1
|
Di Girolamo D, Di Iorio E, Missero C. Molecular and Cellular Function of p63 in Skin Development and Genetic Diseases. J Invest Dermatol 2025; 145:766-779. [PMID: 39340489 DOI: 10.1016/j.jid.2024.08.011] [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: 05/07/2024] [Revised: 08/02/2024] [Accepted: 08/16/2024] [Indexed: 09/30/2024]
Abstract
The transcription factor p63 is a master regulator of multiple ectodermal derivatives. During epidermal commitment, p63 interacts with several chromatin remodeling complexes to transactivate epidermal-specific genes and repress transcription of simple epithelial and nonepithelial genes. In the postnatal epidermis, p63 is required to control the proliferative potential of progenitor cells, maintain epidermal integrity, and contribute to epidermal differentiation. Autosomal dominant sequence variant in p63 cause a spectrum of syndromic disorders that affect several tissues, including or derived from stratified epithelia. In this review, we describe the recent studies that have provided novel insights into disease pathogenesis and potential therapeutic targets.
Collapse
Affiliation(s)
- Daniela Di Girolamo
- CEINGE Biotecnologie Avanzate Franco Salvatore, Naples, Italy; Biology Department, University of Naples Federico II, Naples, Italy
| | - Enzo Di Iorio
- Clinical Genetics Unit, University Hospital of Padua, Padua, Italy; Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Caterina Missero
- CEINGE Biotecnologie Avanzate Franco Salvatore, Naples, Italy; Biology Department, University of Naples Federico II, Naples, Italy.
| |
Collapse
|
2
|
Lopez-Pajares V, Bhaduri A, Zhao Y, Gowrishankar G, Donohue LKH, Guo MG, Siprashvili Z, Miao W, Nguyen DT, Yang X, Li AM, Tung ASH, Shanderson RL, Winge MCG, Meservey LM, Srinivasan S, Meyers RM, Guerrero A, Ji AL, Garcia OS, Tao S, Gambhir SS, Long JZ, Ye J, Khavari PA. Glucose modulates IRF6 transcription factor dimerization to enable epidermal differentiation. Cell Stem Cell 2025:S1934-5909(25)00088-8. [PMID: 40120584 DOI: 10.1016/j.stem.2025.02.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 12/18/2024] [Accepted: 02/28/2025] [Indexed: 03/25/2025]
Abstract
Non-energetic roles for glucose are largely unclear, as is the interplay between transcription factors (TFs) and ubiquitous biomolecules. Metabolomic analyses uncovered elevation of intracellular glucose during differentiation of diverse cell types. Human and mouse tissue engineered with glucose sensors detected a glucose gradient that peaked in the outermost differentiated layers of the epidermis. Free glucose accumulation was essential for epidermal differentiation and required the SGLT1 glucose transporter. Glucose affinity chromatography uncovered glucose binding to diverse regulatory proteins, including the IRF6 TF. Direct glucose binding enabled IRF6 dimerization, DNA binding, genomic localization, and induction of IRF6 target genes, including essential pro-differentiation TFs GRHL1, GRHL3, HOPX, and PRDM1. These data identify a role for glucose as a gradient morphogen that modulates protein multimerization in cellular differentiation.
Collapse
Affiliation(s)
- Vanessa Lopez-Pajares
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Aparna Bhaduri
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Program in Cancer Biology, Stanford University, Stanford, CA 94305, USA
| | - Yang Zhao
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Gayatri Gowrishankar
- Departments of Bioengineering and Radiology, Stanford University, Stanford, CA 94305, USA; Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA
| | - Laura K H Donohue
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Margaret G Guo
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Program in Biomedical Informatics, Stanford University, Stanford, CA 94305, USA
| | - Zurab Siprashvili
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Weili Miao
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Duy T Nguyen
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xue Yang
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Program in Cancer Biology, Stanford University, Stanford, CA 94305, USA
| | - Albert M Li
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Alan Sheng-Hwa Tung
- Department of Pathology, Stanford University, Stanford, CA 94350, USA; Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA
| | - Ronald L Shanderson
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Program in Cancer Biology, Stanford University, Stanford, CA 94305, USA
| | - Marten C G Winge
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lindsey M Meservey
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Suhas Srinivasan
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Robin M Meyers
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Angela Guerrero
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Andrew L Ji
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Omar S Garcia
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Shiying Tao
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sanjiv S Gambhir
- Departments of Bioengineering and Radiology, Stanford University, Stanford, CA 94305, USA; Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA
| | - Jonathan Z Long
- Department of Pathology, Stanford University, Stanford, CA 94350, USA; Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA
| | - Jiangbin Ye
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Paul A Khavari
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Program in Cancer Biology, Stanford University, Stanford, CA 94305, USA; Veterans Affairs Palo Alto Healthcare System, Palo Alto, CA 94304, USA.
| |
Collapse
|
3
|
Ma B, Yang G, Yao J, Wu C, Vega JP, Manske G, Hammoud SS, Sinha S, Singh A, Zhao H, Chen X. Asymmetric Histone Inheritance Regulates Differential Transcription Re-initiation and Cell Fate Decisions in Mouse Olfactory Horizontal Basal Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.02.641101. [PMID: 40093102 PMCID: PMC11908234 DOI: 10.1101/2025.03.02.641101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
To understand epigenetic inheritance in mammals, we investigate cell division modes and histone inheritance patterns in the mouse olfactory epithelium using an injury-induced regeneration model. Horizontal basal cells (HBCs), the adult stem cells in this tissue, undergo asymmetric division, coinciding with asymmetric histone H4 inheritance in vivo. Primary HBCs recapitulate both asymmetric cell division and asymmetric histone inheritance for H4, H3, and H3.3, but not H2A-H2B. Upon mitotic exit, asymmetric histone inheritance correlates with differential enrichment of a key 'stemness' transcription factor p63 and asynchronous transcription re-initiation. Single-cell RNA sequencing of paired daughter cells reveals their asymmetric cell fate priming in this multilineage stem cell system. Furthermore, disruption of asymmetric cell division abolishes these asymmetric cellular features, impairing olfactory epithelium regeneration and smell behavior in mice. Together, these findings reveal asymmetric histone inheritance in a mammalian adult stem cell lineage and highlight its biological significance in tissue regeneration and animal behavior.
