showed that NTZF reduced the level of OS and alleviated the senescence of H2O2-induced KGN. In addition, NTZF activated the protein expression of Sirt1, inhibited the mRNA transcription and protein expression of p53 and p21. Alleviating OGCs senescence and protecting ovarian function through Sirt1/p53 is one of the potential mechanisms of NTZF in the treatment of POI.
Collapse
Affiliation(s)
- Jia-Wen Ma
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zeng-Yan Xiong
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xing-Chu Cai
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiang Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shi-Yan Ren
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuai-Qi An
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zai-Yang Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yi-Zhou Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Famous Chinese Medicine Clinic, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
9
|
Haridevamuthu B, Murugan R, Seenivasan B, Meenatchi R, Pachaiappan R, Almutairi BO, Arokiyaraj S, M K K, Arockiaraj J. Synthetic azo-dye, Tartrazine induces neurodevelopmental toxicity via mitochondria-mediated apoptosis in zebrafish embryos. JOURNAL OF HAZARDOUS MATERIALS 2024; 461:132524. [PMID: 37741213 DOI: 10.1016/j.jhazmat.2023.132524] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/18/2023] [Accepted: 09/09/2023] [Indexed: 09/25/2023]
Abstract
Tartrazine (TZ), or E 102 or C Yellow, is a commonly used azo dye in the food and dyeing industries. Its excessive usage beyond permissible levels threatens human health and the aquatic environment. While previous studies have reported adverse effects such as mutagenicity, carcinogenicity, and reproductive toxicity. Our study aimed to comprehensively evaluate the developmental neurotoxicity of TZ exposure via biochemical and behavioral examinations and explored the underlying mechanism via gene expression analyses. TZ at an environmentally relevant concentration (50 mg/L) significantly induces oxidative stress, altered antioxidant (SOD, CAT and GSH) response, triggered cellular damage (MDA and LDH), and induced neuro-biochemical changes (AChE and NO). Gene expression analyses revealed broad disruptions in genes associated with antioxidant defense (sod1, cat, and gstp1), mitochondrial dysfunction (mfn2, opa1, and fis1),evoked inflammatory response (nfkb, tnfa, and il1b), apoptosis activation (bcl2, bax, and p53), and neural development (bdnf, mbp, and syn2a). Behavioral analysis indicated altered thigmotaxis, touch response, and locomotion depending on the concentration of TZ exposure. Remarkably, the observed effective concentrations were consistent with the permitted levels in food products, highlighting the neurodevelopmental effects of TZ at environmentally relevant concentrations. These findings provide valuable insights into the underlying molecular mechanisms, particularly the role of mitochondria-mediated apoptosis, contributing to TZ-induced neurodevelopmental disorders in vivo.
Collapse
Affiliation(s)
- B Haridevamuthu
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Chengalpattu District, Kattankulathur 603203, Tamil Nadu, India
| | - Raghul Murugan
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Chengalpattu District, Kattankulathur 603203, Tamil Nadu, India
| | - Boopathi Seenivasan
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Chengalpattu District, Kattankulathur 603203, Tamil Nadu, India
| | - Ramu Meenatchi
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Chengalpattu District, Kattankulathur 603203, Tamil Nadu, India
| | - Raman Pachaiappan
- Department of Biotechnology, School of Bioengineering, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Chengalpattu District, Kattankulathur 603203, Tamil Nadu, India
| | - Bader O Almutairi
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Selvaraj Arokiyaraj
- Department of Food Science & Biotechnology, Sejong University, Seoul 05006, South Korea
| | - Kathiravan M K
- Dr APJ Abdul Kalam Research Lab, Department of Pharmaceutical Chemistry, SRM College of Pharmacy, SRM Institute of Science and Technology, Chengalpattu District, Kattankulathur 603203, Tamil Nadu, India
| | - Jesu Arockiaraj
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Chengalpattu District, Kattankulathur 603203, Tamil Nadu, India.
| |
Collapse
|
10
|
Haridevamuthu B, Murugan R, Seenivasan B, Meenatchi R, Pachaiappan R, Almutairi BO, Arokiyaraj S, M. K K, Arockiaraj J. Synthetic azo-dye, Tartrazine induces neurodevelopmental toxicity via mitochondria-mediated apoptosis in zebrafish embryos. JOURNAL OF HAZARDOUS MATERIALS 2024; 461:132524. [DOI: https:/doi.org/10.1016/j.jhazmat.2023.132524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
|
11
|
Kabpha A, Phonsiri K, Pasomboon P, Boonanuntanasarn S. Effects of dietary supplementation of estradiol-17β during fry stage on growth, physiological and immune parameters and gonadal gene expression in adult snakeskin gourami. Animal 2023; 17:100950. [PMID: 37660411 DOI: 10.1016/j.animal.2023.100950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 07/26/2023] [Accepted: 07/30/2023] [Indexed: 09/05/2023] Open
Abstract
In snakeskin gourami (Trichopodus pectoralis), females are generally larger than males, and estradiol-17β (E2)-sex reversal to produce female monosex has gained interest in this species. In this study, we aimed to investigate the effects of E2-induced sex reversal on growth, physiological and immune parameters, and gonadal gene expression in adult snakeskin gourami. Fry (7 days posthatching) were divided into different experimental groups based on the dose of E2: control (no E2 (0 mg kg-1) supplementation), E2-100 (100 mg kg-1), E2-200 (200 mg kg-1), and E2-300 (300 mg kg-1), fed with the E2 doses for 90 d and cultured for 11 months (adult stage). The findings revealed that E2 supplementation produced 88.89-100% of female population. After 11 months of culture, the effects of sexual dimorphism on the growth performance of the E2-100 group were not significant compared to that on the growth performance of the control male and female groups; however, it improved significantly in the E2-200 and E2-300 groups (P < 0.05). E2 elevated the CP and fat contents in body in E2-200 and E2-300 groups (P < 0.05) compared to that in the control group. No sex differences in blood metabolites, haematological values, or immune parameters were identified. Nevertheless, E2-200 and E2-300 groups showed increased blood glucose, triglyceride, haemoglobin, and total immunoglobulin (P < 0.05) compared to control male fish. In addition, all concentrations of E2 increased alternative complement 50 (P < 0.05). Several genes, including bHLH, cyp19a1, daz, deadend, esrb, esrrg, gnrhr, gpa, gsg1l, hsd17β, mospd1, nanos2, p53, piwi2, rerg, rps6ka, tgfb, and vgr, showed differential expression between testis and ovary in control female and E2-treated groups. The expression patterns of the genes were similar in the ovary of the control female and E2-200-treated fish. In conclusion, the findings demonstrate that a feminisation duration of 7-97 days and two doses of E2 at 200 or 300 mg kg-1 successfully produced all-female stocks in snakeskin gourami. Furthermore, the findings showed that E2-treated females were maintained throughout adulthood and exhibited several superior characteristics to male fish. Together with the information generated on differentially expressed sex-related genes, these findings could enable the culturing of faster-growing sex to increase productivity and contribute to the development of intensive snakeskin gourami farming.
Collapse
Affiliation(s)
- A Kabpha
- School of Animal Technology and Innovation, Institute of Agricultural Technology, Suranaree University of Technology, 111 University Avenue, Muang, Nakhon Ratchasima 30000, Thailand
| | - K Phonsiri
- School of Animal Technology and Innovation, Institute of Agricultural Technology, Suranaree University of Technology, 111 University Avenue, Muang, Nakhon Ratchasima 30000, Thailand
| | - P Pasomboon
- School of Animal Technology and Innovation, Institute of Agricultural Technology, Suranaree University of Technology, 111 University Avenue, Muang, Nakhon Ratchasima 30000, Thailand
| | - S Boonanuntanasarn
- School of Animal Technology and Innovation, Institute of Agricultural Technology, Suranaree University of Technology, 111 University Avenue, Muang, Nakhon Ratchasima 30000, Thailand.
| |
Collapse
|
12
|
Basha S, Mukunda DC, Rodrigues J, Gail D'Souza M, Gangadharan G, Pai AR, Mahato KK. A comprehensive review of protein misfolding disorders, underlying mechanism, clinical diagnosis, and therapeutic strategies. Ageing Res Rev 2023; 90:102017. [PMID: 37468112 DOI: 10.1016/j.arr.2023.102017] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 07/14/2023] [Accepted: 07/14/2023] [Indexed: 07/21/2023]
Abstract
INTRODUCTION Proteins are the most common biological macromolecules in living system and are building blocks of life. They are extremely dynamic in structure and functions. Due to several modifications, proteins undergo misfolding, leading to aggregation and thereby developing neurodegenerative and systemic diseases. Understanding the pathology of these diseases and the techniques used to diagnose them is therefore crucial for their effective management . There are several techniques, currently being in use to diagnose them and those will be discussed in this review. AIM/OBJECTIVES Current review aims to discuss an overview of protein aggregation and the underlying mechanisms linked to neurodegeneration and systemic diseases. Also, the review highlights protein misfolding disorders, their clinical diagnosis, and treatment strategies. METHODOLOGY Literature related to neurodegenerative and systemic diseases was explored through PubMed, Google Scholar, Scopus, and Medline databases. The keywords used for literature survey and analysis are protein aggregation, neurodegenerative disorders, Alzheimer's disease, Parkinson's disease, systemic diseases, protein aggregation mechanisms, etc. DISCUSSION /CONCLUSION: This review summarises the pathogenesis of neurodegenerative and systemic disorders caused by protein misfolding and aggregation. The clinical diagnosis and therapeutic strategies adopted for the management of these diseases are also discussed to aid in a better understanding of protein misfolding disorders. Many significant concerns about the role, characteristics, and consequences of protein aggregates in neurodegenerative and systemic diseases are not clearly understood to date. Regardless of technological advancements, there are still great difficulties in the management and cure of these diseases. Therefore, for better understanding, diagnosis, and treatment of neurodegenerative and systemic diseases, more studies to identify novel drugs that may aid in their treatment and management are required.
Collapse
Affiliation(s)
- Shaik Basha
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | | | - Jackson Rodrigues
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Meagan Gail D'Souza
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Gireesh Gangadharan
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Aparna Ramakrishna Pai
- Department of Neurology, Kasturba Medical College - Manipal, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Krishna Kishore Mahato
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| |
Collapse
|
13
|
Chan Y, Xu W, Feng Y, Zhang Y, Li S, Geng Z, Liu Z, Zhao Q, Zhang J, Zhu B. Association of TP53 rs1042522 G > C, MDM2 rs2279744 T > G, and miR-34b/c rs4938723 T > C polymorphisms with aneuploidy pregnancy susceptibility. BMC Pregnancy Childbirth 2023; 23:624. [PMID: 37648962 PMCID: PMC10469955 DOI: 10.1186/s12884-023-05945-3] [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: 12/22/2022] [Accepted: 08/22/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND Aneuploidy pregnancy is a severe major birth defect and causes about 50% spontaneous miscarriages with unknown etiology. To date, only a few epidemiological studies with small sample sizes have investigated the risk factors for aneuploidy pregnancy. TP53, MDM2, and miR-34b/c genes are implicated in tumorigenesis with aneuploidy, yet the function of their polymorphisms in aneuploidy pregnancy susceptibility needs to be clarified. OBJECTIVE To elucidate the association of TP53 rs1042522 G > C, MDM2 rs2279744 309 T > G, and miR-34b/c rs4938723 T > C specific polymorphisms with aneuploidy pregnancy. METHODS In the retrospective case-control study, 330 aneuploidies pregnancy women and 813 normal pregnancy controls were recruited between January 2018 and April 2022 at the First People's Hospital of Yunnan Province, Kunming, China. Three functional polymorphisms, the TP53 rs1042522 G > C (Arg72Pro), MDM2 rs2279744 309 T > G, and miR-34b/c rs4938723 T > C, were genotyped using the snapshot method. RESULTS The frequency distribution of three genotypic variants was not different between case and control pregnant women and was similar to with Hardy-Weinberg Equilibrium (HWE). However, in the younger subgroup (less than 35 years old), a significant difference was detected in allele and recessive model (p = 0.01). In the advanced age subgroup (more than or equal to 35 years old), G of MDM2 rs2279744 T > G revealed a significantly higher frequency in cases than controls (p = 0.045), and miR-34b/c rs4938723 T > C revealed a significant difference under the dominant model (p = 0.03), but no significant differences were observed in other models and in both younger and older subgroup (p > 0.05, respectively). These results suggest that individual polymorphisms were not associated with aneuploidy pregnancy, combined with age, they may serve as a risk factor for aneuploidy pregnancy. CONCLUSION Combination of TP53 rs1042522 G > C, MDM2 rs2279744 T > G, and miR-34b/c rs4938723 T > C polymorphisms with maternal age may be related to aneuploidy pregnancy susceptibility. These findings might elaborate on the genetic etiology of aneuploidy pregnancy.
