1
|
Qi W, Bai J, Wang R, Zeng X, Zhang L. SATB1, senescence and senescence-related diseases. J Cell Physiol 2024; 239:e31327. [PMID: 38801120 DOI: 10.1002/jcp.31327] [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: 01/31/2024] [Revised: 05/06/2024] [Accepted: 05/15/2024] [Indexed: 05/29/2024]
Abstract
Aging leads to an accumulation of cellular mutations and damage, increasing the risk of senescence, apoptosis, and malignant transformation. Cellular senescence, which is pivotal in aging, acts as both a guard against cellular transformation and as a check against cancer progression. It is marked by stable cell cycle arrest, widespread macromolecular changes, a pro-inflammatory profile, and altered gene expression. However, it remains to be determined whether these differing subsets of senescent cells result from unique intrinsic programs or are influenced by their environmental contexts. Multiple transcription regulators and chromatin modifiers contribute to these alterations. Special AT-rich sequence-binding protein 1 (SATB1) stands out as a crucial regulator in this process, orchestrating gene expression by structuring chromatin into loop domains and anchoring DNA elements. This review provides an overview of cellular senescence and delves into the role of SATB1 in senescence-related diseases. It highlights SATB1's potential in developing antiaging and anticancer strategies, potentially contributing to improved quality of life and addressing aging-related diseases.
Collapse
Affiliation(s)
- Wenjing Qi
- Department of Bioscience, Changchun Normal University, Changchun, Jilin, China
- Key Laboratory of Molecular Epigenetics of Ministry of Education, College of Life Sciences, Northeast Normal University, Changchun, Jilin, China
| | - Jinping Bai
- Department of Bioscience, Changchun Normal University, Changchun, Jilin, China
| | - Ruoxi Wang
- Center for Cell Structure and Function, College of Life Sciences, Key Laboratory of Animal Resistance Biology of Shandong Province, Shandong Normal University, Jinan, Shandong, China
| | - Xianlu Zeng
- Key Laboratory of Molecular Epigenetics of Ministry of Education, College of Life Sciences, Northeast Normal University, Changchun, Jilin, China
| | - Lihui Zhang
- Department of Bioscience, Changchun Normal University, Changchun, Jilin, China
| |
Collapse
|
2
|
Fondevila MF, Novoa E, Fernandez U, Dorta V, Parracho T, Kreimeyer H, Garcia-Vence M, Chantada-Vazquez MP, Bravo SB, Porteiro B, Cabaleiro A, Koning M, Senra A, Souto Y, Verheij J, Guallar D, Fidalgo M, Meijnikman AS, da Silva Lima N, Dieguez C, Gonzalez-Rellan MJ, Nogueiras R. Inhibition of hepatic p63 ameliorates steatohepatitis with fibrosis in mice. Mol Metab 2024; 85:101962. [PMID: 38815625 PMCID: PMC11180345 DOI: 10.1016/j.molmet.2024.101962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/12/2024] [Accepted: 05/22/2024] [Indexed: 06/01/2024] Open
Abstract
OBJECTIVE p63 is a transcription factor involved in multiple biological functions. In the liver, the TAp63 isoform induces lipid accumulation in hepatocytes. However, the role of liver TAp63 in the progression of metabolic dysfunction-associated steatohepatitis (MASH) with fibrosis is unknown. METHODS We evaluated the hepatic p63 levels in different mouse models of steatohepatitis with fibrosis induced by diet. Next, we used virogenetic approaches to manipulate the expression of TAp63 in adult mice under diet-induced steatohepatitis with fibrosis and characterized the disease condition. Finally, we performed proteomics analysis in mice with overexpression and knockdown of hepatic TAp63. RESULTS Levels of TAp63, but not of ΔN isoform, are increased in the liver of mice with diet-induced steatohepatitis with fibrosis. Both preventive and interventional strategies for the knockdown of hepatic TAp63 significantly ameliorated diet-induced steatohepatitis with fibrosis in mice fed a methionine- and choline-deficient diet (MCDD) and choline deficient and high fat diet (CDHFD). The overexpression of hepatic TAp63 in mice aggravated the liver condition in mice fed a CDHFD. Proteomic analysis in the liver of these mice revealed alteration in multiple proteins and pathways, such as oxidative phosphorylation, antioxidant activity, peroxisome function and LDL clearance. CONCLUSIONS These results indicate that liver TAp63 plays a critical role in the progression of diet-induced steatohepatitis with fibrosis, and its inhibition ameliorates the disease.
Collapse
Affiliation(s)
- Marcos F Fondevila
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain; CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, 15782, Spain; Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA.
| | - Eva Novoa
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain; CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, 15782, Spain
| | - Uxia Fernandez
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain; CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, 15782, Spain
| | - Valentina Dorta
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Tamara Parracho
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Henriette Kreimeyer
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Maria Garcia-Vence
- Proteomic Unit, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, 15705, Spain
| | - Maria P Chantada-Vazquez
- Proteomic Unit, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, 15705, Spain
| | - Susana B Bravo
- Proteomic Unit, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, 15705, Spain
| | - Begoña Porteiro
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Alba Cabaleiro
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Mijra Koning
- Department of Pathology, Amsterdam University Medical Centers, Location AMC, Amsterdam, the Netherlands
| | - Ana Senra
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Yara Souto
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Joanne Verheij
- Department of Pathology, Amsterdam University Medical Centers, Location AMC, Amsterdam, the Netherlands
| | - Diana Guallar
- Department of Biochemistry and Molecular Biology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Miguel Fidalgo
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Abraham S Meijnikman
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Natalia da Silva Lima
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Carlos Dieguez
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Maria J Gonzalez-Rellan
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain; CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, 15782, Spain; Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON, M5T 3H7, Canada
| | - Ruben Nogueiras
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain; CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, 15782, Spain; Galicia Agency of Innovation (GAIN), Xunta de Galicia, Santiago de Compostela, 15702, Spain.
| |
Collapse
|
3
|
Fondevila MF, Novoa E, Gonzalez-Rellan MJ, Fernandez U, Heras V, Porteiro B, Parracho T, Dorta V, Riobello C, da Silva Lima N, Seoane S, Garcia-Vence M, Chantada-Vazquez MP, Bravo SB, Senra A, Leiva M, Marcos M, Sabio G, Perez-Fernandez R, Dieguez C, Prevot V, Schwaninger M, Woodhoo A, Martinez-Chantar ML, Schwabe R, Cubero FJ, Varela-Rey M, Crespo J, Iruzubieta P, Nogueiras R. p63 controls metabolic activation of hepatic stellate cells and fibrosis via an HER2-ACC1 pathway. Cell Rep Med 2024; 5:101401. [PMID: 38340725 PMCID: PMC10897550 DOI: 10.1016/j.xcrm.2024.101401] [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: 02/09/2023] [Revised: 06/19/2023] [Accepted: 01/09/2024] [Indexed: 02/12/2024]
Abstract
The p63 protein has pleiotropic functions and, in the liver, participates in the progression of nonalcoholic fatty liver disease (NAFLD). However, its functions in hepatic stellate cells (HSCs) have not yet been explored. TAp63 is induced in HSCs from animal models and patients with liver fibrosis and its levels positively correlate with NAFLD activity score and fibrosis stage. In mice, genetic depletion of TAp63 in HSCs reduces the diet-induced liver fibrosis. In vitro silencing of p63 blunts TGF-β1-induced HSCs activation by reducing mitochondrial respiration and glycolysis, as well as decreasing acetyl CoA carboxylase 1 (ACC1). Ectopic expression of TAp63 induces the activation of HSCs and increases the expression and activity of ACC1 by promoting the transcriptional activity of HER2. Genetic inhibition of both HER2 and ACC1 blunt TAp63-induced activation of HSCs. Thus, TAp63 induces HSC activation by stimulating the HER2-ACC1 axis and participates in the development of liver fibrosis.
Collapse
Affiliation(s)
- Marcos F Fondevila
- Department of Physiology, CIMUS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), 15782 Santiago de Compostela, Spain.
| | - Eva Novoa
- Department of Physiology, CIMUS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), 15782 Santiago de Compostela, Spain
| | - Maria J Gonzalez-Rellan
- Department of Physiology, CIMUS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Uxia Fernandez
- Department of Physiology, CIMUS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), 15782 Santiago de Compostela, Spain
| | - Violeta Heras
- Department of Physiology, CIMUS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Begoña Porteiro
- Department of Physiology, CIMUS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Tamara Parracho
- Department of Physiology, CIMUS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Valentina Dorta
- Department of Physiology, CIMUS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Cristina Riobello
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Natalia da Silva Lima
- Department of Physiology, CIMUS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Samuel Seoane
- Department of Physiology, CIMUS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Maria Garcia-Vence
- Proteomic Unit, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), 15705 Santiago de Compostela, Spain
| | - Maria P Chantada-Vazquez
- Proteomic Unit, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), 15705 Santiago de Compostela, Spain
| | - Susana B Bravo
- Proteomic Unit, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), 15705 Santiago de Compostela, Spain
| | - Ana Senra
- Department of Physiology, CIMUS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Magdalena Leiva
- Department of Immunology, Ophthalmology, & ENT, Complutense University School of Medicine, 28040 Madrid, Spain; Health Research Institute Gregorio Marañón (IiSGM), 28007 Madrid, Spain; CIBER Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Miguel Marcos
- University of Salamanca, Department of Internal Medicine, University Hospital of Salamanca-IBSAL, 37008 Salamanca, Spain
| | - Guadalupe Sabio
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | - Roman Perez-Fernandez
- Department of Physiology, CIMUS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Carlos Dieguez
- Department of Physiology, CIMUS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Vincent Prevot
- University Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, European Genomic Institute for Diabetes (EGID), 59000 Lille, France
| | - Markus Schwaninger
- University of Lübeck, Institute for Experimental and Clinical Pharmacology and Toxicology, 23562 Lübeck, Germany
| | - Ashwin Woodhoo
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Maria L Martinez-Chantar
- Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Bizkaia, Spain
| | - Robert Schwabe
- Department of Medicine, Columbia University, New York, NY 10027, USA
| | - Francisco J Cubero
- Department of Immunology, Ophthalmology, & ENT, Complutense University School of Medicine, 28040 Madrid, Spain; Health Research Institute Gregorio Marañón (IiSGM), 28007 Madrid, Spain; CIBER Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Marta Varela-Rey
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Javier Crespo
- Gastroenterology and Hepatology Department, Marqués de Valdecilla University Hospital, Clinical and Translational Digestive Research Group, IDIVAL, 39008 Santander, Spain
| | - Paula Iruzubieta
- Gastroenterology and Hepatology Department, Marqués de Valdecilla University Hospital, Clinical and Translational Digestive Research Group, IDIVAL, 39008 Santander, Spain
| | - Ruben Nogueiras
- Department of Physiology, CIMUS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), 15782 Santiago de Compostela, Spain; Galicia Agency of Innovation (GAIN), Xunta de Galicia, 15702 Santiago de Compostela, Spain.
| |
Collapse
|
4
|
Schwartz B, Levi H, Menon G, Maria R, Upcher A, Kotlovski Y, Oss-Ronen L, Cohen I. ZNF750 Regulates Skin Barrier Function by Driving Cornified Envelope and Lipid Processing Pathways. J Invest Dermatol 2024; 144:296-306.e3. [PMID: 37660780 DOI: 10.1016/j.jid.2023.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/02/2023] [Accepted: 08/16/2023] [Indexed: 09/05/2023]
Abstract
The epidermis is a constantly renewing stratified epithelial tissue that provides essential protective barrier functions. The major barrier is located at the outermost layers of the epidermis, formed by terminally differentiated keratinocytes reinforced by proteins of their cornified envelope and sequestered intercellular lipids. Disruptions to epidermal differentiation characterize various skin disorders. ZNF750 is an epithelial transcription factor essential for in vitro keratinocyte differentiation, whose truncating mutation in humans causes autosomal dominant psoriasis-like skin disease. In this study, we utilized an epidermal-specific Znf750 conditional knockout mouse model to uncover the role ZNF750 plays in epidermal development. We show that deletion of Znf750 in the developing skin does not block epidermal differentiation completely, suggesting in vivo compensatory feedback mechanisms, although it does result in impaired barrier function and perinatal lethality. Molecular dissection revealed ultrastructural defects in the differentiated layers of the epidermis, accompanied by alterations in the expression of ZNF750-dependent genes encoding key cornified envelope precursor proteins and lipid-processing enzymes, including gene subsets known to be mutated in human skin diseases involving impaired barrier function. Together, our findings provide molecular insights into the pathogenesis of human skin disease by linking ZNF750 to a subset of epidermal differentiation genes involved in barrier formation pathways.
Collapse
Affiliation(s)
- Bar Schwartz
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Hilla Levi
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | | | - Raquel Maria
- Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Alexander Upcher
- Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Yulia Kotlovski
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Liat Oss-Ronen
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Idan Cohen
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.
| |
Collapse
|
5
|
Eyermann CE, Chen X, Somuncu OS, Li J, Joukov AN, Chen J, Alexandrova EM. ΔNp63 Regulates Homeostasis, Stemness, and Suppression of Inflammation in the Adult Epidermis. J Invest Dermatol 2024; 144:73-83.e10. [PMID: 37543242 DOI: 10.1016/j.jid.2023.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 06/22/2023] [Accepted: 07/10/2023] [Indexed: 08/07/2023]
Abstract
The p63 transcription factor is critical for epidermis formation in embryonic development, but its role in the adult epidermis is poorly understood. In this study, we show that acute genetic ablation of ΔNp63, the main p63 isoform, in adult epidermis disrupts keratinocyte proliferation and self-maintenance and, unexpectedly, triggers an inflammatory psoriasis-like condition. Mechanistically, single-cell RNA sequencing revealed the downregulation of cell cycle genes, upregulation of differentiation markers, and induction of several proinflammatory pathways in ΔNp63-ablated keratinocytes. Intriguingly, ΔNp63-ablated cells disappear by 3 weeks after ablation, at the expense of the remaining nonablated cells. This is not associated with active cell death and is likely due to reduced self-maintenance and enhanced differentiation. Indeed, in vivo wound healing, a physiological readout of the epidermal stem cell function, is severely impaired upon ΔNp63 ablation. We found that the Wnt signaling pathway (Wnt10A, Fzd6, Fzd10) and the activator protein 1 (JunB, Fos, FosB) factors are the likely ΔNp63 effectors responsible for keratinocyte proliferation/stemness and suppression of differentiation, respectively, whereas IL-1a, IL-18, IL-24, and IL-36γ are the likely negative effectors responsible for suppression of inflammation. These data establish ΔNp63 as a critical node that coordinates epidermal homeostasis, stemness, and suppression of inflammation, upstream of known regulatory pathways.