Collapse
Affiliation(s)
- Binbin Ma
- Howard Hughes Medical Institute, Department of Biology, Johns Hopkins University, Baltimore, MD
- Department of Biology, Johns Hopkins University, Baltimore, MD
| | - Guanghui Yang
- Howard Hughes Medical Institute, Department of Biology, Johns Hopkins University, Baltimore, MD
- Department of Biology, Johns Hopkins University, Baltimore, MD
| | - Jonathan Yao
- Department of Biology, Johns Hopkins University, Baltimore, MD
| | - Charles Wu
- Department of Biology, Johns Hopkins University, Baltimore, MD
| | | | - Gabriel Manske
- Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, MI
- Department of Human Genetics, University of Michigan, Ann Arbor, MI
| | | | - Satrajit Sinha
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, SUNY at Buffalo, 955 Main St, Buffalo, NY
| | - Abhyudai Singh
- Electrical & Computer Engineering, University of Delaware, Newark, DE
| | - Haiqing Zhao
- Department of Biology, Johns Hopkins University, Baltimore, MD
| | - Xin Chen
- Howard Hughes Medical Institute, Department of Biology, Johns Hopkins University, Baltimore, MD
- Department of Biology, Johns Hopkins University, Baltimore, MD
| |
Collapse
|
4
|
Shiraishi M, Sowa Y, Sunaga A, Yamamoto K, Okazaki M. Bioengineering strategies for regeneration of skin integrity: A literature review. Regen Ther 2025; 28:153-160. [PMID: 39790492 PMCID: PMC11713503 DOI: 10.1016/j.reth.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 11/21/2024] [Accepted: 12/04/2024] [Indexed: 01/12/2025] Open
Abstract
Objective The skin is a complex organ that includes various stem cell populations. Current approaches for non-healing skin defects are sometimes inadequate and many attempts have been made to regenerate skin integrity. The aim of this review is to bridge the gap between basic research and clinical application of skin integrity regeneration. Methods A literature search was carried out in PubMed using combinations of the keywords "skin integrity", "tissue-engineered skin", "bioengineered skin", and "skin regeneration". Articles published from 1968 to 2023 reporting evidence from in vivo and in vitro skin regeneration experiments were included. Results These articles showed that stem cells can be differentiated into normal skin cells, including keratinocytes, and are a significant source of skin organoids, which are useful for investigating skin biology; and that emerging direct reprogramming methods have great potential to regenerate skin from the wounded skin surface. Conclusion Recent advances in skin regeneration will facilitate further advancement of both basic and clinical research in skin biology.
Collapse
Affiliation(s)
- Makoto Shiraishi
- Department of Plastic and Reconstructive Surgery, The University of Tokyo Hospital, Tokyo, Japan
| | - Yoshihiro Sowa
- Department of Plastic Surgery, Jichi Medical University, Japan
| | - Ataru Sunaga
- Department of Plastic Surgery, Jichi Medical University, Japan
| | - Kenta Yamamoto
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Mutsumi Okazaki
- Department of Plastic and Reconstructive Surgery, The University of Tokyo Hospital, Tokyo, Japan
| |
Collapse
|
5
|
Chen X, Sun W, Kong X, Ming X, Zhang Y, Yan W, Mohibi S, Chen M, Mitchell K, Zhang J. TAp63γ is the primary isoform of TP63 for tumor suppression but not development. Cell Death Discov 2025; 11:51. [PMID: 39915463 PMCID: PMC11802870 DOI: 10.1038/s41420-025-02326-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/19/2024] [Accepted: 01/24/2025] [Indexed: 02/09/2025] Open
Abstract
TP63 is expressed as TAp63 and ΔNp63 from the P1 and P2 promoters, respectively. While TAp63 and ΔNp63 are expressed as three TAp63α/β/γ and ΔNp63α/β/γ due to alternative splicing, only p63α (TA and ΔN) and p63γ (TA and ΔN) proteins are found to be detectable and likely to be responsible for p63-dependent activity. Previous studies implied and/or demonstrated that TAp63α, which contains an N-terminal activation domain conserved in p53, functions as a tumor suppressor by regulating an array of genes for growth suppression. By contrast, ΔNp63α, which also contains an N-terminal activation domain but is different from that in TAp63, regulates a unique set of genes and functions as a master regulator for development of epidermis and other stratified epithelial tissues. However, the biological function of p63γ is largely unexplored. To explore this, we generated a mouse model in that exon 10', a coding exon specific for p63γ, was deleted by CRISPR-cas9. We showed that mice deficient in p63γ are viable and futile, which is different from mice deficient in total TP63 or p63α. Like TAp63-deficient mice, p63γ-deficient mice have a short lifespan and are prone to spontanenous tumors. Additionally, loss of p63γ shortens the lifespan of tumor-free mice potentially via increased cellular senescence. Moreover, mice deficient in p63γ are prone to chronic inflammation in multiple organs and liver steatosis potentially via altered lipid metabolism. Single-cell RNA-seq revealed that loss of p63γ increases the expression of SCD1, a rate-limiting enzyme for synthesis of monounsaturated fatty acids, leading to altered lipid homeostasis. Together, our data indicate that TP63γ is the primary isoform of TP63 for tumor suppression but not development by maintaining normal inflammatory response and lipid homeostasis.
Collapse
Affiliation(s)
- Xinbin Chen
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, Davis, USA
| | - Wenqiang Sun
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, Davis, USA
- Department of Animal Science and Technology, Sichuan Agricultural University, Ya'an, China
| | - Xiangmudong Kong
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, Davis, USA
| | - Xin Ming
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, Davis, USA
| | - Yanhong Zhang
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, Davis, USA
| | - Wensheng Yan
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, Davis, USA
| | - Shakur Mohibi
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, Davis, USA
| | - Mingyi Chen
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, 75390, USA
| | - Keith Mitchell
- Department of Physiology and Membrane Biology, University of California Davis School of Medicine, Davis, Davis, CA, USA
| | - Jin Zhang
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, University of California, Davis, USA.
| |
Collapse
|
6
|
Lv K, Li Q, Jiang N, Chen Q. Role of TRIM29 in disease: What is and is not known. Int Immunopharmacol 2025; 147:113983. [PMID: 39755113 DOI: 10.1016/j.intimp.2024.113983] [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: 08/10/2024] [Revised: 12/17/2024] [Accepted: 12/28/2024] [Indexed: 01/06/2025]
Abstract
Tripartite motif-containing proteins (TRIMs), comprising the greatest subfamily of E3 ubiquitin ligases with approximately 80 members of this family, are widely distributed in mammalian cells. TRIMs actively participate in ubiquitination of target proteins, a type of post-translational modification associated with protein degradation and other functions. Tripartite motif-containing protein 29 (TRIM29), a member of the TRIM family, differs from other members of this family in that it lacks the RING finger structural domain containing cysteine and histidine residues that mediates DNA binding, protein-protein interactions, and ubiquitin ligase, at its N-terminus. The expression of TRIM29 was initially found to be associated with cancer and diabetic nephropathy progression, and antiviral immunity which is triggered by virus-derived nucleic acids binding to pattern recognition receptors (PRRs) on immune cells. Recently, TRIM29 has also been explored as a diagnostic biomarker and therapeutic target for some immune-related diseases. Here, we review the functions of TRIM29 in the progression of diseases and the inherent mechanisms, as well as the remaining gaps in the literature. A thorough understanding of the detailed regulatory mechanisms of TRIM29 will ultimately facilitate the development of different therapeutic strategies for various diseases.