Collapse
Affiliation(s)
- Ying Chan
- Department of Medical Genetics, NHC Key Laboratory of Periconception Health Birth in Western China, Yunnan Provincial Key Laboratory for Birth Defects and Genetic Diseases, First People's Hospital of Yunnan Province, 157, Jinbi Road, Kunming, 650032, China
- Medical Faculty & Affiliated Hospital, Kunming University of Science and Technology, Kunming, Yunnan Province, 650500, China
| | - Weiming Xu
- Medical Faculty & Affiliated Hospital, Kunming University of Science and Technology, Kunming, Yunnan Province, 650500, China
| | - Yan Feng
- Department of Medical Genetics, NHC Key Laboratory of Periconception Health Birth in Western China, Yunnan Provincial Key Laboratory for Birth Defects and Genetic Diseases, First People's Hospital of Yunnan Province, 157, Jinbi Road, Kunming, 650032, China
| | - Yan Zhang
- Department of Medical Genetics, NHC Key Laboratory of Periconception Health Birth in Western China, Yunnan Provincial Key Laboratory for Birth Defects and Genetic Diseases, First People's Hospital of Yunnan Province, 157, Jinbi Road, Kunming, 650032, China
| | - Suyun Li
- Department of Medical Genetics, NHC Key Laboratory of Periconception Health Birth in Western China, Yunnan Provincial Key Laboratory for Birth Defects and Genetic Diseases, First People's Hospital of Yunnan Province, 157, Jinbi Road, Kunming, 650032, China
| | - Zibiao Geng
- Department of Medical Genetics, NHC Key Laboratory of Periconception Health Birth in Western China, Yunnan Provincial Key Laboratory for Birth Defects and Genetic Diseases, First People's Hospital of Yunnan Province, 157, Jinbi Road, Kunming, 650032, China
| | - Zhijiao Liu
- Department of Medical Genetics, NHC Key Laboratory of Periconception Health Birth in Western China, Yunnan Provincial Key Laboratory for Birth Defects and Genetic Diseases, First People's Hospital of Yunnan Province, 157, Jinbi Road, Kunming, 650032, China
| | - Qingfen Zhao
- Department of Medical Genetics, NHC Key Laboratory of Periconception Health Birth in Western China, Yunnan Provincial Key Laboratory for Birth Defects and Genetic Diseases, First People's Hospital of Yunnan Province, 157, Jinbi Road, Kunming, 650032, China
| | - Jinman Zhang
- Department of Medical Genetics, NHC Key Laboratory of Periconception Health Birth in Western China, Yunnan Provincial Key Laboratory for Birth Defects and Genetic Diseases, First People's Hospital of Yunnan Province, 157, Jinbi Road, Kunming, 650032, China
- Medical Faculty & Affiliated Hospital, Kunming University of Science and Technology, Kunming, Yunnan Province, 650500, China
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan Province, 650500, China
| | - Baosheng Zhu
- Department of Medical Genetics, NHC Key Laboratory of Periconception Health Birth in Western China, Yunnan Provincial Key Laboratory for Birth Defects and Genetic Diseases, First People's Hospital of Yunnan Province, 157, Jinbi Road, Kunming, 650032, China.
- Medical Faculty & Affiliated Hospital, Kunming University of Science and Technology, Kunming, Yunnan Province, 650500, China.
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan Province, 650500, China.
| |
Collapse
|
14
|
Lopez J, Hohensee G, Liang J, Sela M, Johnson J, Kallen AN. The Aging Ovary and the Tales Learned Since Fetal Development. Sex Dev 2023; 17:156-168. [PMID: 37598664 PMCID: PMC10841896 DOI: 10.1159/000532072] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 07/13/2023] [Indexed: 08/22/2023] Open
Abstract
BACKGROUND While the term "aging" implies a process typically associated with later life, the consequences of ovarian aging are evident by the time a woman reaches her forties, and sometimes earlier. This is due to a gradual decline in the quantity and quality of oocytes which occurs over a woman's reproductive lifespan. Indeed, the reproductive potential of the ovary is established even before birth, as the proper formation and assembly of the ovarian germ cell population during fetal life determines the lifetime endowment of oocytes and follicles. In the ovary, sophisticated molecular processes have been identified that regulate the timing of ovarian aging and these are critical to ensuring follicular maintenance. SUMMARY The mechanisms thought to contribute to overall aging have been summarized under the term the "hallmarks of aging" and include such processes as DNA damage, mitochondrial dysfunction, telomere attrition, genomic instability, and stem cell exhaustion, among others. Similarly, in the ovary, molecular processes have been identified that regulate the timing of ovarian aging and these are critical to ensuring follicular maintenance. In this review, we outline critical processes involved in ovarian aging, highlight major achievements for treatment of ovarian aging, and discuss ongoing questions and areas of debate. KEY MESSAGES Ovarian aging is recognized as what may be a complex process in which age, genetics, environment, and many other factors contribute to the size and depletion of the follicle pool. The putative hallmarks of reproductive aging outlined herein include a diversity of plausible processes contributing to the depletion of the ovarian reserve. More research is needed to clarify if and to what extent these putative regulators do in fact govern follicle and oocyte behavior, and how these signals might be integrated in order to control the overall pattern of ovarian aging.
Collapse
Affiliation(s)
- Jesus Lopez
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Gabe Hohensee
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Jing Liang
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Meirav Sela
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Joshua Johnson
- Department of Obstetrics and Gynecology, University of Colorado Denver, Aurora, CO, USA
| | - Amanda N. Kallen
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
15
|
Angwa LM, Nyadanu SD, Kanyugo AM, Adampah T, Pereira G. Fluoride-induced apoptosis in non-skeletal tissues of experimental animals: A systematic review and meta-analysis. Heliyon 2023; 9:e18646. [PMID: 37560699 PMCID: PMC10407679 DOI: 10.1016/j.heliyon.2023.e18646] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 07/15/2023] [Accepted: 07/24/2023] [Indexed: 08/11/2023] Open
Abstract
Different studies have suggested that fluoride can induce apoptosis in non-skeletal tissues, however, evidence from these experimental studies is still controversial. This meta-analysis aims to clarify the mechanism of fluoride-induced apoptosis in non-skeletal tissues of experimental animals. Primary studies which measured apoptosis were identified through exhaustive database searching in PubMed, Embase, Web of Science Core Collection, Scopus, and references of included studies. A random effects model with standardized mean difference (SMD) was used for meta-analyses. The heterogeneity of the studies was evaluated using Higgin's I2 statistics. The risk of bias and publication bias were assessed using the SYRCLE's risk of bias tool and Egger's test, respectively. There was an increase in total apoptotic cells, and the expression of Bax, Bax/Bcl-2 ratio, caspase-3, caspase-8, caspase-9, Cyt c, and p53, and a decrease in the expression of Bcl-2 in the fluoride-treated groups as compared to the control groups. However, there was no evidence of a difference in the expression of APAF-1 in the two groups. The subgroup analysis highlighted the role of the intervention period in modification of the apoptotic effect of fluoride and that the susceptibility and tolerance of different animal species and tissues vary. Meta-regression analysis indicated that the studies' effect size for total apoptotic cells was influenced by animal species and that of Bax by the sample source. The results of this meta-analysis revealed that fluoride causes apoptosis by up-regulating caspase-3, -8, and -9, Cyt c, p53, Bax, and down-regulating Bcl-2 with a concomitant up-regulation of the Bax/Bcl-2 ratio.
Collapse
Affiliation(s)
- Linet Musungu Angwa
- Department of Clinical Medicine, Kabarak University, Private Bag, 20157, Kabarak, Kenya
| | - Sylvester Dodzi Nyadanu
- Curtin School of Population Health, Curtin University, Perth, Kent Street, Bentley, Western Australia, 6102, Australia
- Education, Culture, and Health Opportunities (ECHO) Research Group International, Aflao, Ghana
| | - Anne Murugi Kanyugo
- Department of Clinical Medicine, Kabarak University, Private Bag, 20157, Kabarak, Kenya
| | - Timothy Adampah
- Education, Culture, and Health Opportunities (ECHO) Research Group International, Aflao, Ghana
| | - Gavin Pereira
- Curtin School of Population Health, Curtin University, Perth, Kent Street, Bentley, Western Australia, 6102, Australia
- Centre for Fertility and Health (CeFH), Norwegian Institute of Public Health, 0473, Oslo, Norway
- enAble Institute, Curtin University, Perth, Kent Street, Bentley, Western Australia, 6102, Australia
| |
Collapse
|
16
|
Amendolare A, Marzano F, Petruzzella V, Vacca RA, Guerrini L, Pesole G, Sbisà E, Tullo A. The Underestimated Role of the p53 Pathway in Renal Cancer. Cancers (Basel) 2022; 14:cancers14235733. [PMID: 36497215 PMCID: PMC9736171 DOI: 10.3390/cancers14235733] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/10/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022] Open
Abstract
The TP53 tumor suppressor gene is known as the guardian of the genome, playing a pivotal role in controlling genome integrity, and its functions are lost in more than 50% of human tumors due to somatic mutations. This percentage rises to 90% if mutations and alterations in the genes that code for regulators of p53 stability and activity are taken into account. Renal cell carcinoma (RCC) is a clear example of cancer that despite having a wild-type p53 shows poor prognosis because of the high rate of resistance to radiotherapy or chemotherapy, which leads to recurrence, metastasis and death. Remarkably, the fact that p53 is poorly mutated does not mean that it is functionally active, and increasing experimental evidences have demonstrated this. Therefore, RCC represents an extraordinary example of the importance of p53 pathway alterations in therapy resistance. The search for novel molecular biomarkers involved in the pathways that regulate altered p53 in RCC is mandatory for improving early diagnosis, evaluating the prognosis and developing novel potential therapeutic targets for better RCC treatment.
Collapse
Affiliation(s)
- Alessandra Amendolare
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70121 Bari, Italy
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council—CNR, 70126 Bari, Italy
| | - Flaviana Marzano
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council—CNR, 70126 Bari, Italy
| | - Vittoria Petruzzella
- Department of Translational Biomedicine and Neuroscience (DiBraiN), University of Bari Aldo Moro, 70121 Bari, Italy
| | - Rosa Anna Vacca
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council—CNR, 70126 Bari, Italy
| | - Luisa Guerrini
- Department of Biosciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Graziano Pesole
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70121 Bari, Italy
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council—CNR, 70126 Bari, Italy
| | - Elisabetta Sbisà
- Institute of Biomedical Technologies, National Research Council—CNR, 70126 Bari, Italy
| | - Apollonia Tullo
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council—CNR, 70126 Bari, Italy
- Correspondence: ; Tel.: +39-0805929672
| |
Collapse
|
17
|
p53 Controls Meiotic Prophase Progression and Crossover Formation. Int J Mol Sci 2022; 23:ijms23179818. [PMID: 36077210 PMCID: PMC9456223 DOI: 10.3390/ijms23179818] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/12/2022] [Accepted: 08/17/2022] [Indexed: 11/17/2022] Open
Abstract
Meiosis initiates with the formation of double strand breaks (DSBs) throughout the genome. To avoid genomic instability, these DSBs need to be correctly repaired by homologous recombination. Surveillance mechanisms involving the DNA damage response (DDR) pathway ATM-CHK2-p53 can detect the persistence of unrepaired DBSs and activate the recombination-dependent arrest at the pachytene stage. However, a complete understanding of p53 functions under normal physiological conditions remains lacking. Here, we report a detailed analysis of the p53 role during meiotic prophase in mice spermatocytes. We show that the absence of p53 regulates prophase progression by slowing down the pachytene stage when the recombination-dependent arrest occurs. Furthermore, our results show that p53 is necessary for proper crossover (CO) formation and localization. Our study contributes to a deeper understanding of p53 roles during the meiotic prophase.