Collapse
Affiliation(s)
- Christopher E Eyermann
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA; Stony Brook Cancer Center, Stony Brook, New York, USA
| | - Xi Chen
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA; Stony Brook Cancer Center, Stony Brook, New York, USA
| | - Ozge S Somuncu
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA; Stony Brook Cancer Center, Stony Brook, New York, USA
| | - Jinyu Li
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA; Stony Brook Cancer Center, Stony Brook, New York, USA
| | | | - Jiang Chen
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA; Stony Brook Cancer Center, Stony Brook, New York, USA
| | - Evguenia M Alexandrova
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA; Stony Brook Cancer Center, Stony Brook, New York, USA.
| |
Collapse
|
6
|
Pham LNG, Niimi T, Suzuki S, Nguyen MD, Nguyen LCH, Nguyen TD, Hoang KA, Nguyen DM, Sakuma C, Hayakawa T, Hiyori M, Natsume N, Furukawa H, Imura H, Akashi J, Ohta T, Natsume N. Association between IRF6, TP63, GREM1 Gene Polymorphisms and Non-Syndromic Orofacial Cleft Phenotypes in Vietnamese Population: A Case-Control and Family-Based Study. Genes (Basel) 2023; 14:1995. [PMID: 38002937 PMCID: PMC10671090 DOI: 10.3390/genes14111995] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 10/18/2023] [Accepted: 10/24/2023] [Indexed: 11/26/2023] Open
Abstract
This study aims to identify potential variants in the TP63-IRF6 pathway and GREM1 for the etiology of non-syndromic orofacial cleft (NSOFC) among the Vietnamese population. By collecting 527 case-parent trios and 527 control samples, we conducted a stratified analysis based on different NSOFC phenotypes, using allelic, dominant, recessive and over-dominant models for case-control analyses, and family-based association tests for case-parent trios. Haplotype and linkage disequilibrium analyses were also conducted. IRF6 rs2235375 showed a significant association with an increased risk for non-syndromic cleft lip and palate (NSCLP) and cleft lip with or without cleft palate (NSCL/P) in the G allele, with pallele values of 0.0018 and 0.0003, respectively. Due to the recessive model (p = 0.0011) for the NSCL/P group, the reduced frequency of the GG genotype of rs2235375 was associated with a protective effect against NSCL/P. Additionally, offspring who inherited the G allele at rs2235375 had a 1.34-fold increased risk of NSCL/P compared to the C allele holders. IRF6 rs846810 and a G-G haplotype at rs2235375-rs846810 of IRF6 impacted NSCL/P, with p-values of 0.0015 and 0.0003, respectively. In conclusion, our study provided additional evidence for the association of IRF6 rs2235375 with NSCLP and NSCL/P. We also identified IRF6 rs846810 as a novel marker associated with NSCL/P, and haplotypes G-G and C-A at rs2235375-rs846810 of IRF6 associated with NSOFC.
Collapse
Affiliation(s)
- Loc Nguyen Gia Pham
- Division of Research and Treatment for Oral and Maxillofacial Congenital Anomalies, Aichi Gakuin University, 2–11 Suemori-dori, Chikusa-ku, Nagoya 464-8651, Japan; (L.N.G.P.); (T.N.); (S.S.); (D.M.N.); (C.S.); (N.N.); (H.I.)
- Odonto-Maxillo Facial Hospital of Ho Chi Minh City, 263-265 Tran Hung Dao Street, District 1, Ho Chi Minh City 71000, Vietnam; (M.D.N.); (L.C.H.N.); (T.D.N.); (K.A.H.)
| | - Teruyuki Niimi
- Division of Research and Treatment for Oral and Maxillofacial Congenital Anomalies, Aichi Gakuin University, 2–11 Suemori-dori, Chikusa-ku, Nagoya 464-8651, Japan; (L.N.G.P.); (T.N.); (S.S.); (D.M.N.); (C.S.); (N.N.); (H.I.)
- Cleft Lip and Palate Center, Aichi Gakuin Dental Hospital, 2-11 Suemori-dori, Chikusa-ku, Nagoya 464-8651, Japan;
- Division of Speech, Hearing, and Language, Aichi Gakuin Dental Hospital, 2-11 Suemori-dori, Chikusa-ku, Nagoya 464-8651, Japan; (T.H.); (M.H.)
| | - Satoshi Suzuki
- Division of Research and Treatment for Oral and Maxillofacial Congenital Anomalies, Aichi Gakuin University, 2–11 Suemori-dori, Chikusa-ku, Nagoya 464-8651, Japan; (L.N.G.P.); (T.N.); (S.S.); (D.M.N.); (C.S.); (N.N.); (H.I.)
| | - Minh Duc Nguyen
- Odonto-Maxillo Facial Hospital of Ho Chi Minh City, 263-265 Tran Hung Dao Street, District 1, Ho Chi Minh City 71000, Vietnam; (M.D.N.); (L.C.H.N.); (T.D.N.); (K.A.H.)
| | - Linh Cao Hoai Nguyen
- Odonto-Maxillo Facial Hospital of Ho Chi Minh City, 263-265 Tran Hung Dao Street, District 1, Ho Chi Minh City 71000, Vietnam; (M.D.N.); (L.C.H.N.); (T.D.N.); (K.A.H.)
| | - Tuan Duc Nguyen
- Odonto-Maxillo Facial Hospital of Ho Chi Minh City, 263-265 Tran Hung Dao Street, District 1, Ho Chi Minh City 71000, Vietnam; (M.D.N.); (L.C.H.N.); (T.D.N.); (K.A.H.)
| | - Kien Ai Hoang
- Odonto-Maxillo Facial Hospital of Ho Chi Minh City, 263-265 Tran Hung Dao Street, District 1, Ho Chi Minh City 71000, Vietnam; (M.D.N.); (L.C.H.N.); (T.D.N.); (K.A.H.)
| | - Duc Minh Nguyen
- Division of Research and Treatment for Oral and Maxillofacial Congenital Anomalies, Aichi Gakuin University, 2–11 Suemori-dori, Chikusa-ku, Nagoya 464-8651, Japan; (L.N.G.P.); (T.N.); (S.S.); (D.M.N.); (C.S.); (N.N.); (H.I.)
- School of Odonto-Stomatology, Hanoi Medical University, Hanoi 10000, Vietnam
| | - Chisato Sakuma
- Division of Research and Treatment for Oral and Maxillofacial Congenital Anomalies, Aichi Gakuin University, 2–11 Suemori-dori, Chikusa-ku, Nagoya 464-8651, Japan; (L.N.G.P.); (T.N.); (S.S.); (D.M.N.); (C.S.); (N.N.); (H.I.)
- Cleft Lip and Palate Center, Aichi Gakuin Dental Hospital, 2-11 Suemori-dori, Chikusa-ku, Nagoya 464-8651, Japan;
- Division of Speech, Hearing, and Language, Aichi Gakuin Dental Hospital, 2-11 Suemori-dori, Chikusa-ku, Nagoya 464-8651, Japan; (T.H.); (M.H.)
| | - Toko Hayakawa
- Division of Speech, Hearing, and Language, Aichi Gakuin Dental Hospital, 2-11 Suemori-dori, Chikusa-ku, Nagoya 464-8651, Japan; (T.H.); (M.H.)
| | - Makino Hiyori
- Division of Speech, Hearing, and Language, Aichi Gakuin Dental Hospital, 2-11 Suemori-dori, Chikusa-ku, Nagoya 464-8651, Japan; (T.H.); (M.H.)
| | - Nagana Natsume
- Division of Research and Treatment for Oral and Maxillofacial Congenital Anomalies, Aichi Gakuin University, 2–11 Suemori-dori, Chikusa-ku, Nagoya 464-8651, Japan; (L.N.G.P.); (T.N.); (S.S.); (D.M.N.); (C.S.); (N.N.); (H.I.)
- Cleft Lip and Palate Center, Aichi Gakuin Dental Hospital, 2-11 Suemori-dori, Chikusa-ku, Nagoya 464-8651, Japan;
- Division of Speech, Hearing, and Language, Aichi Gakuin Dental Hospital, 2-11 Suemori-dori, Chikusa-ku, Nagoya 464-8651, Japan; (T.H.); (M.H.)
| | - Hiroo Furukawa
- Cleft Lip and Palate Center, Aichi Gakuin Dental Hospital, 2-11 Suemori-dori, Chikusa-ku, Nagoya 464-8651, Japan;
- Division of Speech, Hearing, and Language, Aichi Gakuin Dental Hospital, 2-11 Suemori-dori, Chikusa-ku, Nagoya 464-8651, Japan; (T.H.); (M.H.)
| | - Hideto Imura
- Division of Research and Treatment for Oral and Maxillofacial Congenital Anomalies, Aichi Gakuin University, 2–11 Suemori-dori, Chikusa-ku, Nagoya 464-8651, Japan; (L.N.G.P.); (T.N.); (S.S.); (D.M.N.); (C.S.); (N.N.); (H.I.)
- Cleft Lip and Palate Center, Aichi Gakuin Dental Hospital, 2-11 Suemori-dori, Chikusa-ku, Nagoya 464-8651, Japan;
- Division of Speech, Hearing, and Language, Aichi Gakuin Dental Hospital, 2-11 Suemori-dori, Chikusa-ku, Nagoya 464-8651, Japan; (T.H.); (M.H.)
| | - Junko Akashi
- Division of Research and Treatment for Oral and Maxillofacial Congenital Anomalies, Aichi Gakuin University, 2–11 Suemori-dori, Chikusa-ku, Nagoya 464-8651, Japan; (L.N.G.P.); (T.N.); (S.S.); (D.M.N.); (C.S.); (N.N.); (H.I.)
| | - Tohru Ohta
- Advanced Research Promotion Center, Health Sciences University of Hokkaido, Ishikari-Tobetsu 061-0293, Japan;
| | - Nagato Natsume
- Division of Research and Treatment for Oral and Maxillofacial Congenital Anomalies, Aichi Gakuin University, 2–11 Suemori-dori, Chikusa-ku, Nagoya 464-8651, Japan; (L.N.G.P.); (T.N.); (S.S.); (D.M.N.); (C.S.); (N.N.); (H.I.)
- Cleft Lip and Palate Center, Aichi Gakuin Dental Hospital, 2-11 Suemori-dori, Chikusa-ku, Nagoya 464-8651, Japan;
- Division of Speech, Hearing, and Language, Aichi Gakuin Dental Hospital, 2-11 Suemori-dori, Chikusa-ku, Nagoya 464-8651, Japan; (T.H.); (M.H.)
| |
Collapse
|
7
|
Terrinoni A, Sala G, Bruno E, Pitolli C, Minieri M, Pieri M, Gambacurta A, Campione E, Belardi R, Bernardini S. Partial Loss of Function ABCA12 Mutations Generate Reduced Deposition of Glucosyl-Ceramide, Leading to Patchy Ichthyosis and Erythrodermia Resembling Erythrokeratodermia Variabilis et Progressiva (EKVP). Int J Mol Sci 2023; 24:13962. [PMID: 37762265 PMCID: PMC10530436 DOI: 10.3390/ijms241813962] [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: 07/18/2023] [Revised: 08/25/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
Ichthyoses are genetically determined cornification disorders of the epidermis characterized by the presence of different degrees of scaling, hyperkeratosis, and erythroderma often associated with palmoplantar keratoderma. Different classifications of these diseases have been proposed, often based upon the involved genes and/or the clinical presentation. The clinical features of these diseases present some overlap of phenotypes among distinct genetic entities, depending mainly on the penetrance of mutations. In this study, using a clinical, genetic, and molecular approach, we analyzed a family with two affected members who had clinical and histological features resembling erythrokeratodermia variabilis (EKV) or a type of erythrodermic hyperkeratosis with palmoplantar keratoderma. Despite of the clinical presentation, we demonstrated that the affected patients were genetically double heterozygous for two different mutations in the ABCA12 gene, known to be responsible for harlequin ichthyosis. To explain the mild phenotype of our patients, we performed a molecular characterization of the skin. In the upper layers of the epidermis, the results showed a patchy presence of the glucosyl-ceramides (GlcCer), which is the lipid transported by ABCA12, fundamental in contributing to skin impermeability. Indeed, the two mutations detected do not completely abolish ABCA12 activity, indicating that the mild phenotype is due to a partial loss of function of the enzyme, thus giving rise to an intermediate phenotype resembling EKVP, due to a partial depletion of GlcCer deposition.
Collapse
Affiliation(s)
- Alessandro Terrinoni
- Department of Experimental Medicine, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (G.S.); (M.M.); (M.P.); (A.G.); (R.B.); (S.B.)
| | - Gabriele Sala
- Department of Experimental Medicine, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (G.S.); (M.M.); (M.P.); (A.G.); (R.B.); (S.B.)
| | - Ernesto Bruno
- Department of Clinical Sciences and Translational Medicine University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy;
| | - Consuelo Pitolli
- Department of Neuroscience, Section of Human Anatomy, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy;
| | - Marilena Minieri
- Department of Experimental Medicine, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (G.S.); (M.M.); (M.P.); (A.G.); (R.B.); (S.B.)
| | - Massimo Pieri
- Department of Experimental Medicine, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (G.S.); (M.M.); (M.P.); (A.G.); (R.B.); (S.B.)
| | - Alessandra Gambacurta
- Department of Experimental Medicine, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (G.S.); (M.M.); (M.P.); (A.G.); (R.B.); (S.B.)
| | - Elena Campione
- Department of System Medicine, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy;
| | - Riccardo Belardi
- Department of Experimental Medicine, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (G.S.); (M.M.); (M.P.); (A.G.); (R.B.); (S.B.)
| | - Sergio Bernardini
- Department of Experimental Medicine, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (G.S.); (M.M.); (M.P.); (A.G.); (R.B.); (S.B.)
| |
Collapse
|
8
|
Innovative Therapeutic Approaches for the Treatment of the Ocular Morbidities in Patients with EEC Syndrome. Cells 2023; 12:cells12030495. [PMID: 36766837 PMCID: PMC9914602 DOI: 10.3390/cells12030495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/19/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Ectrodactyly-Ectodermal dysplasia-Clefting (EEC) syndrome is caused by heterozygous missense point mutations in the p63 gene, an important transcription factor during embryogenesis and for stem cell differentiation in stratified epithelia. Most of the cases are sporadic, related to de novo mutations arising during early-stage development. Familial cases show an autosomic dominant inheritance. The major cause of visual morbidity is limbal stem cell failure, which develops in the second to third decade of life. Patients often show ocular surface alterations, such as recurrent blepharitis and conjunctivitis, superficial microlesions of the cornea, and spontaneous corneal perforation and ulceration, leading to progressive corneal clouding and eventually visual loss. No definitive cures are currently available, and treatments to alleviate symptoms are only palliative. In this review, we will discuss the proposed therapeutic strategies that have been tested or are under development for the management of the ocular defects in patients affected by EEC syndrome: (i) gene therapy-based approaches by means of Allele-Specific (AS) siRNAs to correct the p63 mutations; (ii) cell therapy-based approaches to replenish the pool of limbal stem cells; and (iii) drug therapy to correct/bypass the genetic defect. However, as the number of patients with EEC syndrome is too limited, further studies are still necessary to prove the effectiveness (and safety) of these innovative therapeutic approaches to counteract the premature differentiation of limbal stem cells.