Collapse
Affiliation(s)
- Kunying Lv
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang 110866, China
| | - Qilong Li
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang 110866, China
| | - Ning Jiang
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang 110866, China
| | - Qijun Chen
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, 120 Dongling Road, Shenyang 110866, China; The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang 110866, China.
| |
Collapse
|
7
|
Zhuang J, Li Y, Chen Y, Zhang H, Liu S, Hu M, Chen C. Molecular characterization of a rare TP63 variant associated with split-hand/split-foot malformation 4 and incomplete penetrance: disruption of the p63-Dlx signaling pathway. BMC Genomics 2025; 26:113. [PMID: 39910461 PMCID: PMC11800522 DOI: 10.1186/s12864-025-11297-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 01/28/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Split-hand/foot malformation (SHFM) is a congenital disability characterized by the absence or hypoplasia of the central ray of the hands and/or feet. This study reports a causative variant in the TP63 gene in a Chinese family exhibiting limb anomalies associated with SHFM4. METHODS Enrolled in this study was a Chinese family with limb anomalies without any other clinical features. Karyotype analysis and chromosomal microarray analysis (CMA) were conducted to identify chromosomal abnormalities. Whole exome sequencing (WES) was utilized to investigate sequence variants, while RNA sequencing assessed differentially expressed genes, with findings confirmed through quantitative PCR (qPCR). RESULTS Karyotype analysis and CMA revealed no chromosomal abnormalities in the family. Subsequently, WES identified a rare heterozygous variant of NM_003722.5: c.956G > A (p.Arg319His) in the TP63 gene in the proband, which was inherited from her father who also presented with limb deformities. However, both of the sister and grandfather of the proband had the same variant but exhibited normal limb morphology. RNA sequencing results demonstrated an increased expression level of TP63 and its downstream genes (PERP, CDH3, and DLX5) compared with the controls, indicating an enrichment of cell adhesion molecules the differentially expressed genes in the patient. However, significant differences were noted only for the CDH3 and DLX5 genes in qPCR analysis (p<0.05). CONCLUSION This study identifies, for the first time, the TP63 gene variant c.956G > A (p.Arg319His) as a causative factor for SHFM4 in Chinese individuals with incomplete penetrance. In addition, we hypothesize that the p.Arg319His variant functions as a gain-of-function variant, leading to the upregulation of cell adhesion target genes. Such upregulation then disrupts the p63-Dlx signaling pathway and causes AER stratification failure.
Collapse
Affiliation(s)
- Jianlong Zhuang
- Prenatal Diagnosis Center, Quanzhou Women's and Children's Hospital, Quanzhou, 362000, Fujian Province, China.
| | - Yanqing Li
- Prenatal Diagnosis Center, Quanzhou Women's and Children's Hospital, Quanzhou, 362000, Fujian Province, China
| | - Yu'e Chen
- Department of Ultrasound, Quanzhou Women's and Children's Hospital, Quanzhou, 362000, Fujian Province, China
| | - Hegan Zhang
- Department of Gynecology, Quanzhou Women's and Children's Hospital, Quanzhou, 362000, Fujian Province, China
| | - Shufen Liu
- Department of Neurology, Rare Disease Medical Center, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, Fujian Province, China
| | - Manman Hu
- Berry Genomics Corporation, Beijing, 102200, China
| | - Chunnuan Chen
- Department of Neurology, Rare Disease Medical Center, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, Fujian Province, China.
| |
Collapse
|
8
|
Manoukian P, Kuhnen LC, van Laarhoven HWM, Bijlsma MF. Association of epigenetic landscapes with heterogeneity and plasticity in pancreatic cancer. Crit Rev Oncol Hematol 2025; 206:104573. [PMID: 39581245 DOI: 10.1016/j.critrevonc.2024.104573] [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: 07/22/2024] [Revised: 11/13/2024] [Accepted: 11/17/2024] [Indexed: 11/26/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis. Due to a lack of clear symptoms, patients often present with advanced disease, with limited clinical intervention options. The high mortality rate of PDAC is, however, also a result of several other factors that include a high degree of heterogeneity and treatment resistant cellular phenotypes. Molecular subtypes of PDAC have been identified that are thought to represent cellular phenotypes at the tissue level. The epigenetic landscape is an important factor that dictates these subtypes. Permissive epigenetic landscapes serve as drivers of molecular heterogeneity and cellular plasticity in developing crypts as well as metaplastic lesions. Drawing parallels with other cancers, we hypothesize that epigenetic permissiveness is a potential driver of cellular plasticity in PDAC. In this review will explore the epigenetic alterations that underlie PDAC cell states and relate them to cellular plasticity from other contexts. In doing so, we aim to highlight epigenomic drivers of PDAC heterogeneity and plasticity and, with that, offer some insight to guide pre-clinical research.
Collapse
Affiliation(s)
- Paul Manoukian
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Amsterdam, the Netherlands; Cancer Center Amsterdam, Cancer Biology, Amsterdam, the Netherlands.
| | - Leo C Kuhnen
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Amsterdam, the Netherlands
| | - Hanneke W M van Laarhoven
- Cancer Center Amsterdam, Cancer Biology, Amsterdam, the Netherlands; Amsterdam UMC location University of Amsterdam, Department of Medical Oncology, Amsterdam, the Netherlands
| | - Maarten F Bijlsma
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Amsterdam, the Netherlands; Cancer Center Amsterdam, Cancer Biology, Amsterdam, the Netherlands
| |
Collapse
|
9
|
Liu Z, Zheng Q, Li Z, Huang M, Zhong C, Yu R, Jiang R, Dai H, Zhang J, Gu X, Xu Y, Li C, Shan S, Xu F, Hong Y, Ren T. Epithelial stem cells from human small bronchi offer a potential for therapy of idiopathic pulmonary fibrosis. EBioMedicine 2025; 112:105538. [PMID: 39753035 PMCID: PMC11754162 DOI: 10.1016/j.ebiom.2024.105538] [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: 08/27/2024] [Revised: 11/21/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a fibrosing interstitial pneumonia with restrictive ventilation. Recently, the structural and functional defects of small airways have received attention in the early pathogenesis of IPF. This study aimed to elucidate the characteristics of small airway epithelial dysfunction in patients with IPF and explore novel therapeutic interventions to impede IPF progression by targeting the dysfunctional small airways. METHODS Airway trees spanning the proximal-distal axis were harvested from control lungs and explanted lungs with end-stage IPF undergoing transplant. Qualified basal cells (BCs, p63/Krt5/ITGA6/NGFR) were expanded, and their cellular functions, feasibility, safety and efficacy for transplantation therapy in IPF were validated with experiments in vitro and mouse model. Single-cell RNA-sequencing was employed to elucidate the underlying mechanisms governing the BCs based therapy. Based upon these evidences, three patients with advanced IPF and small airway dysfunction received autologous-BCs transplantation. Post-transplantation assessments included lung function, exercise capacity and high resolution computed tomography (HRCT) scans were analyzed to quantify the clinical benefits conferred by the BCs transplantation. FINDINGS An overall landscape of senescent phenotype in airway epithelial cells and airway stem/progenitor cells along the proximal-distal axis of the airway tree in IPF were outlined. In contrast to the cells situated in distal airways, BCs located in small bronchi in IPF displayed a non-senescent phenotype, with comparable proliferative, differentiative capabilities, and similar transcriptomic profiles to normal controls. In a mouse model of pulmonary fibrosis, BCs exhibited promising protective efficacy and safety for transplantation therapy. Autologous BCs transplantation in three advanced IPF patients with small airway dysfunction yielded significant clinical improvements in pulmonary function, particularly evidence in lung volume and small airway function. INTERPRETATION Epithelia of small bronchi in IPF contain functional and expandable basal stem cells, which exert therapeutic benefits via bronchoscopic implantation. Our findings offer a potential for IPF treatment by targeting small airways. FUNDING National Natural Science Foundation of China (82430001, 81930001, and 81900059), Shanghai Shenkang Hospital Development Center (SHDC2020CR3063B), Department of Science and Technology of Shandong Province (2024HWYQ-058).