Collapse
|
18
|
Liu C, Moten A, Ma Z, Lin HK. The foundational framework of tumors: Gametogenesis, p53, and cancer. Semin Cancer Biol 2022; 81:193-205. [PMID: 33940178 PMCID: PMC9382687 DOI: 10.1016/j.semcancer.2021.04.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 04/20/2021] [Accepted: 04/26/2021] [Indexed: 12/20/2022]
Abstract
The completion-of-tumor hypothesis involved in the dynamic interplay between the initiating oncogenic event and progression is essential to better recognize the foundational framework of tumors. Here we review and extend the gametogenesis-related hypothesis of tumors, because high embryonic/germ cell traits are common in tumors. The century-old gametogenesis-related hypothesis of tumors postulated that tumors arise from displaced/activated trophoblasts, displaced (lost) germ cells, and the reprogramming/reactivation of gametogenic program in somatic cells. Early primordial germ cells (PGCs), embryonic stem (ES) cells, embryonic germ cells (EGCs), and pre-implantation embryos at the stage from two-cell stage to blastocysts originating from fertilization or parthenogenesis have the potential to develop teratomas/teratocarcinomas. In addition, the teratomas/teratocarcinomas/germ cells occur in gonads and extra-gonads. Undoubtedly, the findings provide strong support for the hypothesis. However, it was thought that these tumor types were an exception rather than verification. In fact, there are extensive similarities between somatic tumor types and embryonic/germ cell development, such as antigens, migration, invasion, and immune escape. It was documented that embryonic/germ cell genes play crucial roles in tumor behaviors, e.g. tumor initiation and metastasis. Of note, embryonic/germ cell-like tumor cells at different developmental stages including PGC and oocyte to the early embryo-like stage were identified in diverse tumor types by our group. These embryonic/germ cell-like cancer cells resemble the natural embryonic/germ cells in morphology, gene expression, the capability of teratoma formation, and the ability to undergo the process of oocyte maturation and parthenogenesis. These embryonic/germ cell-like cancer cells are derived from somatic cells and contribute to tumor formation, metastasis, and drug resistance, establishing asexual meiotic embryonic life cycle. p53 inhibits the reactivation of embryonic/germ cell state in somatic cells and oocyte-like cell maturation. Based on earlier and our recent studies, we propose a novel model to complete the gametogenesis-related hypothesis of tumors, which can be applied to certain somatic tumors. That is, tumors tend to establish a somatic asexual meiotic embryonic cycle through the activation of somatic female gametogenesis and parthenogenesis in somatic tumor cells during the tumor progression, thus passing on corresponding embryonic/germ cell traits leading to the malignant behaviors and enhancing the cells' independence. This concept may be instrumental to better understand the nature and evolution of tumors. We rationalize that targeting the key events of somatic pregnancy is likely a better therapeutic strategy for cancer treatment than directly targeting cell mitotic proliferation, especially for those tumors with p53 inactivation.
Collapse
Affiliation(s)
- Chunfang Liu
- Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China.
| | - Asad Moten
- Medical Sciences Division, University of Oxford, Oxford OX3 9DU, UK
| | - Zhan Ma
- Department of Laboratory Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai 200040, China
| | - Hui-Kuan Lin
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
19
|
Activation of Esterase D by FPD5 Inhibits Growth of A549 Lung Cancer Cells via JAB1/p53 Pathway. Genes (Basel) 2022; 13:genes13050786. [PMID: 35627173 PMCID: PMC9141839 DOI: 10.3390/genes13050786] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 04/18/2022] [Accepted: 04/21/2022] [Indexed: 12/04/2022] Open
Abstract
Esterase D (ESD) is widely distributed in mammals, and it plays an important role in drug metabolism, detoxification, and biomarkers and is closely related to the development of tumors. In our previous work, we found that a chemical small-molecule fluorescent pyrazoline derivative, FPD5, an ESD activator, could inhibit tumor growth by activating ESD, but its molecular mechanism is still unclear. Here, by using RNA interference (RNAi), andco-immunoprecipitation techniques, we found that ESD suppressed the nucleus exportation of p53 through reducing the interaction between p53 and JAB1. The protein level of p53 in the nucleus was upregulated and the downstream targets of p53 were found by Human Gene Expression Array. p53 inhibited the expression of CDCA8 and CDC20. Lastly, the cell cycle of A549 cells was arrested at the G0/G1 phase. Together, our data suggest that ESD inhibited the cancer cell growth by arresting the cell cycle of A549 cells via the JAB1/p53 signaling pathway. Our findings provide a new insight into how to inhibit the growth of lung cancer with the activation of ESD by FPD5.
Collapse
|
20
|
Gan J, Gu T, Hong L, Cai G. Ferroptosis-related genes involved in animal reproduction: An Overview. Theriogenology 2022; 184:92-99. [DOI: 10.1016/j.theriogenology.2022.02.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/20/2022] [Accepted: 02/25/2022] [Indexed: 11/30/2022]
|
21
|
Palomares AR, Castillo-Domínguez AA, Ruiz-Galdón M, Rodriguez-Wallberg KA, Reyes-Engel A. Genetic variants in the p53 pathway influence implantation and pregnancy maintenance in IVF treatments using donor oocytes. J Assist Reprod Genet 2021; 38:3267-3275. [PMID: 34618298 PMCID: PMC8666387 DOI: 10.1007/s10815-021-02324-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 09/17/2021] [Indexed: 11/12/2022] Open
Abstract
PURPOSE Single-nucleotide polymorphisms (SNPs) in the p53 pathways have shown to play a role in endometrial receptivity and implantation in infertile women undergoing in vitro fertilization (IVF). The present study aimed to assess the influence of these gene variants over pregnancy success through a receptivity model in recipients of egg donation treatments, when factors such as age and quality of the oocytes are standardized. METHODS A nested case-control study was performed on 234 female patients undergoing their first fresh IVF treatment as recipients of donor oocytes. Genotyping of TP53 Arg72Pro (rs1042522), LIF (rs929271), MDM4 (rs1563828), and USP7 (rs1529916) SNPs in the recipients allowed comparison of allele and genotype frequencies and their association with the IVF treatment outcome. RESULTS Grouped by genotypes, patients showed differences in IVF outcomes after the embryo transfer. Arg72Pro (rs1042522) gene variant was associated to changes in implantation and clinical pregnancy rates. The polymorphisms USP7 (rs1529916) and MDM4 (rs1563828) were associated to differential ongoing pregnancy rates and variable miscarriage events, respectively. CONCLUSIONS This study highlights the association between gene polymorphisms related to P53 function and their influence over IVF reproductive outcomes. Arg72Pro variant may influence early events, as lower implantation rates were found in homozygous for Pro72 allele. By contrast, MDM4 (rs1563828) and USP7 (rs1529916) gene variants were associated with the later maintenance of pregnancy.
Collapse
Affiliation(s)
- Arturo R Palomares
- IVF Unit, Instituto de Fertilidad Clínica Rincón, 29730, Malaga, Spain.
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Malaga, 29071, Malaga, Spain.
- Laboratory of Translational Fertility Preservation, Department of Oncology and Pathology, BioClinicum J 5:30, New Karolinska Hospital, Karolinska Institutet, Visionsgatan 4, 17164, Solna, Stockholm, Sweden.
| | | | - Maximiliano Ruiz-Galdón
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Malaga, 29071, Malaga, Spain
- Clinical Analysis Service, Virgen de La Victoria University Hospital, 29010, Malaga, Spain
| | - Kenny A Rodriguez-Wallberg
- Laboratory of Translational Fertility Preservation, Department of Oncology and Pathology, BioClinicum J 5:30, New Karolinska Hospital, Karolinska Institutet, Visionsgatan 4, 17164, Solna, Stockholm, Sweden
- Karolinska University Hospital, Section of Reproductive Medicine, Novumhuset Plan 4, 14186, Stockholm, Sweden
| | - Armando Reyes-Engel
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Malaga, 29071, Malaga, Spain
| |
Collapse
|
22
|
p53: A Double-Edged Sword in Tumor Ferroptosis. Pharmacol Res 2021; 177:106013. [PMID: 34856333 DOI: 10.1016/j.phrs.2021.106013] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 11/11/2021] [Accepted: 11/26/2021] [Indexed: 12/12/2022]
Abstract
Ferroptosis is a type of lipid peroxidation-induced cell death that can be regulated in various ways, from changing the activity of antioxidant enzymes to the levels of transcription factors. The p53 tumor suppressor gene is the "guardian of the genome" and is involved in controlling cell survival and division under various pressures. In addition to its effects on apoptosis, autophagy, and cell cycle, p53, through the way of transcription dependent or independent two-way, also regulates the biological processes of tumor cell sensitivity to ferroptosis, including the metabolism of amino acids, nicotinamide adenine dinucleotide phosphate, and lipid peroxidation, as well as the biosynthesis of glutathione, phospholipids, NADPH and coenzyme Q10.As reviewed here, we summarized the metabolic network of p53 and its signaling pathway in regulating ferroptosis and elucidated possible factors and potential clinical application of p53 regulating ferroptosis. This review will provide a basis for further understanding the role of p53 in tumor ferroptosis and new strategies for cancer therapeutic avenues.
Collapse
|
23
|
Sharma AK, Sah S, Singla SK, Chauhan MS, Manik RS, Palta P. Exposure to Pulsed Electromagnetic Fields Improves the Developmental Competence and Quality of Somatic Cell Nuclear Transfer Buffalo ( Bubalus bubalis) Embryos Produced Using Fibroblast Cells and Alters Their Epigenetic Status and Gene Expression. Cell Reprogram 2021; 23:304-315. [PMID: 34597162 DOI: 10.1089/cell.2021.0028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
We examined the effects of treatment with pulsed electromagnetic fields (PEMFs) on cumulus cells and buffalo somatic cell nuclear transfer (SCNT) embryos. PEMF treatment (30 μT for 3 hours) of cumulus cells increased (p < 0.05) the relative cell viability and cell proliferation and the expression level of OCT4, NANOG, SOX2, P53, CCNB1, and GPX, but decreased (p < 0.05) that of DNMT1, DNMT3a, GSK3b, and BAX, whereas the expression level of DNMT3b, GLUT1, BCL2, CASPASE3, SOD1, and CATALASE was not affected. PEMF treatment of SCNT embryos at the beginning of in vitro culture increased (p < 0.05) the blastocyst rate (51.4% ± 1.36% vs. 42.8% ± 1.29%) and decreased (p < 0.01) the apoptotic index to the level in in vitro fertilization blastocysts, but did not significantly alter the total cell number and the inner cell mass:trophectoderm cell number ratio of blastocysts compared to the controls. PEMF treatment increased the expression level of NANOG, SOX2, CDX2, GLUT1, P53, and BCL2 and decreased that of BAX, CASPASE3, GSK3b, and HSP70, but not OCT4, DNMT1, DNMT3a, DNMT3b, HDAC1, and CCNB1 in blastocysts. It increased (p < 0.001) the global level of H3K27me3 but not H3K18ac. These results suggest that PEMF treatment of SCNT embryos improves their developmental competence, reduces the level of apoptosis, and alters the expression level of several important genes related to pluripotency, apoptosis, metabolism, and stress.
Collapse
Affiliation(s)
- Aditya Kumar Sharma
- Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, India
| | - Shrutika Sah
- Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, India
| | - Suresh Kumar Singla
- Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, India
| | | | - Radhey Shyam Manik
- Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, India
| | - Prabhat Palta
- Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, India.,Embryo Biotechnology Laboratory, Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, India
| |
Collapse
|
24
|
Vagnini LD, Renzi A, Petersen CG, Dieamant F, Oliveira JBA, Canas MDCT, Franco Junior JG, Oliani AH. Correlation of TP53 (rs1625895), TP73 (rs3765730), MMP9 (rs17576), and MTHFR (rs868014) polymorphisms with low ovarian reserve. Eur J Obstet Gynecol Reprod Biol 2021; 269:132-137. [PMID: 34953598 DOI: 10.1016/j.ejogrb.2021.08.034] [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: 09/22/2020] [Revised: 06/18/2021] [Accepted: 08/26/2021] [Indexed: 01/09/2023]
Abstract
OBJECTIVE To investigate the influence of the Single Nucleotide Polymorphisms (SNPs) TP53 rs1625895, TP73 rs3765730, MMP9 rs17576, and MTHFR rs868014 on ovarian reserve (OR) in infertile patients. STUDY DESIGN A prospective cross-sectional study was carried out in 145 infertile women. The patients were divided into two groups according to ovarian reserve, characterized by association between AMH levels and AFC:After patient distribution, both groups were compared (LOR X NOR) regarding the genotypes of the SNPs TP53 T/C rs1625895, TP73 G/A rs3765730, MMP9 Gln/Arg rs17576, and MTHFR A/G rs868014. RESULT(S) The frequency of the TP53-T/T genotype was greater in the LOR and the TP53-C/C genotype was more frequent in patients with NOR. This association was confirmed by the frequency of alleles, where the presence of the T allele was significantly higher in patients who exhibited LOR (P = 0.0003). The frequency of the TP73-G/G genotype and of the G allele was higher in the LOR group (P = 0.01). Considering the MMP9 gene, the frequency of the Gln/Gln genotype was higher in the LOR group. However, the Gln/Arg genotype and the Arg allele prevailed in the NOR group (P = 0.006). The frequency of the MTHFR-A/A genotype was higher in the LOR group, whereas that of the MTHFR-GG genotype was higher in the NOR group. The presence of allele A was significantly higher in the LOR group (P = 0.002). The regression analysis shows that patients who present the TP53-T/T, TP73-G/G, MMP9-Gln/Gln, and MTHFR-A/A genotypes are 3.6X, 3.1X, 3.2X, and 3.7X more likely of having LOR, respectively. In addition, the association of the TP53/TT + TP73/GG genotypes increased the chance of women being included in the LOR group in 5.7-fold. CONCLUSION(S) The genotypes TP53-T/T, TP73-G/G, MMP9-Gln/Gln, and MTHFR-A/A increase the chance of women to exhibit LOR. These polymorphisms could be useful as genetic markers of low ovarian reserve in infertile patients.