Collapse
|
9
|
Vlašić I, Horvat A, Tadijan A, Slade N. p53 Family in Resistance to Targeted Therapy of Melanoma. Int J Mol Sci 2022; 24:ijms24010065. [PMID: 36613518 PMCID: PMC9820688 DOI: 10.3390/ijms24010065] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Metastatic melanoma is one of the most aggressive tumors, with frequent mutations affecting components of the MAPK pathway, mainly protein kinase BRAF. Despite promising initial response to BRAF inhibitors, melanoma progresses due to development of resistance. In addition to frequent reactivation of MAPK or activation of PI3K/AKT signaling pathways, recently, the p53 pathway has been shown to contribute to acquired resistance to targeted MAPK inhibitor therapy. Canonical tumor suppressor p53 is inactivated in melanoma by diverse mechanisms. The TP53 gene and two other family members, TP63 and TP73, encode numerous protein isoforms that exhibit diverse functions during tumorigenesis. The p53 family isoforms can be produced by usage of alternative promoters and/or splicing on the C- and N-terminus. Various p53 family isoforms are expressed in melanoma cell lines and tumor samples, and several of them have already shown to have specific functions in melanoma, affecting proliferation, survival, metastatic potential, invasion, migration, and response to therapy. Of special interest are p53 family isoforms with increased expression and direct involvement in acquired resistance to MAPK inhibitors in melanoma cells, implying that modulating their expression or targeting their functional pathways could be a potential therapeutic strategy to overcome resistance to MAPK inhibitors in melanoma.
Collapse
|
10
|
Weiner AI, Zhao G, Zayas HM, Holcomb NP, Adams-Tzivelekidis S, Wong J, Gentile ME, Reddy D, Wei J, Palashikar G, Quansah KK, Vaughan AE. ΔNp63 drives dysplastic alveolar remodeling and restricts epithelial plasticity upon severe lung injury. Cell Rep 2022; 41:111805. [PMID: 36516758 PMCID: PMC9808897 DOI: 10.1016/j.celrep.2022.111805] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 10/13/2022] [Accepted: 11/18/2022] [Indexed: 12/15/2022] Open
Abstract
The lung exhibits a robust, multifaceted regenerative response to severe injuries such as influenza infection, during which quiescent lung-resident epithelial progenitors participate in two distinct reparative pathways: functionally beneficial regeneration via alveolar type 2 (AT2) cell proliferation and differentiation, and dysplastic tissue remodeling via intrapulmonary airway-resident basal p63+ progenitors. Here we show that the basal cell transcription factor ΔNp63 is required for intrapulmonary basal progenitors to participate in dysplastic alveolar remodeling following injury. We find that ΔNp63 restricts the plasticity of intrapulmonary basal progenitors by maintaining either active or repressive histone modifications at key differentiation gene loci. Following loss of ΔNp63, intrapulmonary basal progenitors are capable of either airway or alveolar differentiation depending on their surrounding environment both in vitro and in vivo. Uncovering these regulatory mechanisms of dysplastic repair and lung basal cell fate choice highlight potential therapeutic targets to promote functional alveolar regeneration following severe lung injuries.
Collapse
Affiliation(s)
- Aaron I Weiner
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gan Zhao
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hanna M Zayas
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nicolas P Holcomb
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Stephanie Adams-Tzivelekidis
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joanna Wong
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Maria E Gentile
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Dyuthi Reddy
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joey Wei
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gargi Palashikar
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kwaku K Quansah
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrew E Vaughan
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
11
|
Distinct interactors define the p63 transcriptional signature in epithelial development or cancer. Biochem J 2022; 479:1375-1392. [PMID: 35748701 PMCID: PMC9250260 DOI: 10.1042/bcj20210737] [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: 04/05/2022] [Revised: 06/01/2022] [Accepted: 06/06/2022] [Indexed: 11/24/2022]
Abstract
The TP63 is an indispensable transcription factor for development and homeostasis of epithelia and its derived glandular tissue. It is also involved in female germline cell quality control, muscle and thymus development. It is expressed as multiple isoforms transcribed by two independent promoters, in addition to alternative splicing occurring at the mRNA 3′-UTR. Expression of the TP63 gene, specifically the amino-deleted p63 isoform, ΔNp63, is required to regulate numerous biological activities, including lineage specification, self-renewal capacity of epithelial stem cells, proliferation/expansion of basal keratinocytes, differentiation of stratified epithelia. In cancer, ΔNp63 is implicated in squamous cancers pathogenesis of different origin including skin, head and neck and lung and in sustaining self-renewal of cancer stem cells. How this transcription factor can control such a diverse set of biological pathways is central to the understanding of the molecular mechanisms through which p63 acquires oncogenic activity, profoundly changing its down-stream transcriptional signature. Here, we highlight how different proteins interacting with p63 allow it to regulate the transcription of several central genes. The interacting proteins include transcription factors/regulators, epigenetic modifiers, and post-transcriptional modifiers. Moreover, as p63 depends on its interactome, we discuss the hypothesis to target the protein interactors to directly affect p63 oncogenic activities and p63-related diseases.
Collapse
|
12
|
Novelli F, Ganini C, Melino G, Nucci C, Han Y, Shi Y, Wang Y, Candi E. p63 in corneal and epidermal differentiation. Biochem Biophys Res Commun 2022; 610:15-22. [DOI: 10.1016/j.bbrc.2022.04.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 04/06/2022] [Indexed: 11/02/2022]
|
13
|
Osterburg C, Dötsch V. Structural diversity of p63 and p73 isoforms. Cell Death Differ 2022; 29:921-937. [PMID: 35314772 PMCID: PMC9091270 DOI: 10.1038/s41418-022-00975-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 01/25/2023] Open
Abstract
Abstract
The p53 protein family is the most studied protein family of all. Sequence analysis and structure determination have revealed a high similarity of crucial domains between p53, p63 and p73. Functional studies, however, have shown a wide variety of different tasks in tumor suppression, quality control and development. Here we review the structure and organization of the individual domains of p63 and p73, the interaction of these domains in the context of full-length proteins and discuss the evolutionary origin of this protein family.
Facts
Distinct physiological roles/functions are performed by specific isoforms.
The non-divided transactivation domain of p63 has a constitutively high activity while the transactivation domains of p53/p73 are divided into two subdomains that are regulated by phosphorylation.
Mdm2 binds to all three family members but ubiquitinates only p53.
TAp63α forms an autoinhibited dimeric state while all other vertebrate p53 family isoforms are constitutively tetrameric.
The oligomerization domain of p63 and p73 contain an additional helix that is necessary for stabilizing the tetrameric states. During evolution this helix got lost independently in different phylogenetic branches, while the DNA binding domain became destabilized and the transactivation domain split into two subdomains.
Open questions
Is the autoinhibitory mechanism of mammalian TAp63α conserved in p53 proteins of invertebrates that have the same function of genomic quality control in germ cells?
What is the physiological function of the p63/p73 SAM domains?
Do the short isoforms of p63 and p73 have physiological functions?
What are the roles of the N-terminal elongated TAp63 isoforms, TA* and GTA?
Collapse
|
14
|
Laubach K, Zhang J, Chen X. The p53 Family: A Role in Lipid and Iron Metabolism. Front Cell Dev Biol 2021; 9:715974. [PMID: 34395447 PMCID: PMC8358664 DOI: 10.3389/fcell.2021.715974] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 07/08/2021] [Indexed: 12/11/2022] Open
Abstract
The p53 family of tumor suppressors, which includes p53, p63, and p73, has a critical role in many biological processes, such as cell cycle arrest, apoptosis, and differentiation. In addition to tumor suppression, the p53 family proteins also participate in development, multiciliogenesis, and fertility, indicating these proteins have diverse roles. In this review, we strive to cover the relevant studies that demonstrate the roles of p53, p63, and p73 in lipid and iron metabolism.
Collapse
Affiliation(s)
| | | | - Xinbin Chen
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
15
|
Post Zygotic, Somatic, Deletion in KERATIN 1 V1 Domain Generates Structural Alteration of the K1/K10 Dimer, Producing a Monolateral Palmar Epidermolytic Nevus. Int J Mol Sci 2021; 22:ijms22136901. [PMID: 34199056 PMCID: PMC8269197 DOI: 10.3390/ijms22136901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/23/2021] [Accepted: 06/23/2021] [Indexed: 11/29/2022] Open
Abstract
Palmoplantar keratodermas (PPKs) are characterized by thickness of stratum corneum and epidermal hyperkeratosis localized in palms and soles. PPKs can be epidermolytic (EPPK) or non epidermolytic (NEPPK). Specific mutations of keratin 16 (K16) and keratin 1 (K1) have been associated to EPPK, and NEPPK. Cases of mosaicism in PPKs due to somatic keratin mutations have also been described in scientific literature. We evaluated a patient presenting hyperkeratosis localized monolaterally in the right palmar area, characterized by linear yellowish hyperkeratotic lesions following the Blaschko lines. No other relatives of the patient showed any dermatological disease. Light and confocal histological analysis confirmed the presence of epidermolityic hyperkeratosis. Genetic analysis performed demonstrates the heterozygous deletion NM_006121.4:r.274_472del for a total of 198 nucleotides, in KRT1 cDNA obtained by a palmar lesional skin biopsy, corresponding to the protein mutation NP_006112.3:p.Gly71_Gly137del. DNA extracted from peripheral blood lymphocytes did not display the presence of the mutation. These results suggest a somatic mutation causing an alteration in K1 N-terminal variable domain (V1). The deleted sequence involves the ISIS subdomain, containing a lysine residue already described as fundamental for epidermal transglutaminases in the crosslinking of IF cytoskeleton. Moreover, a computational analysis of the wild-type and V1-mutated K1/K10 keratin dimers, suggests an unusual interaction between these keratin filaments. The mutation taster in silico analysis also returned a high probability for a deleterious mutation. These data demonstrate once again the importance of the head domain (V1) of K1 in the formation of a functional keratinocyte cytoskeleton. Moreover, this is a further demonstration of the presence of somatic mutations arising in later stages of the embryogenesis, generating a mosaic phenotype.
Collapse
|
16
|
Yu X, Singh PK, Tabrejee S, Sinha S, Buck MJ. ΔNp63 is a pioneer factor that binds inaccessible chromatin and elicits chromatin remodeling. Epigenetics Chromatin 2021; 14:20. [PMID: 33865440 PMCID: PMC8053304 DOI: 10.1186/s13072-021-00394-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 04/02/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND ΔNp63 is a master transcriptional regulator playing critical roles in epidermal development and other cellular processes. Recent studies suggest that ΔNp63 functions as a pioneer factor that can target its binding sites within inaccessible chromatin and induce chromatin remodeling. METHODS In order to examine if ΔNp63 can bind to inaccessible chromatin and to determine if specific histone modifications are required for binding, we induced ΔNp63 expression in two p63-naïve cell lines. ΔNp63 binding was then examined by ChIP-seq and the chromatin at ΔNp63 targets sites was examined before and after binding. Further analysis with competitive nucleosome binding assays was used to determine how ΔNp63 directly interacts with nucleosomes. RESULTS Our results show that before ΔNp63 binding, targeted sites lack histone modifications, indicating ΔNp63's capability to bind at unmodified chromatin. Moreover, the majority of the sites that are bound by ectopic ΔNp63 expression exist in an inaccessible state. Once bound, ΔNp63 induces acetylation of the histone and the repositioning of nucleosomes at its binding sites. Further analysis with competitive nucleosome binding assays reveal that ΔNp63 can bind directly to nucleosome edges with significant binding inhibition occurring within 50 bp of the nucleosome dyad. CONCLUSION Overall, our results demonstrate that ΔNp63 is a pioneer factor that binds nucleosome edges at inaccessible and unmodified chromatin sites and induces histone acetylation and nucleosome repositioning.
Collapse
Affiliation(s)
- Xinyang Yu
- Department of Biochemistry, State University of New York at Buffalo, Buffalo, NY, 14203, USA.,Zhuhai Interventional Medical Center, Zhuhai Precision Medical Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, Guangdong, China
| | - Prashant K Singh
- Department of Biochemistry, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - Shamira Tabrejee
- Department of Biochemistry, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - Satrajit Sinha
- Department of Biochemistry, State University of New York at Buffalo, Buffalo, NY, 14203, USA.
| | - Michael J Buck
- Department of Biochemistry, State University of New York at Buffalo, Buffalo, NY, 14203, USA. .,Department of Biomedical Informatics, Jacobs School of Medicine & Biomedical Sciences, Buffalo, USA.
| |
Collapse
|
17
|
Xu J, Li F, Gao Y, Guo R, Ding L, Fu M, Yi Y, Chen H, Xiao ZXJ, Niu M. E47 upregulates ΔNp63α to promote growth of squamous cell carcinoma. Cell Death Dis 2021; 12:381. [PMID: 33833226 PMCID: PMC8032790 DOI: 10.1038/s41419-021-03662-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/17/2021] [Accepted: 03/23/2021] [Indexed: 12/28/2022]
Abstract
Targeted therapy has greatly improved both survival and prognosis of cancer patients. However, while therapeutic treatment of adenocarcinoma has been advanced greatly, progress in treatment of squamous cell carcinoma (SCC) has been slow and ineffective. Therefore, it is of great importance to decipher mechanisms and identify new drug targets involved in squamous cell carcinoma development. In this study, we demonstrate that E47 plays the distinctive and opposite roles on cell proliferation in adenocarcinoma and squamous cell carcinoma. While E47 suppresses cell proliferation in adenocarcinoma cells, it functions as a oncoprotein to promote cell proliferation and tumor growth of squamous cell carcinoma. Mechanistically, we show that E47 can directly bind to the promoter and transactivate ΔNp63 gene expression in squamous cell carcinoma cells, resulting in upregulation of cyclins D1/E1 and downregulation of p21, and thereby promoting cell proliferation and tumor growth. We further show that expression of E2A (E12/E47) is positively correlated with p63 and that high expression of E2A is associated with poor outcomes in clinical samples of squamous cell carcinoma. These results highlight that the E47-ΔNp63α axis may be potential therapeutic targets for treatment of squamous cell carcinoma.