Collapse
Affiliation(s)
- Zeyu Liu
- Department of Respiratory and Clinical Care Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Qi Zheng
- Department of Respiratory and Clinical Care Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Zhoubin Li
- Department of Lung Transplantation and Thoracic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, PR China
| | - Moli Huang
- Department of Bioinformatics, School of Biological and Basic Medical Sciences, Soochow University, Suzhou, 215123, China
| | - Cheng Zhong
- Department of Respiratory and Clinical Care Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Ruize Yu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China; Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, 250021, Shandong, China
| | - Rong Jiang
- Department of Bioinformatics, School of Biological and Basic Medical Sciences, Soochow University, Suzhou, 215123, China
| | - Haotian Dai
- Department of Respiratory and Clinical Care Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Jingyuan Zhang
- Department of Respiratory and Clinical Care Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Xiaohua Gu
- Department of Respiratory and Clinical Care Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yongle Xu
- Department of Respiratory and Clinical Care Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Chunwei Li
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Shan Shan
- Department of Respiratory and Clinical Care Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Feng Xu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China; Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, 250021, Shandong, China.
| | - Yue Hong
- School of Life and Health Sciences, Hainan University, Haikou, Hainan 570228, China; Hainan Province Key Laboratory of One Health, Collaborative Innovation Center of One Health, Hainan University, Haikou, Hainan 570228, China.
| | - Tao Ren
- Department of Respiratory and Clinical Care Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| |
Collapse
|
10
|
Boncimino F, D'Auria L, Todorova K, van der Zanden SY, Neefjes J, Mandinova A, Missero C, Sol S. Anthracyclines disaggregate and restore mutant p63 function: a potential therapeutic approach for AEC syndrome. Cell Death Discov 2025; 11:24. [PMID: 39863572 PMCID: PMC11762975 DOI: 10.1038/s41420-025-02307-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/24/2024] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
Ankyloblepharon-Ectodermal Defects-Cleft Lip/Palate (AEC) syndrome is a rare genetic disorder caused by mutations in the TP63 gene, which encodes a transcription factor essential for epidermal gene expression. A key feature of AEC syndrome is chronic skin erosion, for which no effective treatment currently exists. Our previous studies demonstrated that mutations associated with AEC syndrome lead to p63 protein misfolding and aggregation, exerting a dominant-negative effect. By performing a high-throughput screening of epigenetic and FDA-approved compounds in a co-transfection model of wild-type and mutant p63, we found that two compounds, Doxorubicin and Epirubicin, alleviate protein aggregation and restore p63 transactivation function. Moreover, treatment with these compounds reduced protein aggregation and restored the expression of keratinocyte-specific p63 target genes in primary keratinocytes derived from a conditional ΔNp63αL514F knock-in AEC mouse model, which mimics the ectodermal defects and skin erosions characteristic of AEC syndrome. A chemical analog of Doxorubicin, diMe-Doxorubicin, which exhibits lower tissue and organ toxicity, was also found to be effective in promoting the disaggregation of mutant p63 and rescuing its transcriptional activity. Our findings identify compounds that can partially resolve mutant p63 aggregation, increase its monomeric isoform, and reactivate its transcriptional function. These results suggest potential therapeutic efficacy for treating skin erosions in AEC syndrome.
Collapse
Affiliation(s)
- Fabiana Boncimino
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Ludovica D'Auria
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80145, Naples, Italy
- Department of Biology, University of Naples Federico II, 80126, Naples, Italy
| | - Kristina Todorova
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Sabina Y van der Zanden
- Department of Cell and Chemical Biology, ONCODE Institute, Leiden University Medical Center, 2333 ZC, Leiden, The Netherlands
| | - Jacques Neefjes
- Department of Cell and Chemical Biology, ONCODE Institute, Leiden University Medical Center, 2333 ZC, Leiden, The Netherlands
| | - Anna Mandinova
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA.
- Broad Institute of Harvard and MIT, 7 Cambridge Center, Cambridge, MA, 02142, USA.
- Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA, 02138, USA.
| | - Caterina Missero
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80145, Naples, Italy.
- Department of Biology, University of Naples Federico II, 80126, Naples, Italy.
| | - Stefano Sol
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA.
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80145, Naples, Italy.
| |
Collapse
|
11
|
Lotz R, Osterburg C, Chaikuad A, Weber S, Akutsu M, Machel AC, Beyer U, Gebel J, Löhr F, Knapp S, Dobbelstein M, Lu X, Dötsch V. Alternative splicing in the DBD linker region of p63 modulates binding to DNA and iASPP in vitro. Cell Death Dis 2025; 16:4. [PMID: 39762243 PMCID: PMC11704248 DOI: 10.1038/s41419-024-07320-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 11/28/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025]
Abstract
The transcription factor p63 is expressed in many different isoforms as a result of differential promoter use and splicing. Some of these isoforms have very specific physiological functions in the development and maintenance of epithelial tissues and surveillance of genetic integrity in oocytes. The ASPP family of proteins is involved in modulating the transcriptional activity of the p53 protein family members, including p63. In particular, iASPP plays an important role in the development and differentiation of epithelial tissues. Here we characterize the interaction of iASPP with p63 and show that it binds to the linker region between the DNA binding domain and the oligomerization domain. We further demonstrate that this binding site is removed in a splice variant of p63 where a stretch of five amino acids is replaced with a single alanine residue. This stretch contains a degenerate class II SH3 domain binding motif that is responsible for interaction with iASPP, as well as two positively charged amino acids. Moreover, the concomitant loss of the charged amino acids in the alternatively spliced version decreases the affinity of p63 to its cognate DNA element two- to threefold. mRNAs encoding full-length p63, as well as its alternatively spliced version, are present in all tissues that we investigated, albeit in differing ratios. We speculate that, through the formation of hetero-complexes of both isoforms, the affinity to DNA, as well as the interaction with iASPP, can be fine-tuned in a tissue-specific manner.
Collapse
Affiliation(s)
- Rebecca Lotz
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, 60438, Frankfurt, Germany
| | - Christian Osterburg
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, 60438, Frankfurt, Germany.
| | - Apirat Chaikuad
- Institute of Pharmaceutical Chemistry, Goethe University, 60438, Frankfurt, Germany
- Structural Genomics Consortium (SGC), Goethe University, 60438, Frankfurt, Germany
| | - Sabrina Weber
- Institute of Molecular Oncology, Center of Molecular Biosciences, University of Göttingen, Göttingen, Germany
| | - Masato Akutsu
- Buchmann Institute for Molecular Life Sciences, Goethe University, 60438, Frankfurt, Germany
- Research Center for Advanced Analysis, National Agriculture and Food Research Organization (NARO), Tsukuba, Ibaraki, Japan
| | - Anne Christin Machel
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, 60438, Frankfurt, Germany
| | - Ulrike Beyer
- Institute of Molecular Oncology, Center of Molecular Biosciences, University of Göttingen, Göttingen, Germany
| | - Jakob Gebel
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, 60438, Frankfurt, Germany
| | - Frank Löhr
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, 60438, Frankfurt, Germany
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry, Goethe University, 60438, Frankfurt, Germany
- Structural Genomics Consortium (SGC), Goethe University, 60438, Frankfurt, Germany
| | - Matthias Dobbelstein
- Institute of Molecular Oncology, Center of Molecular Biosciences, University of Göttingen, Göttingen, Germany
| | - Xin Lu
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7DQ, UK
| | - Volker Dötsch
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance, Goethe University, 60438, Frankfurt, Germany.