Collapse
Affiliation(s)
- Laura D Vagnini
- Paulista Centre for Diagnosis Research and Training, CPDP, Ribeirao Preto, Brazil.
| | - Adriana Renzi
- Paulista Centre for Diagnosis Research and Training, CPDP, Ribeirao Preto, Brazil
| | - Cláudia G Petersen
- Paulista Centre for Diagnosis Research and Training, CPDP, Ribeirao Preto, Brazil; Centre for Human Reproduction Prof Franco Jr, Ribeirao Preto, Brazil
| | - Felipe Dieamant
- Centre for Human Reproduction Prof Franco Jr, Ribeirao Preto, Brazil
| | - João Batista A Oliveira
- Paulista Centre for Diagnosis Research and Training, CPDP, Ribeirao Preto, Brazil; Centre for Human Reproduction Prof Franco Jr, Ribeirao Preto, Brazil
| | | | - José G Franco Junior
- Paulista Centre for Diagnosis Research and Training, CPDP, Ribeirao Preto, Brazil; Centre for Human Reproduction Prof Franco Jr, Ribeirao Preto, Brazil
| | - Antônio H Oliani
- São José do Rio Preto School of Medicine FAMERP, Sao Jose do Rio Preto, Brazil
| |
Collapse
|
25
|
Guo SH, Liu YM, Wang ZY, Wang FF, Mao YK, Hu YW, Han P, Cuthbertson AGS, Qiu BL, Sang W. Transcriptome analysis reveals TOR signalling-mediated plant flush shoots governing Diaphorina citri Kuwayama oviposition. INSECT MOLECULAR BIOLOGY 2021; 30:264-276. [PMID: 33410566 DOI: 10.1111/imb.12693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 12/31/2020] [Accepted: 01/04/2021] [Indexed: 06/12/2023]
Abstract
Asian Citrus Psyllid (ACP), Diaphorina citri, is a key vector transmitting the causative agent of Huanglongbing (HLB) disease. Population growth of ACP is evident after feeding on plant flush shoots, as they only oviposit here. However, the underlying mechanism as to why flush shoots govern oviposition is unclear. This study compares the fecundity and ovarian morphology of ACP between young flush and mature leaves. Furthermore, the transcriptome of mated females infesting Murraya paniculata was analysed. Finally, the gene of the key Target of Rapamycin (TOR) signalling pathway was silenced by RNAi. Results indicated that flush shoot feeding activated the development of the psyllids ovary and therefore induced oviposition. A total of 126 and 2794 differentially expressed genes were detected at 1 and 5 days, respectively, after pest infestation of flush shoots compared to mature leaves. Many genes are involved in protein metabolism, Mitogen-Activated Protein Kinase (MAPK) signalling pathway, hormone synthesis, and TOR signalling pathway: all thought to activate reproduction. Silencing of the positive regulator gene DcRheb in the TOR pathway resulted in lower levels of ecdysone and juvenile hormone and decreased vitellogenin synthesis, further disrupting reproductive ability. This study enhances understanding of the molecular mechanism underlying ACP's reproductive strategy.
Collapse
Affiliation(s)
- S-H Guo
- Key Laboratory of Bio-Pesticide Creation and Application, South China Agricultural University, Guangzhou, China
| | - Y-M Liu
- Key Laboratory of Bio-Pesticide Creation and Application, South China Agricultural University, Guangzhou, China
| | - Z-Y Wang
- Key Laboratory of Bio-Pesticide Creation and Application, South China Agricultural University, Guangzhou, China
| | - F-F Wang
- Key Laboratory of Bio-Pesticide Creation and Application, South China Agricultural University, Guangzhou, China
| | - Y-K Mao
- Guangdong Provincial Bioengineering Institute (Guangzhou Sugarcane Industry Research Institute), Guangzhou, China
| | - Y-W Hu
- Guangdong Provincial Bioengineering Institute (Guangzhou Sugarcane Industry Research Institute), Guangzhou, China
| | - P Han
- CAS Key Laboratory of Biogeography and Bioresource in Arid Land, Chinese Academy of Sciences, Ürümqi, China
| | | | - B-L Qiu
- Key Laboratory of Bio-Pesticide Creation and Application, South China Agricultural University, Guangzhou, China
| | - W Sang
- Key Laboratory of Bio-Pesticide Creation and Application, South China Agricultural University, Guangzhou, China
| |
Collapse
|
26
|
Zhang C, Liu J, Wang J, Hu W, Feng Z. The emerging role of leukemia inhibitory factor in cancer and therapy. Pharmacol Ther 2021; 221:107754. [PMID: 33259884 PMCID: PMC8084904 DOI: 10.1016/j.pharmthera.2020.107754] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 11/17/2020] [Indexed: 12/11/2022]
Abstract
Leukemia inhibitory factor (LIF) is a multi-functional cytokine of the interleukin-6 (IL-6) superfamily. Initially identified as a factor that inhibits the proliferation of murine myeloid leukemia cells, LIF displays a wide variety of important functions in a cell-, tissue- and context-dependent manner in many physiological and pathological processes, including regulating cell proliferation, pluripotent stem cell self-renewal, tissue/organ development and regeneration, neurogenesis and neural regeneration, maternal reproduction, inflammation, infection, immune response, and metabolism. Emerging evidence has shown that LIF plays an important but complex role in human cancers; while LIF displays a tumor suppressive function in some types of cancers, including leukemia, LIF is overexpressed and exerts an oncogenic function in many more types of cancers. Further, targeting LIF has been actively investigated as a novel strategy for cancer therapy. This review summarizes the recent advances in the studies on LIF in human cancers and its potential application in cancer therapy. A better understanding of the role of LIF in different types of cancers and its underlying mechanisms will help to develop more effective strategies for cancer therapy.
Collapse
Affiliation(s)
- Cen Zhang
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers-State University of New Jersey, New Brunswick, NJ 08903, USA
| | - Juan Liu
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers-State University of New Jersey, New Brunswick, NJ 08903, USA
| | - Jianming Wang
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers-State University of New Jersey, New Brunswick, NJ 08903, USA
| | - Wenwei Hu
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers-State University of New Jersey, New Brunswick, NJ 08903, USA.
| | - Zhaohui Feng
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers-State University of New Jersey, New Brunswick, NJ 08903, USA.
| |
Collapse
|
27
|
Zhang C, Liu J, Wang J, Zhang T, Xu D, Hu W, Feng Z. The Interplay Between Tumor Suppressor p53 and Hypoxia Signaling Pathways in Cancer. Front Cell Dev Biol 2021; 9:648808. [PMID: 33681231 PMCID: PMC7930565 DOI: 10.3389/fcell.2021.648808] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 01/29/2021] [Indexed: 12/13/2022] Open
Abstract
Hypoxia is a hallmark of solid tumors and plays a critical role in different steps of tumor progression, including proliferation, survival, angiogenesis, metastasis, metabolic reprogramming, and stemness of cancer cells. Activation of the hypoxia-inducible factor (HIF) signaling plays a critical role in regulating hypoxic responses in tumors. As a key tumor suppressor and transcription factor, p53 responds to a wide variety of stress signals, including hypoxia, and selectively transcribes its target genes to regulate various cellular responses to exert its function in tumor suppression. Studies have demonstrated a close but complex interplay between hypoxia and p53 signaling pathways. The p53 levels and activities can be regulated by the hypoxia and HIF signaling differently depending on the cell/tissue type and the severity and duration of hypoxia. On the other hand, p53 regulates the hypoxia and HIF signaling at multiple levels. Many tumor-associated mutant p53 proteins display gain-of-function (GOF) oncogenic activities to promote cancer progression. Emerging evidence has also shown that GOF mutant p53 can promote cancer progression through its interplay with the hypoxia and HIF signaling pathway. In this review, we summarize our current understanding of the interplay between the hypoxia and p53 signaling pathways, its impact upon cancer progression, and its potential application in cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Wenwei Hu
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers-State University of New Jersey, New Brunswick, NJ, United States
| | - Zhaohui Feng
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers-State University of New Jersey, New Brunswick, NJ, United States
| |
Collapse
|
28
|
Terawaki K, Saegusa Y, Sekiguchi K, Shimizu T, Takiyama M, Matsumoto T, Iizuka S, Matsumoto C, Motoyama F. The ameliorating effects of tokishakuyakusan in a rat model of implantation failure involves endometrial gland leukemia inhibitory factor and decidualization. JOURNAL OF ETHNOPHARMACOLOGY 2021; 265:113288. [PMID: 32841695 DOI: 10.1016/j.jep.2020.113288] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 08/07/2020] [Accepted: 08/16/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tokishakuyakusan (TSS) is a Kampo medicine that is prescribed for the treatment of infertility in Japan. However, its precise mechanism of action remains unclear. AIM OF THE STUDY Leukemia inhibitory factor (LIF) in the endometrium plays an indispensable role in embryo implantation and is linked to infertility or implantation failure. Previously, we demonstrated that TSS ameliorated implantation failure induced by mifepristone (RU-486), an antagonist of progesterone, in rats. Herein, we aimed to clarify whether the ameliorating effect of TSS on implantation failure in the rat model involves endometrial LIF. Additionally, we determined whether decidualization, the dysfunction of which is linked to infertility or implantation failure similar to LIF, progesterone, and other implantation-related factors, are involved in the effect of TSS. MATERIALS AND METHODS The implantation failure rat model was developed via the subcutaneous administration of RU-486 (7 mg/kg) on day 3 post-coitus. Sesame oil was administered as the vehicle control. Rats were fed a diet containing 1% or 3% TSS or a control diet from day 13 pre-coitus. Subsequently, the implantation sites were assessed, and plasma progesterone levels were analyzed by liquid chromatography-tandem mass spectrometry (LC-MS/MS) on day 8 post-coitus. The LIF mRNA of the endometrial gland, which was segmented via laser-microdissection from the endometrial tissue, was measured, and endometrial LIF immunostaining was carried out on day 5. The gene expression of different factors related to implantation, including decidualization and progesterone-responsiveness on days 5 and 6, were measured. The human endometrial Ishikawa cell line derived from human adenocarcinoma was treated with TSS (30-300 μg/mL) for 24 h, and the LIF concentrations in the cell culture supernatants were measured. RESULTS RU-486 decreased the number of implantation sites in the uterus of rats; however, the decrease was significantly alleviated by TSS (3%-diet), which tended to increase plasma progesterone. In rats with RU-486-induced implantation failure, endometrial gland LIF mRNA and endometrial LIF protein were markedly decreased while the gene expression of both decidualization-related factors such as interleukin-11, insulin-like growth factor binding protein-1, and cyclooxygenase-2, and progesterone responsive-related factors such as FK506 binding protein 5, were significantly decreased. These changes in the uterus of rats with implantation failure were significantly alleviated by TSS (3%-diet). Additionally, TSS significantly enhanced LIF protein production and LIF mRNA in Ishikawa cells. CONCLUSIONS The mechanism whereby TSS ameliorates RU-486-induced implantation failure in rats may involve the alleviation of decreased LIF production derived from the endometrial gland, and a dysfunction of decidualization, including lower progesterone responsiveness in the model. These findings may partly contribute to the interpretation of the beneficial effects of TSS on infertility.