Collapse
Affiliation(s)
- Jing Xu
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Fengtian Li
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Ya Gao
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Rongtian Guo
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Liangping Ding
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Mengyuan Fu
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yong Yi
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Hu Chen
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Zhi-Xiong Jim Xiao
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Mengmeng Niu
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China.
| |
Collapse
|
18
|
Kubo T, Sato S, Hida T, Minowa T, Hirohashi Y, Tsukahara T, Kanaseki T, Murata K, Uhara H, Torigoe T. IL-13 modulates ∆Np63 levels causing altered expression of barrier- and inflammation-related molecules in human keratinocytes: A possible explanation for chronicity of atopic dermatitis. IMMUNITY INFLAMMATION AND DISEASE 2021; 9:734-745. [PMID: 33792188 PMCID: PMC8342210 DOI: 10.1002/iid3.427] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/15/2021] [Accepted: 03/17/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND Barrier disruption and an excessive immune response in keratinocytes are now considered to have important roles in the pathophysiology of atopic dermatitis (AD). Furthermore, disturbed keratinocyte differentiation is considered to underlie AD. ΔNp63, a p53-like transcription factor, is a major regulator of keratinocyte differentiation. However, the functional significance of ΔNp63 in AD has not been clarified. OBJECTIVE In this study, we aimed to investigate the influence of the type 2 inflammatory environment on ΔNp63 expression and AD-associated molecules regulated by ΔNp63 in keratinocytes. METHODS The immunohistochemical expression profiles of ΔNp63 and AD-related molecules were evaluated in human skin tissue. The function of ΔNp63 in the regulation of AD-related molecules and the influence of the type 2 inflammatory environment on ΔNp63 expression were investigated using human primary keratinocytes. Expression of ΔNp63 was manipulated using the RNA interfering method. RESULTS In healthy skin tissue, we observed an inverse expression pattern between ∆Np63 and some barrier-related proteins including filaggrin, caspase-14, claudin-1, and claudin-4. ΔNp63 regulated expression of these genes and proteins. In addition, production of IL-1β and IL-33, pro-inflammatory cytokines, was modulated by ΔNp63. Furthermore, prolonged IL-13 exposure increased the thickness of the three-dimensional culture of keratinocytes. IL-13 interfered with ΔNp63 downregulation during calcium-induced keratinocyte differentiation. IL-13 modulated some barrier-related and inflammation-related molecules, which were regulated by ΔNp63. CONCLUSIONS We have shown that ΔNp63 modulated AD-related barrier and inflammatory molecules. In addition, ΔNp63 expression was affected by IL-4/IL-13. IL-13-ΔNp63 axis would integrate two major factors of AD pathogenesis: dysregulated barrier and inflammation.
Collapse
Affiliation(s)
- Terufumi Kubo
- Department of Pathology, School of Medicine, Sapporo Medical University, Sapporo, Japan
| | - Sayuri Sato
- Department of Dermatology, School of Medicine, Sapporo Medical University, Sapporo, Japan
| | - Tokimasa Hida
- Department of Dermatology, School of Medicine, Sapporo Medical University, Sapporo, Japan
| | - Tomoyuki Minowa
- Department of Pathology, School of Medicine, Sapporo Medical University, Sapporo, Japan.,Department of Dermatology, School of Medicine, Sapporo Medical University, Sapporo, Japan
| | - Yoshihiko Hirohashi
- Department of Pathology, School of Medicine, Sapporo Medical University, Sapporo, Japan
| | - Tomohide Tsukahara
- Department of Pathology, School of Medicine, Sapporo Medical University, Sapporo, Japan
| | - Takayuki Kanaseki
- Department of Pathology, School of Medicine, Sapporo Medical University, Sapporo, Japan
| | - Kenji Murata
- Department of Pathology, School of Medicine, Sapporo Medical University, Sapporo, Japan
| | - Hisashi Uhara
- Department of Dermatology, School of Medicine, Sapporo Medical University, Sapporo, Japan
| | - Toshihiko Torigoe
- Department of Pathology, School of Medicine, Sapporo Medical University, Sapporo, Japan
| |
Collapse
|
19
|
Dong Y, Long J, Luo X, Xie G, Xiao ZJ, Tong Y. Targeting of ΔNp63α by miR-522 promotes the migration of breast epithelial cells. FEBS Open Bio 2021; 11:468-481. [PMID: 33369228 PMCID: PMC7876488 DOI: 10.1002/2211-5463.13072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 12/10/2020] [Accepted: 12/21/2020] [Indexed: 01/09/2023] Open
Abstract
The TP63 gene, which encodes the p63 protein, is involved in multiple biological processes, including embryonic development and tumorigenesis. ΔNp63α, the predominant isoform of p63 in epithelial cells, acts as an oncogene in early-stage tumors, but paradoxically acts as a potent antimetastatic factor in advanced cancers. Here, we report that ΔNp63α is a direct target of hsa-miR-522 (miR-522). Induced expression of miR-522 reduced the levels of ΔNp63α, predisposing breast epithelial cells to a loss of epithelial and acquisition of mesenchymal morphology, resulting in accelerated collective and single-cell migration. Restoration of ΔNp63α repressed miR-522-induced migration. Interestingly, overexpression of miR-522 did not affect breast epithelial cell proliferation, suggesting that miR-522 acts specifically through ΔNp63α in this context. Furthermore, expression of miR-522-3p and p63 was negatively correlated in human cancer samples. Thus, miR-522 might be a causative factor for breast tumorigenesis and cancer metastasis. In summary, our results reveal a novel miR-522/p63 axis in cell migration and thus suggest a potential strategy for therapeutic treatment of cancer metastasis.
Collapse
Affiliation(s)
- Yuanyuan Dong
- Center of Growth, Metabolism and AgingKey Laboratory of Bio‐Resource and Eco‐Environment of Ministry of EducationCollege of Life SciencesSichuan UniversityChengduChina
| | - Juan Long
- Center of Growth, Metabolism and AgingKey Laboratory of Bio‐Resource and Eco‐Environment of Ministry of EducationCollege of Life SciencesSichuan UniversityChengduChina
| | - Xingyong Luo
- Center of Growth, Metabolism and AgingKey Laboratory of Bio‐Resource and Eco‐Environment of Ministry of EducationCollege of Life SciencesSichuan UniversityChengduChina
| | - Gang Xie
- Sichuan Integrative Medicine HospitalChengduChina
| | - Zhi‐Xiong Jim Xiao
- Center of Growth, Metabolism and AgingKey Laboratory of Bio‐Resource and Eco‐Environment of Ministry of EducationCollege of Life SciencesSichuan UniversityChengduChina
| | - Ying Tong
- Center of Growth, Metabolism and AgingKey Laboratory of Bio‐Resource and Eco‐Environment of Ministry of EducationCollege of Life SciencesSichuan UniversityChengduChina
| |
Collapse
|
20
|
Isoform-Specific Roles of Mutant p63 in Human Diseases. Cancers (Basel) 2021; 13:cancers13030536. [PMID: 33572532 PMCID: PMC7866788 DOI: 10.3390/cancers13030536] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/25/2021] [Accepted: 01/27/2021] [Indexed: 12/26/2022] Open
Abstract
Simple Summary The protein p63 belongs to the family of the p53 tumor suppressor. Mouse models have, however, shown that it is not a classical tumor suppressor but instead involved in developmental processes. Mutations in the p63 gene cause several developmental defects in human patients characterized by limb deformation, cleft lip/palate, and ectodermal dysplasia due to p63’s role as a master regulator of epidermal development. In addition, p63 plays a key role as a quality control factor in oocytes and p63 mutations can result either in compromised genetic quality control or premature cell death of all oocytes. Abstract The p63 gene encodes a master regulator of epidermal commitment, development, and differentiation. Heterozygous mutations in the DNA binding domain cause Ectrodactyly, Ectodermal Dysplasia, characterized by limb deformation, cleft lip/palate, and ectodermal dysplasia while mutations in in the C-terminal domain of the α-isoform cause Ankyloblepharon-Ectodermal defects-Cleft lip/palate (AEC) syndrome, a life-threatening disorder characterized by skin fragility, severe, long-lasting skin erosions, and cleft lip/palate. The molecular disease mechanisms of these syndromes have recently become elucidated and have enhanced our understanding of the role of p63 in epidermal development. Here we review the molecular cause and functional consequences of these p63-mutations for skin development and discuss the consequences of p63 mutations for female fertility.
Collapse
|
21
|
Lena AM, Rossi V, Osterburg S, Smirnov A, Osterburg C, Tuppi M, Cappello A, Amelio I, Dötsch V, De Felici M, Klinger FG, Annicchiarico-Petruzzelli M, Valensise H, Melino G, Candi E. The p63 C-terminus is essential for murine oocyte integrity. Nat Commun 2021; 12:383. [PMID: 33452256 PMCID: PMC7810856 DOI: 10.1038/s41467-020-20669-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 11/26/2020] [Indexed: 01/21/2023] Open
Abstract
The transcription factor p63 mediates distinct cellular responses, primarily regulating epithelial and oocyte biology. In addition to the two amino terminal isoforms, TAp63 and ΔNp63, the 3'-end of p63 mRNA undergoes tissue-specific alternative splicing that leads to several isoforms, including p63α, p63β and p63γ. To investigate in vivo how the different isoforms fulfil distinct functions at the cellular and developmental levels, we developed a mouse model replacing the p63α with p63β by deletion of exon 13 in the Trp63 gene. Here, we report that whereas in two organs physiologically expressing p63α, such as thymus and skin, no abnormalities are detected, total infertility is evident in heterozygous female mice. A sharp reduction in the number of primary oocytes during the first week after birth occurs as a consequence of the enhanced expression of the pro-apoptotic transcriptional targets Puma and Noxa by the tetrameric, constitutively active, TAp63β isoform. Hence, these mice show a condition of ovary dysfunction, resembling human primary ovary insufficiency. Our results show that the p63 C-terminus is essential in TAp63α-expressing primary oocytes to control cell death in vivo, expanding the current understanding of human primary ovarian insufficiency.
Collapse
Affiliation(s)
- Anna Maria Lena
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Valerio Rossi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Susanne Osterburg
- Institute of Biophysical Chemistry, Center for Biomolecular Magnetic Resonance and Cluster of Excellence Macromolecular Complexes (CEF), Goethe University, Frankfurt, Germany
| | - Artem Smirnov
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
- Ludwig Institute for Cancer Research, University of Oxford, Oxford, OX3 7DQ, UK
| | - Christian Osterburg
- Institute of Biophysical Chemistry, Center for Biomolecular Magnetic Resonance and Cluster of Excellence Macromolecular Complexes (CEF), Goethe University, Frankfurt, Germany
| | - Marcel Tuppi
- Institute of Biophysical Chemistry, Center for Biomolecular Magnetic Resonance and Cluster of Excellence Macromolecular Complexes (CEF), Goethe University, Frankfurt, Germany
- The Francis Crick Institute, London, NW11ST, UK
| | - Angela Cappello
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Ivano Amelio
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Volker Dötsch
- Institute of Biophysical Chemistry, Center for Biomolecular Magnetic Resonance and Cluster of Excellence Macromolecular Complexes (CEF), Goethe University, Frankfurt, Germany
| | - Massimo De Felici
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | | | | | - Herbert Valensise
- Department of Surgery, University of Rome "Tor Vergata", Rome, Italy
- Policlinico "Casilino", Rome, Italy
| | - Gerry Melino
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy.
| | - Eleonora Candi
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy.