| |
Collapse
|
12
|
Genna VG, Maurizi E, Rama P, Pellegrini G. Biology and medicine on ocular surface restoration: Advancements and limits of limbal stem cell deficiency treatments. Ocul Surf 2025; 35:57-67. [PMID: 39580144 DOI: 10.1016/j.jtos.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 11/08/2024] [Accepted: 11/13/2024] [Indexed: 11/25/2024]
Abstract
Ocular vision can be hampered by corneal damages, sensibly reducing patients' quality of life and having important social and economic consequences. Ocular surface diseases, which often lead to corneal opacities with visual impairment are the most severe forms of the Limbal Stem Cell Deficiency (LSCD). The present review provides an updated perspective on the available treatments for LSCD, focusing on clinical and biological features, as well as critical points to monitor during clinical translation. Recently developed surgical treatments for LSCD are described, along with their benefits and limitations, with the aim of addressing the issue of correct patient selection. Autologous surgical approaches have been attempted, such as conjunctival limbal autograft (CLAU), simple limbal epithelial transplantation (SLET), and others. Allogeneic limbal stem cell transplantation represents an alternative but carries risk of rejection and requires immunosuppression. Other potential treatments are based on induced pluripotent stem cells (iPSCs), but they require further investigation. The development of advanced therapy medicinal products (ATMPs) such as cultivated limbal epithelial transplantation (CLET), or the use of other epithelia as cultivated oral mucosal epithelial cell transplantation (COMET), has opened additional therapeutic possibilities. Some common critical issues in clinical translation are described, such as patient selection, biopsy procurement, or the use of human/animal derived components, which require rigorous validation to ensure safety and efficacy. Personalized medicine is a promising field for ocular surface restoration, where long-term follow-up studies and standardized criteria are crucial to evaluate the efficacy of these treatments and their cost-effectiveness in providing high-value healthcare.
Collapse
Affiliation(s)
| | - Eleonora Maurizi
- Centre for Regenerative Medicine ''S. Ferrari'', University of Modena and Reggio Emilia, Modena, Italy
| | - Paolo Rama
- Department of Ophthalmology, IRCCS Policlinico San Matteo, Pavia, Italy
| | - Graziella Pellegrini
- Centre for Regenerative Medicine ''S. Ferrari'', University of Modena and Reggio Emilia, Modena, Italy.
| |
Collapse
|
13
|
Hrabal V, Stenckova M, Zavadil Kokas F, Muller P, Nenutil R, Vojtesek B, Coates PJ. TAp73 and ΔTAp73 isoforms show cell-type specific distributions and alterations in cancer. Sci Rep 2024; 14:29949. [PMID: 39622910 PMCID: PMC11612387 DOI: 10.1038/s41598-024-80927-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/22/2024] [Indexed: 12/06/2024] Open
Abstract
TP73 is a member of the TP53 gene family and produces N- and C-terminal protein isoforms through alternative promoters, alternative translation initiation and alternative splicing. Most notably, p73 protein isoforms may either contain a p53-like transactivation domain (TAp73 isoforms) or lack this domain (ΔTAp73 isoforms) and these variants have opposing or independent functions. To date, there is a lack of well-characterised isoform-specific p73 antibodies. Here, we produced polyclonal and monoclonal antibodies to N-terminal p73 variants and the C-terminal p73α isoform, the most common variant in human tissues. These reagents show that TAp73 is a marker of multiciliated epithelial cells, while ΔTAp73 is a marker of non-proliferative basal/reserve cells in squamous epithelium. We were unable to detect ΔNp73 variant proteins, in keeping with recent data that this is a minor form in human tissues. Most cervical squamous cell carcinomas (79%) express p73α, and the distribution of staining in basal cells correlated with lower tumour grade. TAp73 was found in 17% of these tumours, with a random distribution and no association with clinicopathological features. These data indicate roles for ΔTAp73 in maintaining a non-proliferative state of undifferentiated squamous epithelial cells and for TAp73 in the production of differentiated multiciliated cells.
Collapse
Affiliation(s)
- Vaclav Hrabal
- Research Center for Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, Zluty kopec 7, Brno, 656 53, Czech Republic.
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic.
| | - Michaela Stenckova
- Research Center for Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, Zluty kopec 7, Brno, 656 53, Czech Republic
| | - Filip Zavadil Kokas
- Research Center for Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, Zluty kopec 7, Brno, 656 53, Czech Republic
| | - Petr Muller
- Research Center for Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, Zluty kopec 7, Brno, 656 53, Czech Republic
| | - Rudolf Nenutil
- Department of Pathology, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Borivoj Vojtesek
- Research Center for Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, Zluty kopec 7, Brno, 656 53, Czech Republic
| | - Philip J Coates
- Research Center for Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, Zluty kopec 7, Brno, 656 53, Czech Republic.
| |
Collapse
|
14
|
Mendes J, Guimarães AR, Ribeiro JM, Oliveiros B, Mesquita LA, Fernandes MH, do Vale FJF, Silva HC. Role of MTHFR, IRF6, PAX7 and TP63 SNPs in susceptibility to non-syndromic orofacial cleft, a candidate gene study in a Portuguese population. Orthod Craniofac Res 2024; 27:950-958. [PMID: 39049717 DOI: 10.1111/ocr.12838] [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: 02/26/2024] [Revised: 06/04/2024] [Accepted: 07/13/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Non-syndromic orofacial cleft (NSOC) is a complex phenotype, involving multiple genetic and environmental factors. Association studies exploring the genetic susceptibility to this prevalent oral malformation show variability of results in different populations. Using a candidate gene approach, we aimed to verify the role of four single-nucleotide polymorphisms (SNPs) in the susceptibility to NSOC in Portuguese patients. METHODS A total of 254 non-consanguineous individuals of Portuguese were recruited, including 120 patients with NSOC and 134 controls. About 92% of these patients had non-syndromic cleft lip with or without cleft palate (NSCL/P) and 8% had only non-syndromic cleft palate (NSCP). SNPs in the MTHFR (rs1801133), IRF6 (rs642961), PAX7 (rs742071) and TP63 (rs9332461) genes were studied, using a real-time approach with TaqMan probes. Allelic, genotypic, dominant, recessive and over-dominant models were explored using a chi-squared test. Adjusted p-value was calculated for multiple comparisons using the Benjamini-Hochberg false discovery rate (FDR). RESULTS All SNPs were in Hardy-Weinberg equilibrium. For MTHFR, IRF6, and PAX7 SNPs, no statistically significant difference was highlighted for any of the evaluated models. For TP63 SNP, data fitted an over-dominant model, with a protective effect for heterozygotes (OR 1.897; CI 95% [1.144-3.147]; p < .016, when comparing controls vs. cases), but significance was lost when applying adjusted p-value for multiple comparisons (4 × 5 tests). CONCLUSION In this Portuguese population, there was no evidence of an association between the evaluated SNPs and NSOC. For TP63 SNP, the possibility of a protective effect of heterozygotes should be further investigated.