Collapse
Affiliation(s)
- Kiyoshi Terawaki
- Tsumura Kampo Research Laboratories, Kampo Research & Development Division, Tsumura & Co., 3586 Yoshiwara, Ami-machi, Inashiki-gun, Ibaraki, 300-1192, Japan.
| | - Yayoi Saegusa
- Tsumura Kampo Research Laboratories, Kampo Research & Development Division, Tsumura & Co., 3586 Yoshiwara, Ami-machi, Inashiki-gun, Ibaraki, 300-1192, Japan
| | - Kyoji Sekiguchi
- Tsumura Kampo Research Laboratories, Kampo Research & Development Division, Tsumura & Co., 3586 Yoshiwara, Ami-machi, Inashiki-gun, Ibaraki, 300-1192, Japan
| | - Tomofumi Shimizu
- Tsumura Kampo Research Laboratories, Kampo Research & Development Division, Tsumura & Co., 3586 Yoshiwara, Ami-machi, Inashiki-gun, Ibaraki, 300-1192, Japan
| | - Mikina Takiyama
- Tsumura Kampo Research Laboratories, Kampo Research & Development Division, Tsumura & Co., 3586 Yoshiwara, Ami-machi, Inashiki-gun, Ibaraki, 300-1192, Japan
| | - Takashi Matsumoto
- Tsumura Kampo Research Laboratories, Kampo Research & Development Division, Tsumura & Co., 3586 Yoshiwara, Ami-machi, Inashiki-gun, Ibaraki, 300-1192, Japan
| | - Seiichi Iizuka
- Tsumura Kampo Research Laboratories, Kampo Research & Development Division, Tsumura & Co., 3586 Yoshiwara, Ami-machi, Inashiki-gun, Ibaraki, 300-1192, Japan
| | - Chinami Matsumoto
- Tsumura Kampo Research Laboratories, Kampo Research & Development Division, Tsumura & Co., 3586 Yoshiwara, Ami-machi, Inashiki-gun, Ibaraki, 300-1192, Japan.
| | | |
Collapse
|
29
|
The Regulation of Ferroptosis by Tumor Suppressor p53 and its Pathway. Int J Mol Sci 2020; 21:ijms21218387. [PMID: 33182266 PMCID: PMC7664917 DOI: 10.3390/ijms21218387] [Citation(s) in RCA: 176] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 11/01/2020] [Accepted: 11/04/2020] [Indexed: 12/11/2022] Open
Abstract
Tumor suppressor p53 plays a key role in tumor suppression. In addition to tumor suppression, p53 is also involved in many other biological and pathological processes, such as immune response, maternal reproduction, tissue ischemia/reperfusion injuries and neurodegenerative diseases. While it has been widely accepted that the role of p53 in regulation of cell cycle arrest, senescence and apoptosis contributes greatly to the function of p53 in tumor suppression, emerging evidence has implicated that p53 also exerts its tumor suppressive function through regulation of many other cellular processes, such as metabolism, anti-oxidant defense and ferroptosis. Ferroptosis is a unique iron-dependent form of programmed cell death driven by lipid peroxidation in cells. Ferroptosis has been reported to be involved in cancer, tissue ischemia/reperfusion injuries and neurodegenerative diseases. Recent studies have shown that ferroptosis can be regulated by p53 and its signaling pathway as well as tumor-associated mutant p53. Interestingly, the regulation of ferroptosis by p53 appears to be highly context-dependent. In this review, we summarize recent advances in the regulation of ferroptosis by p53 and its signaling pathway. Further elucidation of the role and molecular mechanism of p53 in ferroptosis regulation will yield new therapeutic strategies for cancer and other diseases, including neurodegenerative diseases and tissue ischemia/reperfusion injuries.
Collapse
|
30
|
Khokhlova EV, Fesenko ZS, Sopova JV, Leonova EI. Features of DNA Repair in the Early Stages of Mammalian Embryonic Development. Genes (Basel) 2020; 11:genes11101138. [PMID: 32992616 PMCID: PMC7599644 DOI: 10.3390/genes11101138] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 09/20/2020] [Accepted: 09/25/2020] [Indexed: 12/12/2022] Open
Abstract
Cell repair machinery is responsible for protecting the genome from endogenous and exogenous effects that induce DNA damage. Mutations that occur in somatic cells lead to dysfunction in certain tissues or organs, while a violation of genomic integrity during the embryonic period often leads to death. A mammalian embryo’s ability to respond to damaged DNA and repair it, as well as its sensitivity to specific lesions, is still not well understood. In this review, we combine disparate data on repair processes in the early stages of preimplantation development in mammalian embryos.
Collapse
Affiliation(s)
- Evgenia V. Khokhlova
- Institute of Translational Biomedicine, St. Petersburg State University, 199034 St. Petersburg, Russia; (E.V.K.); (Z.S.F.); (J.V.S.)
- Institute of Cytology of the Russian Academy of Sciences, 194064 St. Petersburg, Russia
| | - Zoia S. Fesenko
- Institute of Translational Biomedicine, St. Petersburg State University, 199034 St. Petersburg, Russia; (E.V.K.); (Z.S.F.); (J.V.S.)
| | - Julia V. Sopova
- Institute of Translational Biomedicine, St. Petersburg State University, 199034 St. Petersburg, Russia; (E.V.K.); (Z.S.F.); (J.V.S.)
- Laboratory of Amyloid Biology, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Elena I. Leonova
- Institute of Translational Biomedicine, St. Petersburg State University, 199034 St. Petersburg, Russia; (E.V.K.); (Z.S.F.); (J.V.S.)
- Preclinical Research Center, University of Science and Technology, 1 Olympic Ave, 354340 Sochi, Russia
- Correspondence: ; Tel.: +8-(999)-232-92-58
| |
Collapse
|
31
|
Liu J, Zhang C, Wang X, Hu W, Feng Z. Tumor suppressor p53 cross-talks with TRIM family proteins. Genes Dis 2020; 8:463-474. [PMID: 34179310 PMCID: PMC8209353 DOI: 10.1016/j.gendis.2020.07.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/06/2020] [Accepted: 07/07/2020] [Indexed: 12/11/2022] Open
Abstract
p53 is a key tumor suppressor. As a transcription factor, p53 accumulates in cells in response to various stress signals and selectively transcribes its target genes to regulate a wide variety of cellular stress responses to exert its function in tumor suppression. In addition to tumor suppression, p53 is also involved in many other physiological and pathological processes, e.g. anti-infection, immune response, development, reproduction, neurodegeneration and aging. To maintain its proper function, p53 is under tight and delicate regulation through different mechanisms, particularly the posttranslational modifications. The tripartite motif (TRIM) family proteins are a large group of proteins characterized by the RING, B-Box and coiled-coil (RBCC) domains at the N-terminus. TRIM proteins play important roles in regulation of many fundamental biological processes, including cell proliferation and death, DNA repair, transcription, and immune response. Alterations of TRIM proteins have been linked to many diseases including cancer, infectious diseases, developmental disorders, and neurodegeneration. Interestingly, recent studies have revealed that many TRIM proteins are involved in the regulation of p53, and at the same time, many TRIM proteins are also regulated by p53. Here, we review the cross-talk between p53 and TRIM proteins, and its impact upon cellular biological processes as well as cancer and other diseases.
Collapse
Affiliation(s)
- Juan Liu
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers-State University of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA
| | - Cen Zhang
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers-State University of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA
| | - Xue Wang
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers-State University of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA
| | - Wenwei Hu
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers-State University of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA
| | - Zhaohui Feng
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers-State University of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA
| |
Collapse
|
32
|
Liu J, Zhang C, Hu W, Feng Z. Tumor suppressor p53 and metabolism. J Mol Cell Biol 2020; 11:284-292. [PMID: 30500901 PMCID: PMC6487777 DOI: 10.1093/jmcb/mjy070] [Citation(s) in RCA: 202] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/19/2018] [Accepted: 11/29/2018] [Indexed: 12/13/2022] Open
Abstract
p53 plays a key role in tumor suppression. The tumor suppressive function of p53 has long been attributed to its ability to induce apoptosis, cell cycle arrest, and senescence in cells. However, recent studies suggest that other functions of p53 also contribute to its role as a tumor suppressor, such as its function in metabolic regulation. p53 regulates various metabolic pathways to maintain the metabolic homeostasis of cells and adapt cells to stress. In addition, recent studies have also shown that gain-of-function (GOF) mutant p53 proteins drive metabolic reprogramming in cancer cells, contributing to cancer progression. Further understanding of p53 and its GOF mutants in metabolism will provide new opportunities for cancer therapy.
Collapse
Affiliation(s)
- Juan Liu
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, State University of New Jersey, New Brunswick, NJ, USA
| | - Cen Zhang
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, State University of New Jersey, New Brunswick, NJ, USA
| | - Wenwei Hu
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, State University of New Jersey, New Brunswick, NJ, USA.,Department of Pharmacology, Rutgers Cancer Institute of New Jersey, Rutgers University, State University of New Jersey, New Brunswick, NJ, USA
| | - Zhaohui Feng
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, State University of New Jersey, New Brunswick, NJ, USA.,Department of Pharmacology, Rutgers Cancer Institute of New Jersey, Rutgers University, State University of New Jersey, New Brunswick, NJ, USA
| |
Collapse
|
33
|
Rubin JB, Lagas JS, Broestl L, Sponagel J, Rockwell N, Rhee G, Rosen SF, Chen S, Klein RS, Imoukhuede P, Luo J. Sex differences in cancer mechanisms. Biol Sex Differ 2020; 11:17. [PMID: 32295632 PMCID: PMC7161126 DOI: 10.1186/s13293-020-00291-x] [Citation(s) in RCA: 178] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 03/18/2020] [Indexed: 02/07/2023] Open
Abstract
We now know that cancer is many different diseases, with great variation even within a single histological subtype. With the current emphasis on developing personalized approaches to cancer treatment, it is astonishing that we have not yet systematically incorporated the biology of sex differences into our paradigms for laboratory and clinical cancer research. While some sex differences in cancer arise through the actions of circulating sex hormones, other sex differences are independent of estrogen, testosterone, or progesterone levels. Instead, these differences are the result of sexual differentiation, a process that involves genetic and epigenetic mechanisms, in addition to acute sex hormone actions. Sexual differentiation begins with fertilization and continues beyond menopause. It affects virtually every body system, resulting in marked sex differences in such areas as growth, lifespan, metabolism, and immunity, all of which can impact on cancer progression, treatment response, and survival. These organismal level differences have correlates at the cellular level, and thus, males and females can fundamentally differ in their protections and vulnerabilities to cancer, from cellular transformation through all stages of progression, spread, and response to treatment. Our goal in this review is to cover some of the robust sex differences that exist in core cancer pathways and to make the case for inclusion of sex as a biological variable in all laboratory and clinical cancer research. We finish with a discussion of lab- and clinic-based experimental design that should be used when testing whether sex matters and the appropriate statistical models to apply in data analysis for rigorous evaluations of potential sex effects. It is our goal to facilitate the evaluation of sex differences in cancer in order to improve outcomes for all patients.
Collapse
Affiliation(s)
- Joshua B Rubin
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA.
- Department of Neuroscience, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA.
| | - Joseph S Lagas
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Lauren Broestl
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Jasmin Sponagel
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Nathan Rockwell
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Gina Rhee
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Sarah F Rosen
- Department of Medicine, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Si Chen
- Department of Biomedical Engineering, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Robyn S Klein
- Department of Neuroscience, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
- Department of Medicine, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Princess Imoukhuede
- Department of Biomedical Engineering, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| | - Jingqin Luo
- Department of Surgery, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, 63110, USA
| |
Collapse
|
34
|
Cai Y, Lei X, Chen Z, Mo Z. The roles of cirRNA in the development of germ cells. Acta Histochem 2020; 122:151506. [PMID: 32008790 DOI: 10.1016/j.acthis.2020.151506] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 12/27/2019] [Accepted: 01/02/2020] [Indexed: 12/12/2022]
Abstract
Circular RNA (CircRNA), a type of endogenous non-coding RNAs (ncRNAs), is generally generated from precursor mRNA (pre-mRNA) by canonical splicing and head-to-tail back splicing. The structure without a polyA tail renders circRNA highly insensitive to ribonuclease. Simultaneously, the distribution of circRNAs is tissue and developmental stage-specific. There are five potential biological functions of circRNAs: 1) promote transcription of their parental genes; 2) function as a miRNA sponge; 3) RNA binding protein (RBP) sponge; 4) encode protein; 5) act as an mRNA trap. Recently, circRNA has attracted attention because studies have shown that circRNAs are associated with follicular development, ovarian senescence, spermatogenesis, and germ cell development process, suggesting that circRNAs may function in germ cells regulation. The investigation of circRNAs in germ cells will provide an excellent opportunity to understand its potential molecular basis, and potentially improving reproduction status in human. In this article, the relationship between circRNA and germ cell development will be discussed.