- IDI-IRCCS, Via dei Monti di Creta, Rome, Italy.
| |
Collapse
|
22
|
Zhang J, Yang P, Liu D, Gao M, Wang J, Wang X, Liu Y, Zhang X. c-Myc Upregulated by High Glucose Inhibits HaCaT Differentiation by S100A6 Transcriptional Activation. Front Endocrinol (Lausanne) 2021; 12:676403. [PMID: 34060533 PMCID: PMC8163689 DOI: 10.3389/fendo.2021.676403] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 04/30/2021] [Indexed: 01/13/2023] Open
Abstract
Keratinocyte differentiation dysfunction in diabetic skin is closely related to impaired skin barrier functions. We investigated the effects of c-Myc and S100A6 on Human immortal keratinocyte line (HaCaT) or keratinocyte differentiation and potential mechanisms. The expression levels of differentiation makers such as transglutaminase 1 (TGM1), loricrin (LOR), and keratin 1 (K1) were significantly reduced, while the expression of c-Myc was significantly increased in HaCaT cells cultured in high glucose and wound margin keratinocytes from diabetic rats and human patients. Overexpression of c-Myc caused differentiation dysfunction of HaCaT, while knocking down c-Myc promoted differentiation. High glucose increased the expression of c-Myc and inhibited differentiation in HaCaT cells by activating the WNT/β-catenin pathway. Moreover, inhibition of c-Myc transcriptional activity alleviated the differentiation dysfunction caused by high glucose or overexpression of c-Myc. c-Myc binds to the S100A6 promoter to directly regulate S100A6 expression and high glucose promoted S100A6 transcription. The expression of S100A6 was increased in HaCaT cultured with high glucose and wound margin keratinocytes from diabetic rats and human patients. However, the expression of S100A6 was decreased during normal HaCaT differentiation. HaCaT cells treated with S100A6 recombinant protein showed differentiation dysfunction. The expressions of TGM1, LOR and K1 in knockdown S100A6 HaCaT cells were higher than those in the control group. Overexpression of c-Myc or high glucose caused differentiation dysfunction of HaCaT cells, and was rescued by knocking down S100A6. These findings illustrate a new mechanism by which c-Myc upregulated by high glucose inhibits HaCaT differentiation by directly activating S100A6 transcription. Thus, c-Myc and S100A6 may be potential targets for the treatment of chronic diabetic wounds.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiqiao Wang
- *Correspondence: Xiong Zhang, ; Yan Liu, ; Xiqiao Wang,
| | - Yan Liu
- *Correspondence: Xiong Zhang, ; Yan Liu, ; Xiqiao Wang,
| | - Xiong Zhang
- *Correspondence: Xiong Zhang, ; Yan Liu, ; Xiqiao Wang,
| |
Collapse
|
23
|
Distinct p63 and p73 Protein Interactions Predict Specific Functions in mRNA Splicing and Polyploidy Control in Epithelia. Cells 2020; 10:cells10010025. [PMID: 33375680 PMCID: PMC7824480 DOI: 10.3390/cells10010025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/20/2020] [Accepted: 12/23/2020] [Indexed: 12/14/2022] Open
Abstract
Epithelial organs are the first barrier against microorganisms and genotoxic stress, in which the p53 family members p63 and p73 have both overlapping and distinct functions. Intriguingly, p73 displays a very specific localization to basal epithelial cells in human tissues, while p63 is expressed in both basal and differentiated cells. Here, we analyse systematically the literature describing p63 and p73 protein-protein interactions to reveal distinct functions underlying the aforementioned distribution. We have found that p73 and p63 cooperate in the genome stability surveillance in proliferating cells; p73 specific interactors contribute to the transcriptional repression, anaphase promoting complex and spindle assembly checkpoint, whereas p63 specific interactors play roles in the regulation of mRNA processing and splicing in both proliferating and differentiated cells. Our analysis reveals the diversification of the RNA and DNA specific functions within the p53 family.
Collapse
|
24
|
Guan Y, Yang YJ, Nagarajan P, Ge Y. Transcriptional and signalling regulation of skin epithelial stem cells in homeostasis, wounds and cancer. Exp Dermatol 2020; 30:529-545. [PMID: 33249665 DOI: 10.1111/exd.14247] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 10/10/2020] [Accepted: 11/13/2020] [Indexed: 02/06/2023]
Abstract
The epidermis and skin appendages are maintained by their resident epithelial stem cells, which undergo long-term self-renewal and multilineage differentiation. Upon injury, stem cells are activated to mediate re-epithelialization and restore tissue function. During this process, they often mount lineage plasticity and expand their fates in response to damage signals. Stem cell function is tightly controlled by transcription machineries and signalling transductions, many of which derail in degenerative, inflammatory and malignant dermatologic diseases. Here, by describing both well-characterized and newly emerged pathways, we discuss the transcriptional and signalling mechanisms governing skin epithelial homeostasis, wound repair and squamous cancer. Throughout, we highlight common themes underscoring epithelial stem cell plasticity and tissue-level crosstalk in the context of skin physiology and pathology.
Collapse
Affiliation(s)
- Yinglu Guan
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Youn Joo Yang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Priyadharsini Nagarajan
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yejing Ge
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
25
|
Yi M, Tan Y, Wang L, Cai J, Li X, Zeng Z, Xiong W, Li G, Li X, Tan P, Xiang B. TP63 links chromatin remodeling and enhancer reprogramming to epidermal differentiation and squamous cell carcinoma development. Cell Mol Life Sci 2020; 77:4325-4346. [PMID: 32447427 PMCID: PMC7588389 DOI: 10.1007/s00018-020-03539-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 02/21/2020] [Accepted: 04/24/2020] [Indexed: 12/19/2022]
Abstract
Squamous cell carcinoma (SCC) is an aggressive malignancy that can originate from various organs. TP63 is a master regulator that plays an essential role in epidermal differentiation. It is also a lineage-dependent oncogene in SCC. ΔNp63α is the prominent isoform of TP63 expressed in epidermal cells and SCC, and overexpression promotes SCC development through a variety of mechanisms. Recently, ΔNp63α was highlighted to act as an epidermal-specific pioneer factor that binds closed chromatin and enhances chromatin accessibility at epidermal enhancers. ΔNp63α coordinates chromatin-remodeling enzymes to orchestrate the tissue-specific enhancer landscape and three-dimensional high-order architecture of chromatin. Moreover, ΔNp63α establishes squamous-like enhancer landscapes to drive oncogenic target expression during SCC development. Importantly, ΔNp63α acts as an upstream regulator of super enhancers to activate a number of oncogenic transcripts linked to poor prognosis in SCC. Mechanistically, ΔNp63α activates genes transcription through physically interacting with a number of epigenetic modulators to establish enhancers and enhance chromatin accessibility. In contrast, ΔNp63α also represses gene transcription via interacting with repressive epigenetic regulators. ΔNp63α expression is regulated at multiple levels, including transcriptional, post-transcriptional, and post-translational levels. In this review, we summarize recent advances of p63 in epigenomic and transcriptional control, as well as the mechanistic regulation of p63.
Collapse
Affiliation(s)
- Mei Yi
- NHC Key Laboratory of Carcinogenesis, Hunan Provincial Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, 410078, Hunan, China
| | - Yixin Tan
- Department of Dermatology, The Second Xiangya Hospital, The Central South University, Changsha, 410011, Hunan, China
| | - Li Wang
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Jing Cai
- NHC Key Laboratory of Carcinogenesis, Hunan Provincial Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, 410078, Hunan, China
| | - Xiaoling Li
- NHC Key Laboratory of Carcinogenesis, Hunan Provincial Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, 410078, Hunan, China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis, Hunan Provincial Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, 410078, Hunan, China
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis, Hunan Provincial Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, 410078, Hunan, China
| | - Guiyuan Li
- NHC Key Laboratory of Carcinogenesis, Hunan Provincial Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, 410078, Hunan, China
| | - Xiayu Li
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China.
| | - Pingqing Tan
- NHC Key Laboratory of Carcinogenesis, Hunan Provincial Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.
- Department of Head and Neck Surgery, Hunan Provincial Cancer Hospital and Cancer Hospital Affiliated to Xiangya Medical School, Central South University, Changsha, 410013, Hunan, China.
| | - Bo Xiang
- NHC Key Laboratory of Carcinogenesis, Hunan Provincial Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China.
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, 410078, Hunan, China.
| |
Collapse
|
26
|
Ishida K, Tomita H, Kanayama T, Noguchi K, Niwa A, Kawaguchi M, Miyai M, Matsuo M, Imaizumi Y, Kato K, Hatano Y, Hirata A, Okada H, Shibata T, Hara A. Specific Deletion of p16 INK4a with Retention of p19 ARF Enhances the Development of Invasive Oral Squamous Cell Carcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:1332-1342. [PMID: 32194051 DOI: 10.1016/j.ajpath.2020.01.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 01/05/2020] [Accepted: 01/30/2020] [Indexed: 12/15/2022]
Abstract
The cyclin-dependent kinase inhibitor 2A (CDKN2A)/alternate reading frame (ARF) locus consists of two overlapping tumor suppressor genes, p16INK4a and p14ARF (p19ARF in mice), encoding two unrelated proteins in alternative reading frames. Previous reports suggest that p16INK4a and p14ARF alterations independently exhibit differential roles, and p16INK4a is more closely associated with a poor prognosis in oral cancer. However, the role of p16INK4a-specific loss in oral squamous cell carcinogenesis remains unclear. The authors assessed chemical carcinogen 4-nitroquinoline 1-oxide (4NQO)-induced multistep oral squamous cell carcinogenesis in mice carrying p16INK4a-specific loss with retention of the p19ARF gene (p16INK4a-/-). 4NQO-treated p16-/- mice exhibited a higher incidence and multiplicity of oral squamous cell carcinoma (OSCC) development relative to 4NQO-treated wild-type mice. 4NQO-treated p16INK4a-/- OSCC cells exhibited higher proliferation and up-regulation of Arf, transcription factor E2f1, tumor protein p63 (tp63), and oncogenic ΔNp63, an isoform p63, compared with observations in 4NQO-treated wild-type OSCC cells. Furthermore, the overexpression of oncogenic ΔNp63 was associated with human OSCC. In conclusion, these results in mice indicate the biological significance of p16INK4a-specific loss with retention of p19ARF in oral squamous cell carcinogenesis, and ΔNp63 may be a potential target for OSCC.
Collapse
Affiliation(s)
- Kazuhisa Ishida
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan; Department of Oral Maxillofacial Surgery, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Hiroyuki Tomita
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan.
| | - Tomohiro Kanayama
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Kei Noguchi
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Ayumi Niwa
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Masaya Kawaguchi
- Department of Radiology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Masafumi Miyai
- Department of Neuronal Surgery, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Mikiko Matsuo
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Yuko Imaizumi
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Keizo Kato
- Department of Oral Maxillofacial Surgery, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Yuichiro Hatano
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Akihiro Hirata
- Division of Animal Experiment, Life Science Research Center, Gifu University, Gifu, Japan
| | - Hideshi Okada
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Toshiyuki Shibata
- Department of Oral Maxillofacial Surgery, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Akira Hara
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan
| |
Collapse
|
27
|
Diluvio L, Caporali S, Lozzi F, Campione E, Mazzilli S, Lanna C, Bianchi L, Bernardini S, Minieri M, Mauriello A, Ferlosio A, Candi E, Terrinoni A. Birt-Hogg-Dubé syndrome, from non-invasive dermatologic assessment to gene testing, molecular and ultrastructural histologic analysis. J Eur Acad Dermatol Venereol 2020; 34:e206-e209. [PMID: 31876332 DOI: 10.1111/jdv.16168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- L Diluvio
- Dermatology Unit of Department of Systems Medicine, University of Rome Tor vergata, Rome, Italy
| | - S Caporali
- Department of Industrial Engineering, University of Rome Tor Vergata, Rome, Italy
| | - F Lozzi
- Dermatology Unit of Department of Systems Medicine, University of Rome Tor vergata, Rome, Italy
| | - E Campione
- Dermatology Unit of Department of Systems Medicine, University of Rome Tor vergata, Rome, Italy
| | - S Mazzilli
- Dermatology Unit of Department of Systems Medicine, University of Rome Tor vergata, Rome, Italy
| | - C Lanna
- Dermatology Unit of Department of Systems Medicine, University of Rome Tor vergata, Rome, Italy
| | - L Bianchi
- Dermatology Unit of Department of Systems Medicine, University of Rome Tor vergata, Rome, Italy
| | - S Bernardini
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - M Minieri
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - A Mauriello
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - A Ferlosio
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - E Candi
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy.,Biochemistry Laboratory, Fondazione Luigi Maria Monti, IDI-IRCCS, Rome, Italy
| | - A Terrinoni
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
28
|
Panatta E, Lena AM, Mancini M, Smirnov A, Marini A, Delli Ponti R, Botta-Orfila T, Tartaglia GG, Mauriello A, Zhang X, Calin GA, Melino G, Candi E. Long non-coding RNA uc.291 controls epithelial differentiation by interfering with the ACTL6A/BAF complex. EMBO Rep 2020; 21:e46734. [PMID: 32017402 PMCID: PMC7054673 DOI: 10.15252/embr.201846734] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 12/23/2019] [Accepted: 01/08/2020] [Indexed: 11/29/2022] Open
Abstract
The mechanisms that regulate the switch between epidermal progenitor state and differentiation are not fully understood. Recent findings indicate that the chromatin remodelling BAF complex (Brg1‐associated factor complex or SWI/SNF complex) and the transcription factor p63 mutually recruit one another to open chromatin during epidermal differentiation. Here, we identify a long non‐coding transcript that includes an ultraconserved element, uc.291, which physically interacts with ACTL6A and modulates chromatin remodelling to allow differentiation. Loss of uc.291 expression, both in primary keratinocytes and in three‐dimensional skin equivalents, inhibits differentiation as indicated by epidermal differentiation complex genes down‐regulation. ChIP experiments reveal that upon uc.291 depletion, ACTL6A is bound to the differentiation gene promoters and inhibits BAF complex targeting to induce terminal differentiation genes. In the presence of uc.291, the ACTL6A inhibitory effect is released, allowing chromatin changes to promote the expression of differentiation genes. Thus, uc.291 interacts with ACTL6A to modulate chromatin remodelling activity, allowing the transcription of late differentiation genes.
Collapse
Affiliation(s)
- Emanuele Panatta
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy.,MRC Toxicology Unit, University of Cambridge, Cambridge, UK
| | - Anna Maria Lena
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | | | - Artem Smirnov
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Alberto Marini
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy.,MRC Toxicology Unit, University of Cambridge, Cambridge, UK
| | - Riccardo Delli Ponti
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Teresa Botta-Orfila
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Gian Gaetano Tartaglia
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain.,Institucio Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Alessandro Mauriello
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Xinna Zhang
- The Center for RNA Interference and Non-coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - George A Calin
- The Center for RNA Interference and Non-coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gerry Melino
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy.,MRC Toxicology Unit, University of Cambridge, Cambridge, UK
| | - Eleonora Candi
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy.,IDI-IRCCS, Rome, Italy
| |
Collapse
|
29
|
Guan Y, Wang G, Fails D, Nagarajan P, Ge Y. Unraveling cancer lineage drivers in squamous cell carcinomas. Pharmacol Ther 2020; 206:107448. [PMID: 31836455 PMCID: PMC6995404 DOI: 10.1016/j.pharmthera.2019.107448] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 12/03/2019] [Indexed: 12/12/2022]
Abstract
Cancer hijacks embryonic development and adult wound repair mechanisms to fuel malignancy. Cancer frequently originates from de-regulated adult stem cells or progenitors, which are otherwise essential units for postnatal tissue remodeling and repair. Cancer genomics studies have revealed convergence of multiple cancers across organ sites, including squamous cell carcinomas (SCCs), a common group of cancers arising from the head and neck, esophagus, lung, cervix and skin. In this review, we summarize our current knowledge on the molecular drivers of SCCs, including these five major organ sites. We especially focus our discussion on lineage dependent driver genes and pathways, in the context of squamous development and stratification. We then use skin as a model to discuss the notion of field cancerization during SCC carcinogenesis, and cancer as a wound that never heals. Finally, we turn to the idea of context dependency widely observed in cancer driver genes, and outline literature support and possible explanations for their lineage specific functions. Through these discussions, we aim to provide an up-to-date summary of molecular mechanisms driving tumor plasticity in squamous cancers. Such basic knowledge will be helpful to inform the clinics for better stratifying cancer patients, revealing novel drug targets and providing effective treatment options.