Collapse
Affiliation(s)
- João Mendes
- Institute of Medical Genetics/UCGenomics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | | | - Joana Martins Ribeiro
- Institute of Medical Genetics/UCGenomics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Bárbara Oliveiros
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Luís Alcides Mesquita
- Institute of Medical Genetics/UCGenomics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | | | | | - Henriqueta Coimbra Silva
- Institute of Medical Genetics/UCGenomics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Center of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
15
|
Foffi E, Violante A, Pecorari R, Lena AM, Rugolo F, Melino G, Candi E. BRD4 sustains p63 transcriptional program in keratinocytes. Biol Direct 2024; 19:124. [PMID: 39605045 PMCID: PMC11600901 DOI: 10.1186/s13062-024-00547-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 10/10/2024] [Indexed: 11/29/2024] Open
Abstract
Here, we investigated the potential interaction between bromodomain-containing protein 4 (BRD4), an established epigenetic modulator and transcriptional coactivator, and p63, a member of the p53 transcription factor family, essential for epithelial development and skin homeostasis. Our protein-protein interaction assays demonstrated a strong and conserved physical interaction between BRD4 and the p53 family members-p63, p73, and p53-suggesting a shared binding region among these proteins. While the role of BRD4 in cancer development through its interaction with p53 has been explored, the effects of BRD4 and Bromodomain and Extra Terminal (BET) inhibitors in non-transformed cells, such as keratinocytes, remain largely unknown. Our functional analyses revealed changes in cellular proliferation and differentiation in keratinocytes depleted of either p63 or BRD4, which were further supported by using the BRD4 inhibitor JQ1. Transcriptomic analyses, chromatin immunoprecipitation, and RT-qPCR indicated a synergistic mechanism between p63 and BRD4 in regulating the transcription of keratinocyte-specific p63 target genes, including HK2, FOXM1, and EVPL. This study not only highlights the complex relationship between BRD4 and p53 family members but also suggests a role for BRD4 in maintaining keratinocyte functions. Our findings pave the way for further exploration of potential therapeutic applications of BRD4 inhibitors in treating skin disorders.
Collapse
Affiliation(s)
- E Foffi
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - A Violante
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - R Pecorari
- Istituto Dermopatico Dell'Immacolata, IDI-IRCCS, 00167, Rome, Italy
| | - A M Lena
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - F Rugolo
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - G Melino
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - E Candi
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", 00133, Rome, Italy.
- Istituto Dermopatico Dell'Immacolata, IDI-IRCCS, 00167, Rome, Italy.
| |
Collapse
|
16
|
Lv Z, Liu Z, Liu K, Lin X, Pu W, Li Y, Zhao H, Xi Y, Sui P, Vaughan AE, Gillich A, Zhou B. Alveolar regeneration by airway secretory-cell-derived p63 + progenitors. Cell Stem Cell 2024; 31:1685-1700.e6. [PMID: 39232560 DOI: 10.1016/j.stem.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 03/12/2024] [Accepted: 08/09/2024] [Indexed: 09/06/2024]
Abstract
Lung injury activates epithelial stem or progenitor cells for alveolar repair and regeneration. Unraveling the origin and fate of injury-induced progenitors is crucial for elucidating lung repair mechanisms. Here, we report that p63-expressing progenitors emerge upon bleomycin-induced mouse lung injury. Single-cell RNA sequencing and clonal analysis reveal that these p63+ progenitors proliferate rapidly and differentiate into alveolar type 1 and type 2 cells through different trajectories. Dual recombinase-mediated sequential genetic-lineage tracing demonstrates that p63+ progenitors originate from airway secretory cells and subsequently generate alveolar cells. Functionally, p63 activation is essential for efficient alveolar regeneration from secretory cells post injury. Our study identifies secretory-cell-derived p63+ progenitors as contributors to alveolar repair, suggesting a potential therapeutic avenue for lung regeneration following injury.
Collapse
Affiliation(s)
- Zan Lv
- New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Zixin Liu
- New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Kuo Liu
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Xiuyu Lin
- New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Wenjuan Pu
- New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Yan Li
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Huan Zhao
- New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Ying Xi
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Pengfei Sui
- New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Andrew E Vaughan
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Astrid Gillich
- Calico Life Sciences LLC, South San Francisco, CA 94080, USA
| | - Bin Zhou
- New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
17
|
Branch MC, Weber M, Li MY, Flora P, Ezhkova E. Overview of chromatin regulatory processes during surface ectodermal development and homeostasis. Dev Biol 2024; 515:30-45. [PMID: 38971398 PMCID: PMC11317222 DOI: 10.1016/j.ydbio.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 05/02/2024] [Accepted: 07/02/2024] [Indexed: 07/08/2024]
Abstract
The ectoderm is the outermost of the three germ layers of the early embryo that arise during gastrulation. Once the germ layers are established, the complex interplay of cellular proliferation, differentiation, and migration results in organogenesis. The ectoderm is the progenitor of both the surface ectoderm and the neural ectoderm. Notably, the surface ectoderm develops into the epidermis and its associated appendages, nails, external exocrine glands, olfactory epithelium, and the anterior pituitary. Specification, development, and homeostasis of these organs demand a tightly orchestrated gene expression program that is often dictated by epigenetic regulation. In this review, we discuss the recent discoveries that have highlighted the importance of chromatin regulatory mechanisms mediated by transcription factors, histone and DNA modifications that aid in the development of surface ectodermal organs and maintain their homeostasis post-development.
Collapse
Affiliation(s)
- Meagan C Branch
- Black Family Stem Cell Institute, Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Madison Weber
- Black Family Stem Cell Institute, Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Meng-Yen Li
- Black Family Stem Cell Institute, Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Pooja Flora
- Black Family Stem Cell Institute, Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Elena Ezhkova
- Black Family Stem Cell Institute, Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
18
|
Mutafchieva MZ, Draganova MN, Yaneva BK, Zagorchev PI, Tomov GT. Clinical Improvement and P63-Deficiency Correction in OLP Patients After Photobiomodulation. Dent J (Basel) 2024; 12:338. [PMID: 39590388 PMCID: PMC11593062 DOI: 10.3390/dj12110338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 09/27/2024] [Accepted: 10/10/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Oral lichen planus (OLP) is a chronic inflammatory disease associated with the formation of symptomatic lesions in the mouth. P63 is essential for epidermal development and regeneration. Weak expression of this protein has been shown in OLP lesions. Photobiomodulation (PBM) therapy has been reported to reduce OLP symptoms, but its ability to correct the molecular perturbations of the disease has not been studied. This study aimed to evaluate the efficacy of PBM in OLP treatment by evaluating changes in p63 expression and their association with clinical response. Methods: Twenty OLP patients underwent PBM with a diode laser (810 nm), (0.50 W, 30 s, 1.2 J/cm2), 3 times weekly for a month. The treatment efficacy index (EI) was calculated based on pain-level values and clinical scores of lesions before and after therapy. Biopsies were taken before and after therapy, analyzed immunohistochemically for p63 expression, and compared with 10 healthy controls. Results: P63 levels in OLP lesions were significantly lower than those in normal oral mucosa. After treatment, the pain level and clinical scores of the lesions decreased significantly. The calculated EI showed PBM effectiveness in 90% of cases. Increased p63 positivity and staining intensity were observed after therapy. Conclusions: The established p63 deficiency in OLP lesions is likely an important molecular mechanism in the pathogenesis of the disease. Laser irradiation at 810 nm increased p63 expression to a level close to that found in the healthy epithelium and significantly improved the symptoms and clinical signs of OLP. All of this determines the effectiveness of PBM therapy in the management of OLP.