Collapse
Affiliation(s)
- Yaqin Cai
- Clinical Anatomy & Reproductive Medicine Application Institute, Department of Histology and Embryology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; Institute of Basic Medical Sciences, Center for Diabetic Systems Medicine (Guangxi Key Laboratory of Excellence), Guilin Medical University, Guangxi, Guilin, 541100, China
| | - Xiaocan Lei
- Clinical Anatomy & Reproductive Medicine Application Institute, Department of Histology and Embryology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Zhuo Chen
- Hunan Province Innovative Training Base for Medical Postgraduates, University of South China and Yueyang Women & Children's Medical Center, Institute of Reproductive Medicine, Yueyang, Hunan, 416000, China
| | - Zhongcheng Mo
- Institute of Basic Medical Sciences, Center for Diabetic Systems Medicine (Guangxi Key Laboratory of Excellence), Guilin Medical University, Guangxi, Guilin, 541100, China; Hunan Province Innovative Training Base for Medical Postgraduates, University of South China and Yueyang Women & Children's Medical Center, Institute of Reproductive Medicine, Yueyang, Hunan, 416000, China.
| |
Collapse
|
35
|
Charni-Natan M, Aloni-Grinstein R, Osher E, Rotter V. Liver and Steroid Hormones-Can a Touch of p53 Make a Difference? Front Endocrinol (Lausanne) 2019; 10:374. [PMID: 31244779 PMCID: PMC6581675 DOI: 10.3389/fendo.2019.00374] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 05/28/2019] [Indexed: 12/12/2022] Open
Abstract
The liver is the main metabolic organ in the body, serving as a significant hormonal secretory gland and functioning to maintain hormone balance and homeostasis. Steroid hormones regulate various biological pathways, mainly in the reproductive system and in many metabolic processes. The liver, as well as steroid hormones, contribute significantly, through functional intertwine, to homeostasis maintenance, and proper responses during stress. Malfunction of either has a significant impact on the other and may lead to severe liver diseases as well as to several endocrine syndromes. Thus, the regulation on liver functions as on steroid hormones levels and activities is well-controlled. p53, the well-known tumor suppressor gene, was recently found to regulate metabolism and general homeostasis processes, particularly within the liver. Moreover, p53 was shown to be involved in steroid hormones regulation. In this review, we discuss the bi-directional regulation of the liver and the steroid hormones pointing to p53 as a novel regulator in this axis. A comprehensive understanding of the molecular mechanisms of this axis may help to prevent and treat related disease, especially with the increasing exposure of the population to environmental steroid hormones and steroid hormone-based medication.
Collapse
Affiliation(s)
- Meital Charni-Natan
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ronit Aloni-Grinstein
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Etty Osher
- Sackler Faculty of Medicine, Tel Aviv-Sourasky Medical Center, Institute of Endocrinology Metabolism and Hypertension, Tel Aviv University, Tel Aviv, Israel
| | - Varda Rotter
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
36
|
Treatment of buffalo (Bubalus bubalis) SCNT embryos with microRNA-21 mimic improves their quality and alters gene expression but does not affect their developmental competence. Theriogenology 2019; 126:8-16. [DOI: 10.1016/j.theriogenology.2018.11.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/20/2018] [Accepted: 11/25/2018] [Indexed: 01/15/2023]
|
37
|
Abecia JA, Forcada F, Vázquez MI, Muiño-Blanco T, Cebrián-Pérez JA, Pérez-Pe R, Casao A. Role of melatonin on embryo viability in sheep. Reprod Fertil Dev 2019; 31:82-92. [PMID: 32188544 DOI: 10.1071/rd18308] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Melatonin is a natural hormone synthesised in the pineal gland, the activity of which is regulated by day-night perception and dictates seasonal rhythms in reproduction in ovine species. Exogenous melatonin, administered via subcutaneous implants, is used to prolong the breeding season of ewes and can increase the proportion of pregnant ewes (fertility rate) and litter size. The increased proportion of ewes that become pregnant and the number of lambs born per lambing among melatonin-treated sheep may be caused by increased embryo survival, through enhanced luteal function, reduced antiluteolytic mechanisms, or improved embryo quality. This review focuses on the effects of melatonin on embryo viability and summarises the processes by which this hormone affects the ovary, follicle, oocyte, corpus luteum and embryo. Moreover, the effects of melatonin on the mechanisms of invivo maternal recognition of pregnancy in sheep and the protective action that it appears to have on the invitro procedures that are used to obtain healthy embryos are reviewed.
Collapse
Affiliation(s)
- José-Alfonso Abecia
- Instituto Universitario de Investigación en Ciencias Ambientales de Aragón (IUCA), Universidad de Zaragoza, Facultad de Veterinaria, Miguel Servet, 177, 50013 Zaragoza, Spain
| | - Fernando Forcada
- Instituto Universitario de Investigación en Ciencias Ambientales de Aragón (IUCA), Universidad de Zaragoza, Facultad de Veterinaria, Miguel Servet, 177, 50013 Zaragoza, Spain
| | - María-Isabel Vázquez
- Departamento de Reproducción Animal, Facultad de Agronomía y Veterinaria, Universidad Nacional de Río Cuarto, Ruta Nacional 36, Km 601, 5800 Río Cuarto, Córdoba, Argentina
| | - Teresa Muiño-Blanco
- Instituto Universitario de Investigación en Ciencias Ambientales de Aragón (IUCA), Universidad de Zaragoza, Facultad de Veterinaria, Miguel Servet, 177, 50013 Zaragoza, Spain
| | - José A Cebrián-Pérez
- Instituto Universitario de Investigación en Ciencias Ambientales de Aragón (IUCA), Universidad de Zaragoza, Facultad de Veterinaria, Miguel Servet, 177, 50013 Zaragoza, Spain
| | - Rosaura Pérez-Pe
- Instituto Universitario de Investigación en Ciencias Ambientales de Aragón (IUCA), Universidad de Zaragoza, Facultad de Veterinaria, Miguel Servet, 177, 50013 Zaragoza, Spain
| | - Adriana Casao
- Instituto Universitario de Investigación en Ciencias Ambientales de Aragón (IUCA), Universidad de Zaragoza, Facultad de Veterinaria, Miguel Servet, 177, 50013 Zaragoza, Spain
| |
Collapse
|
38
|
Liu C, Cai Z, Jin G, Peng D, Pan BS, Zhang X, Han F, Xu X, Lin HK. Abnormal gametogenesis induced by p53 deficiency promotes tumor progression and drug resistance. Cell Discov 2018; 4:54. [PMID: 30302273 PMCID: PMC6167385 DOI: 10.1038/s41421-018-0054-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 07/08/2018] [Accepted: 07/12/2018] [Indexed: 01/15/2023] Open
Abstract
The century-old embryonal/gametogenesis hypothesis of tumors could link diverse tumors' malignant features together likely representing the real "stemness" of tumors. However, the genetic evidence to validate abnormal gametogenesis in tumors remains lacking. Here we show that p53 deficiency elicits abnormal gametogenesis from primordial germ cell-like stage to late oocyte-like stage and subsequent parthenogenetic activation. The similar upregulation of abnormal gametogenesis by p53 deficiency is observed both in p53-/- mouse model and cultured cancer cells. Notably, germ cell-like cells isolated from distinct tumors from p53-/- mice and cancer cell lines display potent tumorigenicity potential. Abnormal oogenesis induced by p53 deficiency and then spontaneous parthenogenetic activation endow tumors with imitated embryonic development, life cycle, and therapeutic resistance. Our study establishes the genetic evidence to support embryonal/gametogenesis theory of tumors and reveals a pivotal role of p53 in restricting abnormal gametogenesis that may represent a novel aspect for p53's tumor suppression.
Collapse
Affiliation(s)
- Chunfang Liu
- 1Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040 China
- 2Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- 3Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Zhen Cai
- 2Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- 3Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Guoxiang Jin
- 2Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- 3Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Danni Peng
- 2Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- 3Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Bo-Syong Pan
- 2Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- 3Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Xian Zhang
- 2Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- 3Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Fei Han
- 2Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- 3Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Xiaohong Xu
- 3Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Hui-Kuan Lin
- 2Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157 USA
- 3Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
- 4Graduate Institute of Basic Medical Science, China Medical University, Taichung, 404 Taiwan
- 5Department of Biotechnology, Asia University, Taichung, 41354 Taiwan
| |
Collapse
|
39
|
Tao H, Xiong Q, Zhang F, Zhang N, Liu Y, Suo X, Li X, Yang Q, Chen M. Circular RNA profiling reveals chi_circ_0008219 function as microRNA sponges in pre-ovulatory ovarian follicles of goats (Capra hircus). Genomics 2017; 110:S0888-7543(17)30129-5. [PMID: 29107014 DOI: 10.1016/j.ygeno.2017.10.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 10/13/2017] [Accepted: 10/17/2017] [Indexed: 01/01/2023]
Abstract
Circular RNAs (circRNAs) are a new class of non-coding RNAs in animals and are a novel target of non-coding RNA (ncRNA) regulation. The mechanism and function of circRNAs have been reported in some species and tissues. However, there is little available information on the functions of circRNAs in the goat reproductive system. In the present study, we deeply sequenced and analyzed circRNAs through bioinformatics to reveal the expression profiles, and predicted 13,950 circRNAs in the pre-ovulatory ovarian follicles of goats for the first time. Thirty-seven circRNAs were differentially expressed in the Boer goat compared with the Macheng black goat. The chi_circ_0008219 was involved in a vast circRNA-miRNA-mRNA co-expression network. Via a luciferase activity assay, chi_circ_0008219 is observed to sponge to 3 ovarian follicle-related miRNAs. These findings demonstrate that circRNAs have potential effects in the ovarian follicles of ewes and may represent a promising new research field in ovarian follicular development.
Collapse
Affiliation(s)
- Hu Tao
- Hubei Key Laboratory of Animal Embryo Engineering and Molecular Breeding, Institute of Animal Husbandry and Veterinary, Hubei, Academy of Agricultural Sciences, Wuhan 430064, China
| | - Qi Xiong
- Hubei Key Laboratory of Animal Embryo Engineering and Molecular Breeding, Institute of Animal Husbandry and Veterinary, Hubei, Academy of Agricultural Sciences, Wuhan 430064, China
| | - Feng Zhang
- Hubei Key Laboratory of Animal Embryo Engineering and Molecular Breeding, Institute of Animal Husbandry and Veterinary, Hubei, Academy of Agricultural Sciences, Wuhan 430064, China
| | - Nian Zhang
- Hubei Key Laboratory of Animal Embryo Engineering and Molecular Breeding, Institute of Animal Husbandry and Veterinary, Hubei, Academy of Agricultural Sciences, Wuhan 430064, China
| | - Yang Liu
- Hubei Key Laboratory of Animal Embryo Engineering and Molecular Breeding, Institute of Animal Husbandry and Veterinary, Hubei, Academy of Agricultural Sciences, Wuhan 430064, China
| | - Xiaojun Suo
- Hubei Key Laboratory of Animal Embryo Engineering and Molecular Breeding, Institute of Animal Husbandry and Veterinary, Hubei, Academy of Agricultural Sciences, Wuhan 430064, China
| | - Xiaofeng Li
- Hubei Key Laboratory of Animal Embryo Engineering and Molecular Breeding, Institute of Animal Husbandry and Veterinary, Hubei, Academy of Agricultural Sciences, Wuhan 430064, China
| | - Qianping Yang
- Hubei Key Laboratory of Animal Embryo Engineering and Molecular Breeding, Institute of Animal Husbandry and Veterinary, Hubei, Academy of Agricultural Sciences, Wuhan 430064, China
| | - Mingxin Chen
- Hubei Key Laboratory of Animal Embryo Engineering and Molecular Breeding, Institute of Animal Husbandry and Veterinary, Hubei, Academy of Agricultural Sciences, Wuhan 430064, China.
| |
Collapse
|
40
|
Dai MS, Hall SJ, Vantangoli Policelli MM, Boekelheide K, Spade DJ. Spontaneous testicular atrophy occurs despite normal spermatogonial proliferation in a Tp53 knockout rat. Andrology 2017; 5:1141-1152. [PMID: 28834365 DOI: 10.1111/andr.12409] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 06/12/2017] [Accepted: 06/27/2017] [Indexed: 12/19/2022]
Abstract
The tumor suppressor protein p53 (TP53) has many functions in cell cycle regulation, apoptosis, and DNA damage repair and is also involved in spermatogenesis in the mouse. To evaluate the role of p53 in spermatogenesis in the rat, we characterized testis biology in adult males of a novel p53 knockout rat (SD-Tp53tm1sage ). p53 knockout rats exhibited variable levels of testicular atrophy, including significantly decreased testis weights, atrophic seminiferous tubules, decreased seminiferous tubule diameter, and elevated spermatocyte TUNEL labeling rates, indicating a dysfunction in spermatogenesis. Phosphorylated histone H2AX protein levels and distribution were similar in the non-atrophic seminiferous tubules of both genotypes, showing evidence of pre-synaptic DNA double-strand breaks in leptotene and zygotene spermatocytes, preceding cell death in p53 knockout rat testes. Quantification of the spermatogonial stem cell (SSC) proliferation rate with bromodeoxyuridine (BrdU) labeling, in addition to staining with the undifferentiated type A spermatogonial marker GDNF family receptor alpha-1 (GFRA1), indicated that the undifferentiated spermatogonial population was normal in p53 knockout rats. Following exposure to 0.5 or 5 Gy X-ray, p53 knockout rats exhibited no germ cell apoptotic response beyond their unirradiated phenotype, while germ cell death in wild-type rat testes was elevated to a level similar to the unexposed p53 knockout rats. This study indicates that seminiferous tubule atrophy occurs following spontaneous, elevated levels of spermatocyte death in the p53 knockout rat. This phenomenon is variable across individual rats. These results indicate a critical role for p53 in rat germ cell survival and spermatogenesis.