Collapse
Affiliation(s)
- Yinglu Guan
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Guan Wang
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Danielle Fails
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Priyadharsini Nagarajan
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Yejing Ge
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA.
| |
Collapse
|
30
|
Regulation of Stem Cells by Cullin-RING Ligase. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1217:79-98. [PMID: 31898223 DOI: 10.1007/978-981-15-1025-0_6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Stem cells can remain quiescent, self-renewal, and differentiate into many types of cells and even cancer stem cells. The coordination of these complex processes maintains the homeostasis of the organism. Ubiquitination is an important posttranslational modification process that regulates protein stability and activity. The ubiquitination levels of stem cell-associated proteins are closely related with stem cell characteristics. Cullin-RING Ligases (CRLs) are the largest family of E3 ubiquitin ligases, accounting for approximately 20% of proteins degraded by proteasome. In this review, we discuss the role of CRLs in stem cell homeostasis, self-renewal, and differentiation and expound their ubiquitination substrates. In addition, we also discuss the effect of CRLs on the formation of cancer stem cells that may provide promising therapy strategies for cancer.
Collapse
|
31
|
Frezza V, Fierro C, Gatti E, Peschiaroli A, Lena AM, Petruzzelli MA, Candi E, Anemona L, Mauriello A, Pelicci PG, Melino G, Bernassola F. ΔNp63 promotes IGF1 signalling through IRS1 in squamous cell carcinoma. Aging (Albany NY) 2019; 10:4224-4240. [PMID: 30594912 PMCID: PMC6326668 DOI: 10.18632/aging.101725] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 12/12/2018] [Indexed: 02/07/2023]
Abstract
Accumulating evidence has proved that deregulation of ΔNp63 expression plays an oncogenic role in head and neck squamous cell carcinomas (HNSCCs). Besides p63, the type 1-insulin-like growth factor (IGF) signalling pathway has been implicated in HNSCC development and progression. Most insulin/IGF1 signalling converges intracellularly onto the protein adaptor insulin receptor substrate-1 (IRS-1) that transmits signals from the receptor to downstream effectors, including the PI3K/AKT and the MAPK kinase pathways, which, ultimately, promote proliferation, invasion, and cell survival. Here we report that p63 directly controls IRS1 transcription and cellular abundance and fosters the PI3K/AKT and MAPK downstream signalling pathways. Inactivation of ΔNp63 expression indeed reduces tumour cell responsiveness to IGF1 stimulation, and inhibits the growth potential of HNSCC cells. In addition, a positive correlation was observed between p63 and IRS1 expression in human HNSCC tissue arrays and in publicly available gene expression data. Our findings indicate that aberrant expression of ΔNp63 in HNSSC may act as an oncogenic stimulus by altering the IGF signalling pathway.
Collapse
Affiliation(s)
- Valentina Frezza
- Department of Experimental Medicine, TOR University of Rome "Tor Vergata", Rome 00133, Italy
| | - Claudia Fierro
- Department of Experimental Medicine, TOR University of Rome "Tor Vergata", Rome 00133, Italy
| | - Elena Gatti
- Department of Experimental Oncology European Institute of Oncology, Milan 20139, Italy
| | - Angelo Peschiaroli
- National Research Council of Italy Institute of Translational Pharmacology (IFT-CNR), Rome 00133, Italy
| | - Anna Maria Lena
- Department of Experimental Medicine, TOR University of Rome "Tor Vergata", Rome 00133, Italy
| | | | - Eleonora Candi
- Department of Experimental Medicine, TOR University of Rome "Tor Vergata", Rome 00133, Italy.,Istituto Dermopatico dell'Immacolata, IRCCS,, Rome 00163, Italy
| | - Lucia Anemona
- Department of Experimental Medicine, TOR University of Rome "Tor Vergata", Rome 00133, Italy
| | - Alessandro Mauriello
- Department of Experimental Medicine, TOR University of Rome "Tor Vergata", Rome 00133, Italy
| | - Pier Giuseppe Pelicci
- Department of Experimental Oncology European Institute of Oncology, Milan 20139, Italy
| | - Gerry Melino
- Department of Experimental Medicine, TOR University of Rome "Tor Vergata", Rome 00133, Italy.,Medical Research Council, Toxicology Unit, University of Cambridge, Leicester LE1 9HN, UK
| | - Francesca Bernassola
- Department of Experimental Medicine, TOR University of Rome "Tor Vergata", Rome 00133, Italy
| |
Collapse
|
32
|
Smirnov A, Anemona L, Novelli F, Piro CM, Annicchiarico-Petruzzelli M, Melino G, Candi E. p63 Is a Promising Marker in the Diagnosis of Unusual Skin Cancer. Int J Mol Sci 2019; 20:E5781. [PMID: 31744230 PMCID: PMC6888618 DOI: 10.3390/ijms20225781] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/07/2019] [Accepted: 11/15/2019] [Indexed: 01/02/2023] Open
Abstract
Skin cancer is the most common type of cancer worldwide. Ozone depletion and climate changes might cause a further increase in the incidence rate in the future. Although the early detection of skin cancer enables it to be treated successfully, some tumours can evolve and become more aggressive, especially in the case of melanoma. Therefore, good diagnostic and prognostic markers are needed to ensure correct detection and treatment. Transcription factor p63, a member of the p53 family of proteins, plays an essential role in the development of stratified epithelia such as skin. In this paper, we conduct a comprehensive review of p63 expression in different types of skin cancer and discuss its possible use in the diagnosis and prognosis of cutaneous tumours.
Collapse
Affiliation(s)
- Artem Smirnov
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Lucia Anemona
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Flavia Novelli
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Cristina M. Piro
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | | | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy
- MRC-Toxicology Unit, University of Cambridge, Cambridge CB2 1QP, UK
| | - Eleonora Candi
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133 Rome, Italy
- Istituto Dermopatico dell’Immacolata-IRCCS, 00163 Rome, Italy
| |
Collapse
|
33
|
Stacy AJ, Zhang J, Craig MP, Hira A, Dole N, Kadakia MP. TIP60 up-regulates ΔNp63α to promote cellular proliferation. J Biol Chem 2019; 294:17007-17016. [PMID: 31601649 DOI: 10.1074/jbc.ra119.010388] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/03/2019] [Indexed: 01/08/2023] Open
Abstract
An estimated 5.4 million cases of nonmelanoma skin cancer are reported in the United States at an associated cost of $4.8 billion. ΔNp63α, a proto-oncogene in the p53 family of transcription factors, is overexpressed in squamous cell carcinoma (SCC) and associated with poor prognosis and survival. ΔNp63α elicits its tumorigenic effects in part by promoting cellular proliferation and cell survival. Despite its importance in SCC, the upstream regulation of ΔNp63α is poorly understood. In this study, we identify TIP60 as a novel upstream regulator of ΔNp63α. Using a combination of overexpression, silencing, stable expression, and pharmacological approaches in multiple cell lines, we showed that TIP60 up-regulates ΔNp63α expression. Utilizing cycloheximide treatment, we showed that TIP60 catalytic activity is required for stabilization of ΔNp63α protein levels. We further showed that TIP60 coexpression inhibits ΔNp63α ubiquitination and proteasomal degradation. Stabilization of ΔNp63α protein was further associated with TIP60-mediated acetylation. Finally, we demonstrated that TIP60-mediated regulation of ΔNp63α increases cellular proliferation by promoting G2/M progression through MTS assays and flow cytometry. Taken together, our findings provide evidence that TIP60 may contribute to SCC progression by increasing ΔNp63α protein levels, thereby promoting cellular proliferation.
Collapse
Affiliation(s)
- Andrew J Stacy
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio 45435
| | - Jin Zhang
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio 45435
| | - Michael P Craig
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio 45435
| | - Akshay Hira
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio 45435
| | - Nikhil Dole
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio 45435
| | - Madhavi P Kadakia
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio 45435
| |
Collapse
|
34
|
Werner S, Keller L, Pantel K. Epithelial keratins: Biology and implications as diagnostic markers for liquid biopsies. Mol Aspects Med 2019; 72:100817. [PMID: 31563278 DOI: 10.1016/j.mam.2019.09.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 09/02/2019] [Accepted: 09/04/2019] [Indexed: 02/07/2023]
Abstract
Keratins are essential elements of the cytoskeleton of normal and malignant epithelial cells. Because carcinomas commonly maintain their specific keratin expression pattern during malignant transformation, keratins are extensively used as tumor markers in cancer diagnosis including the detection of circulating tumor cells in blood of carcinoma patients. Interestingly, recent biological insights demonstrate that epithelial keratins should not only be considered as mere tumor markers. Emerging evidence suggests an active biological role of keratins in tumor cell dissemination and metastasis. In this review, we illustrate the family of keratin proteins, summarize the latest biological insights into keratin function related to cancer metastasis and discuss the current use of keratins for detection of CTCs and other blood biomarkers used in oncology.
Collapse
Affiliation(s)
- Stefan Werner
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Laura Keller
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Klaus Pantel
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
35
|
Ectrodactyly-ectodermal dysplasia-clefting syndrome with unusual cutaneous vitiligoid and psoriasiform lesions due to a novel single point TP63 gene mutation. Postepy Dermatol Alergol 2019; 36:358-364. [PMID: 31333354 PMCID: PMC6640015 DOI: 10.5114/ada.2018.73437] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 01/06/2018] [Indexed: 12/11/2022] Open
|
36
|
Molecular Mechanisms of p63-Mediated Squamous Cancer Pathogenesis. Int J Mol Sci 2019; 20:ijms20143590. [PMID: 31340447 PMCID: PMC6678256 DOI: 10.3390/ijms20143590] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/12/2019] [Accepted: 07/15/2019] [Indexed: 12/20/2022] Open
Abstract
The p63 gene is a member of the p53/p63/p73 family of transcription factors and plays a critical role in development and homeostasis of squamous epithelium. p63 is transcribed as multiple isoforms; ΔNp63α, the predominant p63 isoform in stratified squamous epithelium, is localized to the basal cells and is overexpressed in squamous cell cancers of multiple organ sites, including skin, head and neck, and lung. Further, p63 is considered a stem cell marker, and within the epidermis, ΔNp63α directs lineage commitment. ΔNp63α has been implicated in numerous processes of skin biology that impact normal epidermal homeostasis and can contribute to squamous cancer pathogenesis by supporting proliferation and survival with roles in blocking terminal differentiation, apoptosis, and senescence, and influencing adhesion and migration. ΔNp63α overexpression may also influence the tissue microenvironment through remodeling of the extracellular matrix and vasculature, as well as by enhancing cytokine and chemokine secretion to recruit pro-inflammatory infiltrate. This review focuses on the role of ΔNp63α in normal epidermal biology and how dysregulation can contribute to cutaneous squamous cancer development, drawing from knowledge also gained by squamous cancers from other organ sites that share p63 overexpression as a defining feature.
Collapse
|
37
|
Wang Y, Li J, Gao Y, Luo Y, Luo H, Wang L, Yi Y, Yuan Z, Jim Xiao ZX. Hippo kinases regulate cell junctions to inhibit tumor metastasis in response to oxidative stress. Redox Biol 2019; 26:101233. [PMID: 31212215 PMCID: PMC6582208 DOI: 10.1016/j.redox.2019.101233] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/22/2019] [Accepted: 05/31/2019] [Indexed: 12/29/2022] Open
Abstract
Reactive oxygen species (ROS) are key regulators in cell proliferation, survival, tumor initiation and development. However, the role of ROS in tumor metastasis is less clear. Here, we show that oxidative stress inhibited tumor metastasis via activation of Hippo kinase MST1/2, which led to the phosphorylation and nuclear accumulation of FoxO3a, resulting in upregulation of ΔNp63α expression and suppression of cell migration independent of YAP. Strikingly, while loss of MST1 led to and disruption of cell-cell junction exemplified by reduced E-cadherin expression, resulting in scattered cell growth, loss of MST2 led to disruption of cell-matrix adhesion as evidenced by reduced integrin β4, resulting in increased cell migration and tumor metastasis. Furthermore, expression of MST1 and MST2 was down-regulated in human breast carcinoma. Furthermore, oxidative stress inhibited HER2-or PI3K-mediated tumor metastasis via the MST2-FoxO3a-ΔNp63α pathway. Together, these results that this noncanonical Hippo MST2-FoxO3a-ΔNp63α pathway may play a critical role in ROS-mediated regulation of cell migration and tumor metastasis.
Collapse
Affiliation(s)
- Yang Wang
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, China
| | - Juan Li
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, China
| | - Ya Gao
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, China
| | - Yue Luo
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, China
| | - Hong Luo
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, China
| | - Liang Wang
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, China
| | - Yong Yi
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, China
| | - Zengqiang Yuan
- Institute of Basic Medical Sciences, AMMS, Beijing, 100850, China
| | - Zhi-Xiong Jim Xiao
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, China; Department of Biochemistry, Boston University School of Medicine, Boston, MA, 02118, USA.
| |
Collapse
|
38
|
Palombo R, Caporali S, Falconi M, Iacovelli F, Morozzo Della Rocca B, Lo Surdo A, Campione E, Candi E, Melino G, Bernardini S, Terrinoni A. Luteolin-7- O-β-d-Glucoside Inhibits Cellular Energy Production Interacting with HEK2 in Keratinocytes. Int J Mol Sci 2019; 20:ijms20112689. [PMID: 31159225 PMCID: PMC6600217 DOI: 10.3390/ijms20112689] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/06/2019] [Accepted: 05/15/2019] [Indexed: 01/10/2023] Open
Abstract
Flavonoids have been demonstrated to affect the activity of many mammalian enzyme systems. Their functional phenolic groups are able to mediate antioxidant effects by scavenging free radicals. Molecules of this class have been found able to modulate the activity of kinases, phospholipase A2, cyclooxygenases, lipoxygenase, glutathione S-transferase, and many others. Recently, it has been demonstrated that luteolin, in the form of Luteolin-7-O-β-d-glucoside (LUT-7G) is able to induce the keratinocyte differentiation process in vitro. This flavonoid is able to counteract the proliferative effects of IL-22/IL6 pathway by the inhibition of STAT3 activity also in vivo in a psoriatic mouse model. Observations on energy metabolism changes of differentiating cells led us to perform a complete metabolomics analysis using human primary keratinocytes treated with LUT-7G. Our results show that LUT-7G, is not only able to impair the nuclear translocation of STAT3, but it also blocks the energy metabolism pathway, depressing the glycolytic and Krebs pathway by the inhibition of hexokinase 2 activity. These data confirm that LUT-7G can be proposed as a potential candidate for the treatment of inflammatory and proliferative diseases, but its role as a hexokinase 2 (HEK2) inhibitor opens new perspectives in nutritional science, and especially in cancer therapy, in which the inhibition of the Warburg effect could be relevant.