Collapse
Affiliation(s)
- Maria Zaharieva Mutafchieva
- Department of Periodontology and Oral Mucosa Diseases, Faculty of Dental Medicine, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria;
| | - Milena Nenkova Draganova
- Department of Medical Biology, Faculty of Medicine, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria
- Research Institute, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria;
| | - Blagovesta Konstantinova Yaneva
- Department of Periodontology and Oral Mucosa Diseases, Faculty of Dental Medicine, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria;
| | - Plamen Ivanov Zagorchev
- Research Institute, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria;
- Department of Medical Physics and Biophysics, Faculty of Pharmacy, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria
| | - Georgi Tomchev Tomov
- Department of Healthcare and Social Work, New Bulgarian University, 1618 Sofia, Bulgaria;
| |
Collapse
|
19
|
Cho Y, Cao Z, Luo X, Tian JJ, Hukkanen RR, Hussien R, Cancilla B, Chowdhury P, Li F, Ma S, LaGory EL, Schroeder M, Dusenberry A, Marshall L, Hawkins J, van Lookeren Campagne M, Zhou Y. NLRP10 maintains epidermal homeostasis by promoting keratinocyte survival and P63-dependent differentiation and barrier function. Cell Death Dis 2024; 15:759. [PMID: 39424623 PMCID: PMC11492288 DOI: 10.1038/s41419-024-07146-y] [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: 05/10/2024] [Revised: 09/27/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024]
Abstract
Atopic dermatitis (AD) is a common chronic inflammatory skin disorder characterized by disrupted epidermal barrier function and aberrant immune responses. Despite recent developments in new therapeutics for AD, there is still a large unmet medical need for disease management due to the complex and multifactorial nature of AD. Recent genome-wide association studies (GWAS) have identified NLRP10 as a susceptible gene for AD but the physiological role of NLRP10 in skin homeostasis and AD remains unknown. Here we show that NLRP10 is downregulated in AD skin samples. Using an air-lift human skin equivalent culture, we demonstrate that NLRP10 promotes keratinocyte survival and is required for epidermal differentiation and barrier function. Mechanistically, NLRP10 limits cell death by preventing the recruitment of caspase-8 to the death inducing signaling complex (DISC) and by inhibiting its subsequent activation. NLRP10 also stabilizes p63, the master regulator of keratinocyte differentiation, to drive proper keratinocyte differentiation and to reinforce the barrier function. Our findings underscore NLRP10 as a key player in atopic dermatitis pathogenesis, highlighting NLRP10 as a potential target for therapeutic intervention to restore skin barrier function and homeostasis in AD.
Collapse
Affiliation(s)
- Yeonhee Cho
- Inflammation Research, Amgen Inc., South San Francisco, CA, USA
| | - Zhongzheng Cao
- Inflammation Research, Amgen Inc., South San Francisco, CA, USA
- Amgen R&D Postdoctoral Fellows Program, South San Francisco, CA, USA
| | - Xin Luo
- Center for Research Acceleration by Digital Innovation, Amgen Inc., South San Francisco, CA, USA
| | - Jennifer J Tian
- Translational Safety & Bioanalytical Sciences, Amgen Inc., South San Francisco, CA, USA
| | - Renee R Hukkanen
- Translational Safety & Bioanalytical Sciences, Amgen Inc, Cambridge, MA, USA
| | - Rajaa Hussien
- Translational Safety & Bioanalytical Sciences, Amgen Inc., South San Francisco, CA, USA
| | - Belinda Cancilla
- Translational Safety & Bioanalytical Sciences, Amgen Inc., South San Francisco, CA, USA
| | | | - Fei Li
- Structural Biology, Amgen Inc., South San Francisco, CA, USA
| | - Shining Ma
- Center for Research Acceleration by Digital Innovation, Amgen Inc., South San Francisco, CA, USA
| | - Edward L LaGory
- Pharmacokinetics and Drug Metabolism, Amgen Inc., South San Francisco, CA, USA
| | - Mark Schroeder
- Pharmacokinetics and Drug Metabolism, Amgen Inc., South San Francisco, CA, USA
| | | | | | - Jenn Hawkins
- Clinical Biomarkers, Amgen Inc, Thousand Oaks, CA, USA
| | | | - Yi Zhou
- Inflammation Research, Amgen Inc., South San Francisco, CA, USA.
| |
Collapse
|
20
|
Polito MP, Romaldini A, Tagliazucchi L, Marini G, Radice F, Gozza GA, Bergamini G, Costi MP, Enzo E. Biochemical characterization of the feedforward loop between CDK1 and FOXM1 in epidermal stem cells. Biol Direct 2024; 19:91. [PMID: 39396994 PMCID: PMC11472434 DOI: 10.1186/s13062-024-00540-8] [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: 09/23/2024] [Accepted: 10/07/2024] [Indexed: 10/15/2024] Open
Abstract
The complex network governing self-renewal in epidermal stem cells (EPSCs) is only partially defined. FOXM1 is one of the main players in this network, but the upstream signals regulating its activity remain to be elucidated. In this study, we identify cyclin-dependent kinase 1 (CDK1) as the principal kinase controlling FOXM1 activity in human primary keratinocytes. Mass spectrometry identified CDK1 as a key hub in a stem cell-associated protein network, showing its upregulation and interaction with essential self renewal-related markers. CDK1 phosphorylates FOXM1 at specific residues, stabilizing the protein and enhancing its nuclear localization and transcriptional activity, promoting self-renewal. Additionally, FOXM1 binds to the CDK1 promoter, inducing its expression.We identify the CDK1-FOXM1 feedforward loop as a critical axis sustaining EPSCs during in vitro cultivation. Understanding the upstream regulators of FOXM1 activity offers new insights into the biochemical mechanisms underlying self-renewal and differentiation in human primary keratinocytes.
Collapse
Affiliation(s)
- Maria Pia Polito
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Science, University of Modena and Reggio Emilia, Via Glauco Gottardi 100, Modena, Italy
| | - Alessio Romaldini
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Science, University of Modena and Reggio Emilia, Via Glauco Gottardi 100, Modena, Italy
| | - Lorenzo Tagliazucchi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, Modena, 41125, Italy
| | - Grazia Marini
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Science, University of Modena and Reggio Emilia, Via Glauco Gottardi 100, Modena, Italy
| | - Federica Radice
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Science, University of Modena and Reggio Emilia, Via Glauco Gottardi 100, Modena, Italy
| | - Gaia Andrea Gozza
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Science, University of Modena and Reggio Emilia, Via Glauco Gottardi 100, Modena, Italy
| | - Giulia Bergamini
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Science, University of Modena and Reggio Emilia, Via Glauco Gottardi 100, Modena, Italy
| | - Maria Paola Costi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi 103, Modena, 41125, Italy
| | - Elena Enzo
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Science, University of Modena and Reggio Emilia, Via Glauco Gottardi 100, Modena, Italy.