Collapse
Affiliation(s)
- Matthew S Dai
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Susan J Hall
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | | | - Kim Boekelheide
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Daniel J Spade
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| |
Collapse
|
41
|
Yue X, Zhao Y, Xu Y, Zheng M, Feng Z, Hu W. Mutant p53 in Cancer: Accumulation, Gain-of-Function, and Therapy. J Mol Biol 2017; 429:1595-1606. [PMID: 28390900 PMCID: PMC5663274 DOI: 10.1016/j.jmb.2017.03.030] [Citation(s) in RCA: 205] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 03/31/2017] [Accepted: 03/31/2017] [Indexed: 12/19/2022]
Abstract
Tumor suppressor p53 plays a central role in tumor suppression. p53 is the most frequently mutated gene in human cancer, and over half of human cancers contain p53 mutations. Majority of p53 mutations in cancer are missense mutations, leading to the expression of full-length mutant p53 (mutp53) protein. While the critical role of wild-type p53 in tumor suppression has been firmly established, mounting evidence has demonstrated that many tumor-associated mutp53 proteins not only lose the tumor-suppressive function of wild-type p53 but also gain new activities to promote tumorigenesis independently of wild-type p53, termed gain-of-function. Mutant p53 protein often accumulates to very high levels in tumors, contributing to malignant progression. Recently, mutp53 has become an attractive target for cancer therapy. Further understanding of the mechanisms underlying mutp53 protein accumulation and gain-of-function will accelerate the development of targeted therapies for human cancer harboring mutp53. In this review, we summarize the recent advances in the studies on mutp53 protein accumulation and gain-of-function and targeted therapies for mutp53 in human cancer.
Collapse
Affiliation(s)
- Xuetian Yue
- Rutgers Cancer Institute of New Jersey, Rutgers, the State University of New Jersey, New Brunswick, NJ 08903, USA
| | - Yuhan Zhao
- Rutgers Cancer Institute of New Jersey, Rutgers, the State University of New Jersey, New Brunswick, NJ 08903, USA
| | - Yang Xu
- Department of Hematology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Min Zheng
- State Key Lab of Diagnostic and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Zhaohui Feng
- Rutgers Cancer Institute of New Jersey, Rutgers, the State University of New Jersey, New Brunswick, NJ 08903, USA; Department of Pharmacology, Rutgers, the State University of New Jersey, New Brunswick, NJ 08903, USA.
| | - Wenwei Hu
- Rutgers Cancer Institute of New Jersey, Rutgers, the State University of New Jersey, New Brunswick, NJ 08903, USA; Department of Pharmacology, Rutgers, the State University of New Jersey, New Brunswick, NJ 08903, USA.
| |
Collapse
|
42
|
Salemi M, Barone C, Salluzzo MG, Giambirtone M, Scillato F, Galati Rando R, Romano C, Morale MC, Ridolfo F, Romano C. A polymorphism (rs1042522) in TP53 gene is a risk factor for Down Syndrome in Sicilian mothers. J Matern Fetal Neonatal Med 2016; 30:2752-2754. [PMID: 27852141 DOI: 10.1080/14767058.2016.1262343] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Trisomy 21 is the most frequent genetic cause of intellectual disability. Tumor Protein 53 (TP53) gene down-regulation triggers chromosomal instability. A TP53 gene polymorphism c.215G > C (rs1042522) is associated with accumulation of aneuploid cells. We analyzed the TP53 c.215G > C (rs1042522) polymorphism in Sicilian mothers of subjects with Down Syndrome (DS) within a case-control study. METHODS Nucleotide polymorphism was detected by pyrosequencing technology. RESULTS The distribution of TP53 c.215G > C polymorphism showed significant difference between mothers of subjects with DS and controls. CONCLUSIONS Our data show that TP53 c.215G > C polymorphism is a risk factor for DS in Sicilian mothers.
Collapse
Affiliation(s)
- Michele Salemi
- a IRCCS Associazione Oasi Institute for Research on Mental Retardation and Brain Aging , Troina , Italy
| | - Concetta Barone
- a IRCCS Associazione Oasi Institute for Research on Mental Retardation and Brain Aging , Troina , Italy
| | - Maria Grazia Salluzzo
- a IRCCS Associazione Oasi Institute for Research on Mental Retardation and Brain Aging , Troina , Italy
| | - Mariaconcetta Giambirtone
- a IRCCS Associazione Oasi Institute for Research on Mental Retardation and Brain Aging , Troina , Italy
| | - Francesco Scillato
- a IRCCS Associazione Oasi Institute for Research on Mental Retardation and Brain Aging , Troina , Italy
| | - Rosanna Galati Rando
- a IRCCS Associazione Oasi Institute for Research on Mental Retardation and Brain Aging , Troina , Italy
| | - Carmelo Romano
- a IRCCS Associazione Oasi Institute for Research on Mental Retardation and Brain Aging , Troina , Italy
| | - Maria Concetta Morale
- a IRCCS Associazione Oasi Institute for Research on Mental Retardation and Brain Aging , Troina , Italy
| | - Federico Ridolfo
- b Post-Graduate School in Clinical Biochemistry , University of Milan , Milan , Italy
| | - Corrado Romano
- a IRCCS Associazione Oasi Institute for Research on Mental Retardation and Brain Aging , Troina , Italy
| |
Collapse
|
43
|
Influence of TP53 Codon 72 Polymorphism Alone or in Combination with HDM2 SNP309 on Human Infertility and IVF Outcome. PLoS One 2016; 11:e0167147. [PMID: 27898708 PMCID: PMC5127557 DOI: 10.1371/journal.pone.0167147] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Accepted: 11/09/2016] [Indexed: 11/19/2022] Open
Abstract
To evaluate the association of the TP53 codon 72 (rs 1042522) alone or in combination with HDM2 SNP309 (rs 2279744) polymorphisms with human infertility and IVF outcome, we collected 1450 infertility women undergoing their first controlled ovarian stimulation for IVF treatment and 250 fertile controls in the case-control study. Frequencies, distribution, interaction of genes, and correlation with infertility and IVF outcome of clinical pregnancy were analyzed. We found a statistically significant association between TP53 codon 72 polymorphism and IVF outcome (52.10% vs. 47.40%, OR = 0.83, 95%CI:0.71–0.96, p = 0.01). No significant difference was shown between TP53 codon 72, HDM2 SNP309 polymorphisms, human infertility, and between the combination of two genes polymorphisms and the clinical pregnancy outcome of IVF. The data support C allele as a protective factor for IVF pregnancy outcome. Further researches should be focused on the mechanism of these associations.
Collapse
|
44
|
Chen LJ, Xu WM, Yang M, Wang K, Chen Y, Huang XJ, Ma QH. HUWE1 plays important role in mouse preimplantation embryo development and the dysregulation is associated with poor embryo development in humans. Sci Rep 2016; 6:37928. [PMID: 27901130 PMCID: PMC5128802 DOI: 10.1038/srep37928] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 11/02/2016] [Indexed: 11/15/2022] Open
Abstract
HUWE1 is a HECT domain containing ubiquitin ligase implicated in neurogenesis, spermatogenesis and cancer development. The purpose of the current study is to investigate the role of HUWE1 in early embryo development. Here we demonstrate that Huwe1 is expressed in both nucleus and cytoplasm of preimplantation mouse embryos as well as gametes. Hypoxia (5% O2) treatment could significantly increase Huwe1 expression during mouse embryo development process. HUWE1 knockdown inhibited normal embryonic development and reduced blastocyst formation, and increased apoptotic cell numbers were observed in the embryos of HUWE1 knockdown group. Human embryo staining result showed that reduced HUWE1 staining was observed in the poor-quality embryos. Furthermore, Western blot result showed that significantly reduced expression of HUWE1 was observed in the villi of miscarriage embryos compared with the normal control, indicating that reduced expression of HUWE1 is related to poor embryo development. Oxidative reagent, H2O2 inhibited HUWE1 expression in human sperm, indicating that HUWE1 expression in sperm is regulated by oxidative stress. In conclusion, these results suggest that HUWE1 protein could contribute to preimplantation embryo development and dysregulated expression of HUWE1 could be related to poor embryo development and miscarriage in IVF clinic.
Collapse
Affiliation(s)
- L J Chen
- Department of Obstetric and Gynecologic diseases, West China Second University Hospital, Sichuan University, Chengdu 610041, P.R. China.,SCU-CUHK Joint Laboratory of Reproductive Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, (Sichuan University), West China Second University Hospital, Sichuan University, Chengdu 610041, P.R. China
| | - W M Xu
- Department of Obstetric and Gynecologic diseases, West China Second University Hospital, Sichuan University, Chengdu 610041, P.R. China.,SCU-CUHK Joint Laboratory of Reproductive Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, (Sichuan University), West China Second University Hospital, Sichuan University, Chengdu 610041, P.R. China
| | - M Yang
- Department of Obstetric and Gynecologic diseases, West China Second University Hospital, Sichuan University, Chengdu 610041, P.R. China.,SCU-CUHK Joint Laboratory of Reproductive Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, (Sichuan University), West China Second University Hospital, Sichuan University, Chengdu 610041, P.R. China
| | - K Wang
- Department of Obstetric and Gynecologic diseases, West China Second University Hospital, Sichuan University, Chengdu 610041, P.R. China.,SCU-CUHK Joint Laboratory of Reproductive Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, (Sichuan University), West China Second University Hospital, Sichuan University, Chengdu 610041, P.R. China
| | - Y Chen
- Department of Obstetric and Gynecologic diseases, West China Second University Hospital, Sichuan University, Chengdu 610041, P.R. China.,SCU-CUHK Joint Laboratory of Reproductive Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, (Sichuan University), West China Second University Hospital, Sichuan University, Chengdu 610041, P.R. China
| | - X J Huang
- College of Animal Science &Technology, Nanjing Agriculture University, Nanjing, China
| | - Q H Ma
- Department of Obstetric and Gynecologic diseases, West China Second University Hospital, Sichuan University, Chengdu 610041, P.R. China.,SCU-CUHK Joint Laboratory of Reproductive Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, (Sichuan University), West China Second University Hospital, Sichuan University, Chengdu 610041, P.R. China
| |
Collapse
|
45
|
Xie T, Yu CH, Zheng Y, A ZC. The polymorphism G4C14-to-A4T14 in p73 gene may affect the susceptibility to male infertility with severe spermatogenesis impairment in Chinese population. Eur J Obstet Gynecol Reprod Biol 2016; 204:74-7. [PMID: 27525684 DOI: 10.1016/j.ejogrb.2016.07.508] [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: 03/10/2016] [Revised: 06/07/2016] [Accepted: 07/26/2016] [Indexed: 10/21/2022]
Abstract
OBJECTIVE The aim of this study was to explore the association between the polymorphism G4C14-to-A4T14 in the p73 gene and male infertility with severe spermatogenesis impairment in Chinese population. STUDY DESIGN Three hundreds and one infertile patients with severe spermatogenesis impairment (including azoospermia and severe oligospermia) and 252 fertile men were recruited in this study. The polymorphism G4C14-to-A4T14 in the p73 gene was genotyped using polymerase chain reaction and restriction fragment length polymorphism assay. The differences in allelic and genotypic frequencies between patients and controls were evaluated by chi-square test. RESULTS The frequency of allele AT (28.9% vs. 22.4%, P=0.017, OR=1.41, 95% CI=1.07-1.85) in patients with severe spermatogenesis impairment was significantly higher than that in controls, whereas the genotype GC/GC was significantly decreased in patients compared with controls (48.5% vs. 59.1%, P=0.048, OR=0.65, 95% CI=0.46-0.91). CONCULUSION The findings of this study suggested that the polymorphism G4C14-to-A4T14 in p73 gene might be associated with severe spermatogenesis impairment and could affect the susceptibility to male infertility with severe spermatogenesis impairment in Chinese population.