Collapse
Affiliation(s)
- Ramona Palombo
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier, 1, 00133 Rome, Italy.
- Laboratory of Cellular and Molecular Neurobiology, Fondazione Santa Lucia, Via del Fosso di Fiorano, 64, 00143 Rome, Italy.
| | - Sabrina Caporali
- Department of Industrial Engineering, University of Rome Tor Vergata, 00133 Rome, Italy.
| | - Mattia Falconi
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy.
| | - Federico Iacovelli
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy.
| | - Blasco Morozzo Della Rocca
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy.
| | - Alessandro Lo Surdo
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier, 1, 00133 Rome, Italy.
| | - Elena Campione
- Department of Systems Medicine, Dermatologic Unit, University of Rome Tor Vergata, 00133 Rome, Italy.
| | - Eleonora Candi
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier, 1, 00133 Rome, Italy.
- IDI-IRCCS, Biochemistry Laboratory, via dei Monti di Creta, 104, 00167 Rome, Italy.
| | - Gerry Melino
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier, 1, 00133 Rome, Italy.
| | - Sergio Bernardini
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier, 1, 00133 Rome, Italy.
| | - Alessandro Terrinoni
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier, 1, 00133 Rome, Italy.
| |
Collapse
|
39
|
Yi Y, Zhang W, Yi J, Xiao ZX. Role of p53 Family Proteins in Metformin Anti-Cancer Activities. J Cancer 2019; 10:2434-2442. [PMID: 31258748 PMCID: PMC6584340 DOI: 10.7150/jca.30659] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 04/23/2019] [Indexed: 12/13/2022] Open
Abstract
Metformin has been used as therapy for type 2 diabetes for many years. Clinical and basic evidence as indicated that metformin has anti-cancer activities. It has been well-established that metformin activates AMP-activated protein kinase (AMPK), which in turn regulates energy homeostasis. However, the mechanistic aspects of metformin anti-cancer activity remain elusive. p53 family proteins, including p53, p63 and p73, have diverse biological functions, including regulation of cell growth, survival, development, senescence and aging. In this review, we highlight the evidence and mechanisms by which metformin inhibits cancer cell survival and tumor growth. We also aimed to discuss the role of p53 family proteins in metformin-mediated suppression of cancer growth and survival.
Collapse
|
40
|
Fang X, Ni N, Lydon JP, Ivanov I, Bayless KJ, Rijnkels M, Li Q. Enhancer of Zeste 2 Polycomb Repressive Complex 2 Subunit Is Required for Uterine Epithelial Integrity. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1212-1225. [PMID: 30954472 DOI: 10.1016/j.ajpath.2019.02.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 02/11/2019] [Accepted: 02/13/2019] [Indexed: 12/25/2022]
Abstract
Normal proliferation and differentiation of uterine epithelial cells are critical for uterine development and function. Enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2), a core component of polycomb repressive complexes 2, possesses histone methyltransferase activity that catalyzes the trimethylation of lysine 27 of histone H3. EZH2 has been involved in epithelial-mesenchymal transition, a key event in development and carcinogenesis. However, its role in uterine epithelial cell function remains unknown. To determine the role of uterine EZH2, Ezh2 was conditionally deleted using progesterone receptor Cre recombinase, which is expressed in both epithelial and mesenchymal compartments of the uterus. Loss of EZH2 promoted stratification of uterine epithelium, an uncommon and detrimental event in the uterus. The abnormal epithelium expressed basal cell markers, including tumor protein 63, cytokeratin 5 (KRT5), KRT6A, and KRT14. These results suggest that EZH2 serves as a guardian of uterine epithelial integrity, partially via inhibiting the differentiation of basal-like cells and preventing epithelial stratification. The observed epithelial abnormality was accompanied by fertility defects, altered uterine growth and function, and the development of endometrial hyperplasia. Thus, the Ezh2 conditional knockout mouse model may be useful to explore mechanisms that regulate endometrial homeostasis and uterine function.
Collapse
Affiliation(s)
- Xin Fang
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas
| | - Nan Ni
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas
| | - John P Lydon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Ivan Ivanov
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas; Center for Translational Environmental Health Research, Texas A&M University, College Station, Texas
| | - Kayla J Bayless
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, Texas
| | - Monique Rijnkels
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas
| | - Qinglei Li
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas.
| |
Collapse
|
41
|
Fukumoto J, Sidramagowda Patil S, Krishnamurthy S, Saji S, John I, Narala VR, Hernández-Cuervo H, Alleyn M, Breitzig MT, Galam L, Soundararajan R, Chaudhari UK, Hansen BC, Lockey RF, Kolliputi N. Altered expression of p63 isoforms and expansion of p63- and club cell secretory protein-positive epithelial cells in the lung as novel features of aging. Am J Physiol Cell Physiol 2019; 316:C492-C508. [PMID: 30649915 PMCID: PMC6482668 DOI: 10.1152/ajpcell.00330.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 01/04/2019] [Accepted: 01/05/2019] [Indexed: 12/20/2022]
Abstract
Aging is a key contributor for subclinical progression of late-onset lung diseases. Basal, club, and type II alveolar epithelial cells (AECs) are lung epithelial progenitors whose capacities of differentiation are extensively studied. The timely transition of these cells in response to environmental changes helps maintain the intricate organization of lung structure. However, it remains unclear how aging affects their behavior. This paper demonstrates that the protein expression profiles of a type II AEC marker, prosurfactant protein C (pro-SPC), and a basal cell marker, p63, are altered in the lungs of 14-mo-old versus 7- to 9-wk-old mice. Expression of NH2-terminal-truncated forms of p63 (ΔNp63), a basal cell marker, and claudin-10, a club cell marker, in cytoplasmic extracts of lungs of 14-mo-old mice was upregulated. In contrast, nuclear expression of full-length forms of p63 (TAp63) decreases with age. These alterations in protein expression profiles coincide with dramatic changes in lung functions including compliance. Whole tissue lysates of middle-aged versus aged rhesus monkey lungs display similar age-associated alterations in pro-SPC expression. An age-associated decrease of TAp63 in nuclear lysates was observed in aged monkey group. Moreover, the lungs of 14-mo-old versus 7- to 9-wk-old mice display a wider spreading of ΔNp63-positive CCSP-positive bronchiolar epithelial cells. This expansion did not involve upregulation of Ki67, a representative proliferation marker. Collectively, it is postulated that 1) this expansion is secondary to a transition of progenitor cells committed to club cells from ΔNp63-negative to ΔNp63-positive status, and 2) high levels of cytoplasmic ΔNp63 expression trigger club cell migration.
Collapse
Affiliation(s)
- Jutaro Fukumoto
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida , Tampa, Florida
| | - Sahebgowda Sidramagowda Patil
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida , Tampa, Florida
| | - Sudarshan Krishnamurthy
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida , Tampa, Florida
| | - Smita Saji
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida , Tampa, Florida
| | - Irene John
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida , Tampa, Florida
| | - Venkata Ramireddy Narala
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida , Tampa, Florida
- Department of Zoology, Yogi Vemana University, Kadapa, India
| | - Helena Hernández-Cuervo
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida , Tampa, Florida
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida , Tampa, Florida
| | - Matthew Alleyn
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida , Tampa, Florida
| | - Mason T Breitzig
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida , Tampa, Florida
| | - Lakshmi Galam
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida , Tampa, Florida
| | - Ramani Soundararajan
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida , Tampa, Florida
| | - Uddhav K Chaudhari
- Departments of Internal Medicine and Pediatrics, Morsani College of Medicine, University of South Florida , Tampa, Florida
| | - Barbara C Hansen
- Departments of Internal Medicine and Pediatrics, Morsani College of Medicine, University of South Florida , Tampa, Florida
| | - Richard F Lockey
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida , Tampa, Florida
| | - Narasaiah Kolliputi
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida , Tampa, Florida
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida , Tampa, Florida
| |
Collapse
|
42
|
Cox B, Laporte E, Vennekens A, Kobayashi H, Nys C, Van Zundert I, Uji-I H, Vercauteren Drubbel A, Beck B, Roose H, Boretto M, Vankelecom H. Organoids from pituitary as a novel research model toward pituitary stem cell exploration. J Endocrinol 2019; 240:287-308. [PMID: 30475227 DOI: 10.1530/joe-18-0462] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 11/23/2018] [Indexed: 12/18/2022]
Abstract
The pituitary is the master endocrine gland, harboring stem cells of which the phenotype and role remain poorly characterized. Here, we established organoids from mouse pituitary with the aim to generate a novel research model to study pituitary stem cell biology. The organoids originated from the pituitary cells expressing the stem cell marker SOX2 were long-term expandable, displayed a stemness phenotype during expansive culture and showed specific hormonal differentiation ability, although limited, after subrenal transplantation. Application of the protocol to transgenically injured pituitary harboring an activated stem cell population, resulted in more numerous organoids. Intriguingly, these organoids presented with a cystic morphology, whereas the organoids from undamaged gland were predominantly dense and appeared more limited in expandability. Transcriptomic analysis revealed distinct epithelial phenotypes and showed that cystic organoids more resembled the pituitary phenotype, at least to an immature state, and displayed in vitro differentiation, although yet moderate. Organoid characterization further exposed facets of regulatory pathways of the putative stem cells of the pituitary and advanced new injury-activated markers. Taken together, we established a novel organoid research model revealing new insights into the identity and regulation of the putative pituitary stem cells. This organoid model may eventually lead to an interesting tool to decipher pituitary stem cell biology in both healthy and diseased gland.
Collapse
Affiliation(s)
- Benoit Cox
- Department of Development and Regeneration, Cluster of Stem Cell and Developmental Biology, Unit of Stem Cell Research, KU Leuven (University of Leuven), Leuven, Belgium
| | - Emma Laporte
- Department of Development and Regeneration, Cluster of Stem Cell and Developmental Biology, Unit of Stem Cell Research, KU Leuven (University of Leuven), Leuven, Belgium
| | - Annelies Vennekens
- Department of Development and Regeneration, Cluster of Stem Cell and Developmental Biology, Unit of Stem Cell Research, KU Leuven (University of Leuven), Leuven, Belgium
| | - Hiroto Kobayashi
- Department of Development and Regeneration, Cluster of Stem Cell and Developmental Biology, Unit of Stem Cell Research, KU Leuven (University of Leuven), Leuven, Belgium
- Department of Anatomy and Structural Science, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Charlotte Nys
- Department of Development and Regeneration, Cluster of Stem Cell and Developmental Biology, Unit of Stem Cell Research, KU Leuven (University of Leuven), Leuven, Belgium
| | - Indra Van Zundert
- Department of Chemistry, Laboratory of Molecular Imaging and Photonics, KU Leuven, Leuven, Belgium
| | - Hiroshi Uji-I
- Department of Chemistry, Laboratory of Molecular Imaging and Photonics, KU Leuven, Leuven, Belgium
| | | | - Benjamin Beck
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Brussels, Belgium
- WELBIO, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Heleen Roose
- Department of Development and Regeneration, Cluster of Stem Cell and Developmental Biology, Unit of Stem Cell Research, KU Leuven (University of Leuven), Leuven, Belgium
| | - Matteo Boretto
- Department of Development and Regeneration, Cluster of Stem Cell and Developmental Biology, Unit of Stem Cell Research, KU Leuven (University of Leuven), Leuven, Belgium
| | - Hugo Vankelecom
- Department of Development and Regeneration, Cluster of Stem Cell and Developmental Biology, Unit of Stem Cell Research, KU Leuven (University of Leuven), Leuven, Belgium
| |
Collapse
|
43
|
ATDC mediates a TP63-regulated basal cancer invasive program. Oncogene 2019; 38:3340-3354. [PMID: 30643195 PMCID: PMC6499660 DOI: 10.1038/s41388-018-0646-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 12/03/2018] [Accepted: 12/03/2018] [Indexed: 12/20/2022]
Abstract
Basal subtype cancers are deadly malignancies but the molecular events driving tumor lethality are not completely understood. Ataxia-Telangiectasia Group D Complementing gene (ATDC, also known as TRIM29), is highly expressed and drives tumor formation and invasion in human bladder cancers but the factor(s) regulating its expression in bladder cancer are unknown. Molecular subtyping of bladder cancer has identified an aggressive basal subtype which shares molecular features of basal/squamous tumors arising in other organs and is defined by activation of a TP63-driven gene program. Here we demonstrate that ATDC is linked with expression of TP63 and highly expressed in basal bladder cancers. We find that TP63 binds to transcriptional regulatory regions of ATDC and KRT14 directly, increasing their expression, and that ATDC and KRT14 execute a TP63-driven invasive program. In vivo, ATDC is required for TP63-induced bladder tumor invasion and metastasis. These results link TP63 and the basal gene expression program to ATDC and to aggressive tumor behavior. Defining ATDC as a molecular determinant of aggressive, basal cancers may lead to improved biomarkers and therapeutic approaches.
Collapse
|
44
|
Wu-Chou YH, Lu YC, Chen KTP, Chang HF, Lin YT, Lo LJ. Association Studies Between Regulatory Regions of IRF6/ TP63 Genes and Nonsyndromic Oral Clefts. Cleft Palate Craniofac J 2018; 56:778-785. [PMID: 30419764 DOI: 10.1177/1055665618809244] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVE To evaluate genetic variants within the regulatory regions of interferon regulatory factor 6 (IRF6) and TP63 for the etiology of nonsyndromic oral clefts risk factors. DESIGN We performed allelic transmission disequilibrium test analysis on 5 eligible single-nucleotide polymorphisms (SNPs) and SNP haplotypes using the Family-Based Association Test. PARTICIPANTS The study sample consisted of 334 case-parent trios of nonsyndromic oral clefts from Taiwanese population, separated into nonsyndromic cleft lip/palate (NSCL/P) and nonsyndromic cleft palate only (NSCPO) groups. RESULTS We found all 3 selected SNPs of the IRF6 gene show significant association with nonsyndromic oral clefts (rs2235371, P = 5.10E-07; rs642961, P = .00194; and rs77542756, P = 9.08E-07). Haplotype analyses identified 3 possible SNP combination haplotypes in the IRF6 gene and found that C-G-G showed significant undertransmission (P = .058), whereas 2 other haplotypes, T-G-A and C-A-G (P = 2.71E-06 and P = 5.00E-04, respectively), were significantly overtransmitted to the NSCL/P children but not to the NSCPO children. For the TP63 gene, we failed to detect evidence of nonsyndromic oral cleft association in the 2 SNPs within the TP63 large intron 1 region. CONCLUSIONS We used a family-based analysis in 334 Taiwanese case-parent trios to evaluate selected SNPs of IRF6 genes and TP63 genes for a risk of orofacial clefting. This study provides additional evidence for an association between IRF6 and NSCL/P, including the genetic variants within the 5'-noncoding region of the gene. We also confirmed that NSCL/P and NSCPO individuals belong to different groups. For the TP63, our data did not favor the direct involvement of TAp63 isoforms during orofacial development.
Collapse
Affiliation(s)
- Yah-Huei Wu-Chou
- 1 Department of Medical Research, Chang Gung University, Taoyuan.,2 Craniofacial Research Center, Chang Gung Memorial Hospital, Taoyuan
| | - Yi-Chieh Lu
- 1 Department of Medical Research, Chang Gung University, Taoyuan.,2 Craniofacial Research Center, Chang Gung Memorial Hospital, Taoyuan
| | - Kuo-Ting Philip Chen
- 2 Craniofacial Research Center, Chang Gung Memorial Hospital, Taoyuan.,3 Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Taoyuan
| | - Hsien-Fang Chang
- 4 Center of Genomic Medicine, National Taiwan University, Taipei
| | - Yin-Ting Lin
- 1 Department of Medical Research, Chang Gung University, Taoyuan.,2 Craniofacial Research Center, Chang Gung Memorial Hospital, Taoyuan
| | - Lun-Jou Lo
- 2 Craniofacial Research Center, Chang Gung Memorial Hospital, Taoyuan.,3 Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Taoyuan.,5 Graduate Institute of Dental and Craniofacial Science, Chang Gung University, Taoyuan
| |
Collapse
|
45
|
Bhuiyan SA, Ly S, Phan M, Huntington B, Hogan E, Liu CC, Liu J, Pavlidis P. Systematic evaluation of isoform function in literature reports of alternative splicing. BMC Genomics 2018; 19:637. [PMID: 30153812 PMCID: PMC6114036 DOI: 10.1186/s12864-018-5013-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 08/14/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Although most genes in mammalian genomes have multiple isoforms, an ongoing debate is whether these isoforms are all functional as well as the extent to which they increase the functional repertoire of the genome. To ground this debate in data, it would be helpful to have a corpus of experimentally-verified cases of genes which have functionally distinct splice isoforms (FDSIs). RESULTS We established a curation framework for evaluating experimental evidence of FDSIs, and analyzed over 700 human and mouse genes, strongly biased towards genes that are prominent in the alternative splicing literature. Despite this bias, we found experimental evidence meeting the classical definition for functionally distinct isoforms for ~ 5% of the curated genes. If we relax our criteria for inclusion to include weaker forms of evidence, the fraction of genes with evidence of FDSIs remains low (~ 13%). We provide evidence that this picture will not change substantially with further curation and conclude there is a large gap between the presumed impact of splicing on gene function and the experimental evidence. Furthermore, many functionally distinct isoforms were not traceable to a specific isoform in Ensembl, a database that forms the basis for much computational research. CONCLUSIONS We conclude that the claim that alternative splicing vastly increases the functional repertoire of the genome is an extrapolation from a limited number of empirically supported cases. We also conclude that more work is needed to integrate experimental evidence and genome annotation databases. Our work should help shape research around the role of splicing on gene function from presuming large general effects to acknowledging the need for stronger experimental evidence.
Collapse
Affiliation(s)
- Shamsuddin A. Bhuiyan
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4 Canada
- Department of Psychiatry, University of British Columbia, Vancouver, BC V6T 1Z4 Canada
- Graduate Program in Bioinformatics, University of British Columbia, Vancouver, Canada
| | - Sophia Ly
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4 Canada
| | - Minh Phan
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4 Canada
| | - Brandon Huntington
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4 Canada
| | - Ellie Hogan
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4 Canada
| | - Chao Chun Liu
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4 Canada
| | - James Liu
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4 Canada
| | - Paul Pavlidis
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4 Canada
- Department of Psychiatry, University of British Columbia, Vancouver, BC V6T 1Z4 Canada
| |
Collapse
|
46
|
Bongiovanni L, Caposano F, Romanucci M, Grieco V, Malatesta D, Brachelente C, Massimini M, Benazzi C, Thomas RE, Salda LD. Survivin and Sox9: Potential Stem Cell Markers in Canine Normal, Hyperplastic, and Neoplastic Canine Prostate. Vet Pathol 2018; 56:200-207. [PMID: 30131013 DOI: 10.1177/0300985818794161] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Canine prostatic carcinoma is a relevant model for human prostatic carcinoma. Survivin is proposed as a biomarker of malignancy in human prostatic cancer. Sox9 is a stem cell marker required for prostate development and expressed in several adult tissues. The aims of the present study were to evaluate the patterns and expression levels of 2 putative stem cell markers, survivin and Sox9, in canine benign prostatic hyperplasia (BPH) and prostatic carcinoma to investigate their potential as stem cell markers. Immunohistochemistry with specific antibodies was performed on 3 samples of normal prostate gland, 18 samples of canine BPH, and 16 samples of prostatic carcinoma. The basal cell layer of normal and hyperplastic prostatic lobules had nuclear Sox9 immunolabeling and nuclear and rarely cytoplasmic survivin immunostaining, identifying them as potential stem cell markers. Significantly more frequent survivin and Sox9 expression (≥10% of nuclei) was observed in prostatic carcinoma as compared with BPH. The potential coexpression of survivin with Sox9, androgen receptor, and p63 was also investigated in selected BPH and prostatic carcinoma cases with immunofluorescence, and a partial colocalization was observed. Results indicate that Sox9 and survivin could be considered markers of stemness in canine prostate cells. Given its role in proliferation, cells in the basal cell layer with nuclear survivin expression are likely to be transit-amplifying cells that maintain some stem cell proprieties.
Collapse
Affiliation(s)
- Laura Bongiovanni
- 1 Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy.,2 Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | | | | | - Valeria Grieco
- 3 Department of Veterinary Science and Public Health, University of Milan, Milan, Italy
| | - Daniela Malatesta
- 1 Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy
| | - Chiara Brachelente
- 4 Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | | | - Cinzia Benazzi
- 5 Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Rachel E Thomas
- 2 Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | | |
Collapse
|
47
|
Srivastava K, Pickard A, Craig SG, Quinn GP, Lambe SM, James JA, McDade SS, McCance DJ. ΔNp63γ/SRC/Slug Signaling Axis Promotes Epithelial-to-Mesenchymal Transition in Squamous Cancers. Clin Cancer Res 2018; 24:3917-3927. [PMID: 29739791 PMCID: PMC6098695 DOI: 10.1158/1078-0432.ccr-17-3775] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 03/28/2018] [Accepted: 05/03/2018] [Indexed: 02/07/2023]
Abstract
Purpose: To investigate the regulation of epithelial-to-mesenchymal transition (EMT) in head and neck squamous cell carcinoma (HNSCC) and its importance in tumor invasion.Experimental Design: We use a three-dimensional invasive organotypic raft culture model of human foreskin keratinocytes expressing the E6/E7 genes of the human papilloma virus-16, coupled with bioinformatic and IHC analysis of patient samples to investigate the role played by EMT in invasion and identify effectors and upstream regulatory pathways.Results: We identify SNAI2 (Slug) as a critical effector of EMT-activated downstream of TP63 overexpression in HNSCC. Splice-form-specific depletion and rescue experiments further identify the ΔNp63γ isoform as both necessary and sufficient to activate the SRC signaling axis and SNAI2-mediated EMT and invasion. Moreover, elevated SRC levels are associated with poor outcome in patients with HNSCC in The Cancer Genome Atlas dataset. Importantly, the effects on EMT and invasions and SNAI2 expression can be reversed by genetic or pharmacologic inhibition of SRC.Conclusions: Overexpression of ΔNp63γ modulates cell invasion by inducing targetable SRC-Slug-evoked EMT in HNSCC, which can be reversed by inhibitors of the SRC signaling. Clin Cancer Res; 24(16); 3917-27. ©2018 AACR.
Collapse
Affiliation(s)
- Kirtiman Srivastava
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, United Kingdom.
| | - Adam Pickard
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, United Kingdom
- The Wellcome Trust Centre for Cell Matrix Research, University of Manchester, Manchester, United Kingdom
| | - Stephanie G Craig
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, United Kingdom
| | - Gerard P Quinn
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, United Kingdom
| | - Shauna M Lambe
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, United Kingdom
| | - Jacqueline A James
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, United Kingdom
| | - Simon S McDade
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, United Kingdom.
| | - Dennis J McCance
- Department of Pathology, School of Medicine, University of New Mexico, Albuquerque, New Mexico.
| |
Collapse
|
48
|
Chen Y, Li Y, Peng Y, Zheng X, Fan S, Yi Y, Zeng P, Chen H, Kang H, Zhang Y, Xiao ZX, Li C. ΔNp63α down-regulates c-Myc modulator MM1 via E3 ligase HERC3 in the regulation of cell senescence. Cell Death Differ 2018; 25:2118-2129. [PMID: 29880857 DOI: 10.1038/s41418-018-0132-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 04/20/2018] [Accepted: 05/08/2018] [Indexed: 12/31/2022] Open
Abstract
p63 and c-Myc are key transcription factors controlling genes involved in the cell cycle and cellular senescence. We previously reported that p63α can destabilize MM1 protein to derepress c-Myc, resulting in cell cycle progress and tumorigenesis. However, how the proteasomal degradation of MM1 is facilitated remains unclear. In the present study, we identified a novel E3 ligase, HERC3, which can mediate ubiquitination of MM1 and promote its proteasome-dependent degradation. We found that ΔNp63α transcriptionally up-regulates HERC3 and knockdown of HERC3 abrogates ΔNp63α-induced down-regulation of MM1. Either overexpression of MM1 or ablation of HERC3 induces cell senescence, while knockdown of MM1 rescues cell senescence induced by deficiency of either ΔNp63α or HERC3, implicating the involvement of the ΔNp63α/HERC3/MM1/c-Myc axis in the modulation of cell senescence. Additionally, our Oncomine analysis indicates activation of the ΔNp63α/HERC3/MM1/c-Myc axis in invasive breast carcinoma. Together, our data illuminate a novel axis regulating cell senescence: ΔNp63α stimulates transcription of E3 ligase HERC3, which mediates ubiquitination of c-Myc modulator MM1 and targets it to proteasomal degradation; subsequently, c-Myc is derepressed by ΔNp63α, thereby cell senescence is modulated by this axis. Our work provides a new interpretation of crosstalk between p63 and c-Myc, and also sheds new light on ΔNp63α-controlled cell senescence and tumorigenesis.
Collapse
Affiliation(s)
- Yonglong Chen
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, Sichuan, China
| | - Yimin Li
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, Sichuan, China
| | - Yougong Peng
- Department of General Surgery, The Second People's Hospital of Jingmen, Jingmen, 448000, Hubei, China
| | - Xuan Zheng
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, Sichuan, China
| | - Shijie Fan
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, Sichuan, China
| | - Yong Yi
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, Sichuan, China
| | - Peng Zeng
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, Sichuan, China
| | - Hu Chen
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, Sichuan, China
| | - Han Kang
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, Sichuan, China
| | - Yujun Zhang
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, Sichuan, China
| | - Zhi-Xiong Xiao
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, Sichuan, China
| | - Chenghua Li
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, Sichuan, China.
| |
Collapse
|
49
|
He H, Peng Y, Fan S, Chen Y, Zheng X, Li C. Cullin3/KCTD5 induces monoubiquitination of ΔNp63α and impairs its activity. FEBS Lett 2018; 592:2334-2340. [PMID: 29782646 DOI: 10.1002/1873-3468.13104] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 04/26/2018] [Accepted: 05/09/2018] [Indexed: 11/06/2022]
Abstract
Potassium channel tetramerization domain containing 5 (KCTD5) was previously documented as a component of the Cullin3-RING ligase (CRL3). It has been reported that KCTD5 can induce enrichment of polyubiquitinated proteins, and KCTD5-based CRL3 destabilizes several proteins. In our present study, we report that KCTD5 may physically interact with ΔNp63α, which is a member of the p53 family. Our further investigation revealed that Cullin3/KCTD5 can induce monoubiquitination of ΔNp63α. Cullin3/KCTD5 downregulates the DNA-binding affinity of ΔNp63α, impairing either its transactivity or its transinhibitory activity. Functionally, Cullin3/KCTD5 abates the proproliferation activity of ΔNp63α. These findings suggest that KCTD5-based CRL3 may mediate monoubiquitination and is a novel regulator of ΔNp63α.
Collapse
Affiliation(s)
- Hanbing He
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yougong Peng
- Department of General Surgery, The Second People's Hospital of Jingmen, China
| | - Shijie Fan
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yonglong Chen
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Xuan Zheng
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Chenghua Li
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| |
Collapse
|
50
|
Panatta E, Lena AM, Mancini M, Affinati M, Smirnov A, Annicchiarico-Petruzzelli M, Piro MC, Campione E, Bianchi L, Mazzanti C, Melino G, Candi E. Kruppel-like factor 4 regulates keratinocyte senescence. Biochem Biophys Res Commun 2018; 499:389-395. [DOI: 10.1016/j.bbrc.2018.03.172] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 03/22/2018] [Indexed: 01/07/2023]
|