Collapse
Affiliation(s)
- Ting Xie
- College of Basic Medicine, Dali University, Dali, 671000, China
| | - Cheng-He Yu
- College of Basic Medicine, Dali University, Dali, 671000, China; Department of Reproductive Medicine, Affiliated Hospital of Dali University, Dali, 671000, China
| | - Ye Zheng
- Department of Genetics, College of Agriculture and Biology, Dali University, Dali, 671003, China
| | - Zhou-Cun A
- College of Basic Medicine, Dali University, Dali, 671000, China; Department of Genetics, College of Agriculture and Biology, Dali University, Dali, 671003, China.
| |
Collapse
|
46
|
Boddy AM, Kokko H, Breden F, Wilkinson GS, Aktipis CA. Cancer susceptibility and reproductive trade-offs: a model of the evolution of cancer defences. Philos Trans R Soc Lond B Biol Sci 2016; 370:rstb.2014.0220. [PMID: 26056364 PMCID: PMC4581025 DOI: 10.1098/rstb.2014.0220] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The factors influencing cancer susceptibility and why it varies across species are major open questions in the field of cancer biology. One underexplored source of variation in cancer susceptibility may arise from trade-offs between reproductive competitiveness (e.g. sexually selected traits, earlier reproduction and higher fertility) and cancer defence. We build a model that contrasts the probabilistic onset of cancer with other, extrinsic causes of mortality and use it to predict that intense reproductive competition will lower cancer defences and increase cancer incidence. We explore the trade-off between cancer defences and intraspecific competition across different extrinsic mortality conditions and different levels of trade-off intensity, and find the largest effect of competition on cancer in species where low extrinsic mortality combines with strong trade-offs. In such species, selection to delay cancer and selection to outcompete conspecifics are both strong, and the latter conflicts with the former. We discuss evidence for the assumed trade-off between reproductive competitiveness and cancer susceptibility. Sexually selected traits such as ornaments or large body size require high levels of cell proliferation and appear to be associated with greater cancer susceptibility. Similar associations exist for female traits such as continuous egg-laying in domestic hens and earlier reproductive maturity. Trade-offs between reproduction and cancer defences may be instantiated by a variety of mechanisms, including higher levels of growth factors and hormones, less efficient cell-cycle control and less DNA repair, or simply a larger number of cell divisions (relevant when reproductive success requires large body size or rapid reproductive cycles). These mechanisms can affect intra- and interspecific variation in cancer susceptibility arising from rapid cell proliferation during reproductive maturation, intrasexual competition and reproduction.
Collapse
Affiliation(s)
- Amy M Boddy
- Department of Psychology, Arizona State University, Tempe, AZ, USA Center for Evolution and Cancer, University of California San Francisco, San Francisco, CA, USA Wissenschaftskolleg zu Berlin, Institute for Advanced Study, 14193 Berlin, Germany
| | - Hanna Kokko
- Wissenschaftskolleg zu Berlin, Institute for Advanced Study, 14193 Berlin, Germany Institute of Evolutionary Biology and Environmental Studies, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Felix Breden
- Wissenschaftskolleg zu Berlin, Institute for Advanced Study, 14193 Berlin, Germany Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia, Canada V5A 1S6
| | - Gerald S Wilkinson
- Wissenschaftskolleg zu Berlin, Institute for Advanced Study, 14193 Berlin, Germany Department of Biology, University of Maryland, College Park, MD 20742, USA
| | - C Athena Aktipis
- Department of Psychology, Arizona State University, Tempe, AZ, USA Center for Evolution and Cancer, University of California San Francisco, San Francisco, CA, USA Wissenschaftskolleg zu Berlin, Institute for Advanced Study, 14193 Berlin, Germany
| |
Collapse
|
47
|
Chen H, Yang X, Wang Z. Association between p53 Arg72Pro polymorphism and recurrent pregnancy loss: an updated systematic review and meta-analysis. Reprod Biomed Online 2015; 31:149-53. [DOI: 10.1016/j.rbmo.2015.05.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 05/01/2015] [Accepted: 05/05/2015] [Indexed: 11/26/2022]
|
48
|
Velazquez MA. Impact of maternal malnutrition during the periconceptional period on mammalian preimplantation embryo development. Domest Anim Endocrinol 2015; 51:27-45. [PMID: 25498236 DOI: 10.1016/j.domaniend.2014.10.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 10/27/2014] [Accepted: 10/28/2014] [Indexed: 02/07/2023]
Abstract
During episodes of undernutrition and overnutrition the mammalian preimplantation embryo undergoes molecular and metabolic adaptations to cope with nutrient deficits or excesses. Maternal adaptations also take place to keep a nutritional microenvironment favorable for oocyte development and embryo formation. This maternal-embryo communication takes place via several nutritional mediators. Although adaptive responses to malnutrition by both the mother and the embryo may ensure blastocyst formation, the resultant quality of the embryo can be compromised, leading to early pregnancy failure. Still, studies have shown that, although early embryonic mortality can be induced during malnutrition, the preimplantation embryo possesses an enormous plasticity that allows it to implant and achieve a full-term pregnancy under nutritional stress, even in extreme cases of malnutrition. This developmental strategy, however, may come with a price, as shown by the adverse developmental programming induced by even subtle nutritional challenges exerted exclusively during folliculogenesis and the preimplantation period, resulting in offspring with a higher risk of developing deleterious phenotypes in adulthood. Overall, current evidence indicates that malnutrition during the periconceptional period can induce cellular and molecular alterations in preimplantation embryos with repercussions for fertility and postnatal health.
Collapse
Affiliation(s)
- M A Velazquez
- Centre for Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK.
| |
Collapse
|
49
|
Vagnini LD, Renzi A, Oliveira-Pelegrin GR, Canas MDCT, Petersen CG, Mauri AL, Oliveira JBA, Baruffi RLR, Cavagna M, Franco Junior JG. The TP73 gene polymorphism (rs4648551, A>G) is associated with diminished ovarian reserve. PLoS One 2015; 10:e0120048. [PMID: 25794170 PMCID: PMC4368621 DOI: 10.1371/journal.pone.0120048] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 02/03/2015] [Indexed: 11/29/2022] Open
Abstract
It’s known that the members of the TP53 family are involved in the regulation of female reproduction. Studies in mice showed that the TP73 gene (member of this family) plays a role in the size of follicular pool, ovulation rate and maintenance of genomic stability. In the present study we analyzed data from 605 patients with ≤ 37 years attending their first intracytoplasmic sperm injection (ICSI). The association between the TP73 polymorphism (rs4648551, A>G) and the following parameters related to ovarian reserve, like age, antral follicular count (AFC), anti-Mullerian hormone levels (AMH) and ovarian response prediction index (ORPI) was evaluated. Our results showed an association of the AA genotype with diminished ovarian reserve (AMH <1, AFC ≤9). Women presenting the AA genotype had a 2.0-fold increased risk for having AMH <1 and AFC ≤9 (OR 2.0, 95% CI 1.23-3.31, P = 0.005). Patients presenting AA genotype had the lowest levels of AMH (P = 0.02), the lowest number of antral follicles (P = 0.01) and the lowest ORPI (P = 0.007). Analyzing the alleles, we can see an enrichment of the A allele in the group of diminished ovarian reserve (OR 1.4, 95%CI 1.02-1.83, P = 0.04). To the best of our knowledge, the present study is the first to analyze this polymorphism in humans for assessing the numbers of ovarian follicles and AMH levels and, therefore, the ovarian reserve. Our findings can contribute to the use of this polymorphism as a potential marker of diminished ovarian reserve.
Collapse
Affiliation(s)
- Laura Diniz Vagnini
- Paulista Center for Diagnosis, Research and Training, Ribeirão Preto, São Paulo, Brazil
| | - Adriana Renzi
- Paulista Center for Diagnosis, Research and Training, Ribeirão Preto, São Paulo, Brazil
| | | | | | | | - Ana Lucia Mauri
- Center for Human Reproduction Prof. Franco Jr, Ribeirão Preto, São Paulo, Brazil
| | | | | | - Mario Cavagna
- Center for Human Reproduction Prof. Franco Jr, Ribeirão Preto, São Paulo, Brazil
| | | |
Collapse
|
50
|
Yu H, Yue X, Zhao Y, Li X, Wu L, Zhang C, Liu Z, Lin K, Xu-Monette ZY, Young KH, Liu J, Shen Z, Feng Z, Hu W. LIF negatively regulates tumour-suppressor p53 through Stat3/ID1/MDM2 in colorectal cancers. Nat Commun 2014; 5:5218. [PMID: 25323535 PMCID: PMC4203416 DOI: 10.1038/ncomms6218] [Citation(s) in RCA: 148] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 09/10/2014] [Indexed: 12/21/2022] Open
Abstract
Leukaemia inhibitory factor (LIF) has been recently identified as a p53 target gene, which mediates the role of p53 in maternal implantation under normal physiological conditions. Here we report that LIF is a negative regulator of p53; LIF downregulates p53 protein levels and function in human colorectal cancer (CRC) cells. The downregulation of p53 by LIF is mediated by the activation of Stat3, which transcriptionally induces inhibitor of DNA-binding 1 (ID1). ID1 upregulates MDM2, a key negative regulator of p53, and promotes p53 protein degradation. LIF is overexpressed in a large percentage of CRCs. LIF overexpression promotes cellular resistance towards chemotherapeutic agents in cultured CRC cells and colorectal xenograft tumours in a largely p53-dependent manner. Overexpression of LIF is associated with a poor prognosis in CRC patients. Taken together, LIF is a novel negative regulator of p53, overexpression of LIF is an important mechanism for the attenuation of p53, which promotes chemoresistance in CRCs.
Collapse
Affiliation(s)
- Haiyang Yu
- Rutgers Cancer Institute of New Jersey, Rutgers the State University of New Jersey, New Brunswick, NJ 08903
| | - Xuetian Yue
- Rutgers Cancer Institute of New Jersey, Rutgers the State University of New Jersey, New Brunswick, NJ 08903
| | - Yuhan Zhao
- Rutgers Cancer Institute of New Jersey, Rutgers the State University of New Jersey, New Brunswick, NJ 08903
| | - Xiaoyan Li
- Rutgers Cancer Institute of New Jersey, Rutgers the State University of New Jersey, New Brunswick, NJ 08903
- Department of Breast Surgery, Qilu Hospital, Shandong University, Ji’nan, China
| | - Lihua Wu
- Rutgers Cancer Institute of New Jersey, Rutgers the State University of New Jersey, New Brunswick, NJ 08903
- First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Cen Zhang
- Rutgers Cancer Institute of New Jersey, Rutgers the State University of New Jersey, New Brunswick, NJ 08903
| | - Zhen Liu
- Rutgers Cancer Institute of New Jersey, Rutgers the State University of New Jersey, New Brunswick, NJ 08903
| | - Kevin Lin
- Rutgers Cancer Institute of New Jersey, Rutgers the State University of New Jersey, New Brunswick, NJ 08903
| | - Zijun Y. Xu-Monette
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030
| | - Ken H. Young
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030
| | - Juan Liu
- Rutgers Cancer Institute of New Jersey, Rutgers the State University of New Jersey, New Brunswick, NJ 08903
| | - Zhiyuan Shen
- Rutgers Cancer Institute of New Jersey, Rutgers the State University of New Jersey, New Brunswick, NJ 08903
| | - Zhaohui Feng
- Rutgers Cancer Institute of New Jersey, Rutgers the State University of New Jersey, New Brunswick, NJ 08903
| | - Wenwei Hu
- Rutgers Cancer Institute of New Jersey, Rutgers the State University of New Jersey, New Brunswick, NJ 08903
| |
Collapse
|