| |
Collapse
|
21
|
Zhang Y, Karagiannis D, Liu H, Lin M, Fang Y, Jiang M, Chen X, Suresh S, Huang H, She J, Shi F, Liu J, Luo D, Angel JC, Lin G, Yang P, El-Rifai W, Zaika A, Oro AE, Liu K, Rustgi AK, Wang TC, Lu C, Que J. Epigenetic regulation of p63 blocks squamous-to-neuroendocrine transdifferentiation in esophageal development and malignancy. SCIENCE ADVANCES 2024; 10:eadq0479. [PMID: 39383220 PMCID: PMC11463268 DOI: 10.1126/sciadv.adq0479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 09/04/2024] [Indexed: 10/11/2024]
Abstract
While cell fate determination and maintenance are important in establishing and preserving tissue identity and function during development, aberrant cell fate transition leads to cancer cell heterogeneity and resistance to treatment. Here, we report an unexpected role for the transcription factor p63 (Trp63/TP63) in the fate choice of the squamous versus neuroendocrine lineage in esophageal development and malignancy. Deletion of p63 results in extensive neuroendocrine differentiation in the developing mouse esophagus and esophageal progenitors derived from human embryonic stem cells. In human esophageal neuroendocrine carcinoma (eNEC) cells, p63 is transcriptionally silenced by EZH2-mediated H3K27 trimethylation (H3K27me3). Up-regulation of the major p63 isoform ΔNp63α, through either ectopic expression or EZH2 inhibition, promotes squamous transdifferentiation of eNEC cells. Together, these findings uncover p63 as a rheostat in coordinating the transition between squamous and neuroendocrine cell fates during esophageal development and tumor progression.
Collapse
Affiliation(s)
- Yongchun Zhang
- State Key Laboratory of Microbial Metabolism and Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
- Columbia Center for Human Development, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
- Division of Digestive and Liver Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Dimitris Karagiannis
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Helu Liu
- Columbia Center for Human Development, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
- Division of Digestive and Liver Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
- Institute of Deep-sea Science and Engineering, Chinese Academy of Sciences, Sanya 572000, Hainan, China
| | - Mi Lin
- Columbia Center for Human Development, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
- Division of Digestive and Liver Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian, China
| | - Yinshan Fang
- Columbia Center for Human Development, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
- Division of Digestive and Liver Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Ming Jiang
- Center for Genetic Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310030, Zhejiang, China
| | - Xiao Chen
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Supriya Suresh
- Columbia Center for Human Development, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
- Division of Digestive and Liver Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Haidi Huang
- State Key Laboratory of Microbial Metabolism and Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Junjun She
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi, China
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi, China
| | - Feiyu Shi
- Department of General Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi, China
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi, China
| | - Jiangying Liu
- State Key Laboratory of Microbial Metabolism and Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Dan Luo
- State Key Laboratory of Microbial Metabolism and Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - J. Carlos Angel
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Guangtan Lin
- Columbia Center for Human Development, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
- Division of Digestive and Liver Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian, China
| | - Patrick Yang
- Department of Internal Medicine, Westchester Medical Center/New York Medical College, Valhalla, NY 10595, USA
| | - Wael El-Rifai
- Department of Surgery and Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
- Miami Veterans Affairs Healthcare System, Miami, FL 33136, USA
| | - Alexander Zaika
- Department of Surgery and Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
- Miami Veterans Affairs Healthcare System, Miami, FL 33136, USA
| | - Anthony E. Oro
- Programin Epithelial Biology, Stanford University School of Medicine, Stanford 94305, CA, USA
| | - Kuancan Liu
- Central Laboratory, Xiang’an Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Anil K. Rustgi
- Division of Digestive and Liver Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
- Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Timothy C. Wang
- Division of Digestive and Liver Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
- Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Chao Lu
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
- Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Jianwen Que
- Columbia Center for Human Development, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
- Division of Digestive and Liver Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
- Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
22
|
Gall A, Bosticardo M, Ma S, Chen K, Amini K, Pala F, Delmonte OM, Wenger T, Bamshad M, Sleasman J, Blessing M, van Oers NSC, Notarangelo LD, de la Morena MT. Case report: Artificial thymic organoids facilitate clinical decisions for a patient with a TP63 variant and severe persistent T cell lymphopenia. Front Immunol 2024; 15:1438383. [PMID: 39364398 PMCID: PMC11448704 DOI: 10.3389/fimmu.2024.1438383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 08/27/2024] [Indexed: 10/05/2024] Open
Abstract
Pathogenic variants in the transcription factor TP63 are associated with clinically overlapping syndromes including ectrodactyly-ectodermal dysplasia clefting (EEC) and ankyloblepharon-ectodermal defects-cleft lip/palate (AEC). T cell lymphopenia has rarely been described in individuals with TP63 variants and the cause of the T cell defect is unclear. Here, we present a case of a female infant born with TP63-related syndrome and profound T cell lymphopenia, first uncovered through newborn screening. Flow cytometry analysis revealed low CD4+ naïve T cells and nearly absent CD8+ T cells with intact B and NK cell compartments. A de novo heterozygous pathogenic variant c.1040 G>A (C347Y) in exon 8 of TP63 was identified. An artificial thymic organoid system, to assess the intrinsic ability of the patient's hematopoietic cells to develop into T cells, was performed twice using separate peripheral blood samples. Ex vivo T cell differentiation was evident with the artificial organoid system, suggesting that a thymic stromal cell defect may be the cause of the T cell lymphopenia. Consistent with this, interrogation of publicly available data indicated that TP63 expression in the human thymus is restricted to thymic epithelial cells. Based on these data, congenital athymia was suspected and the patient received an allogenic cultured thymus tissue implant (CTTI). This is the first report of suspected congenital athymia and attempted treatment with CTTI associated with TP63 variant. At 9 months post-implant, peripheral lymphocyte analysis revealed measurable T cell receptor excision circles and presence of CD4+ recent thymic emigrants suggestive of early thymopoiesis. She will continue regular monitoring to ensure restoration of T cell immunity.
Collapse
Affiliation(s)
- Alevtina Gall
- Division of Immunology, Department of Pediatrics, University of Washington and Seattle Children’s Hospital, Seattle, WA, United States
| | - Marita Bosticardo
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Stacey Ma
- Division of Allergy and Infectious Diseases, Department of Internal Medicine, University of Washington, Seattle, WA, United States
| | - Karin Chen
- Division of Immunology, Department of Pediatrics, University of Washington and Seattle Children’s Hospital, Seattle, WA, United States
| | - Kayla Amini
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Francesca Pala
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Ottavia M. Delmonte
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Tara Wenger
- Division of Genetic Medicine, Department of Pediatrics, University of Washington and Seattle Children’s Hospital, Seattle, WA, United States
| | - Michael Bamshad
- Division of Genetic Medicine, Department of Pediatrics, University of Washington and Seattle Children’s Hospital, Seattle, WA, United States
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, WA, United States
| | - John Sleasman
- Department of Pediatrics, Duke University Medical Center, Durham, NC, United States
| | - Matthew Blessing
- Division of Craniofacial Medicine, Department of Pediatrics, University of Washington and Seattle Children’s Hospital, Seattle, WA, United States
| | - Nicolai S. C. van Oers
- Department of Immunology, Pediatrics and Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Luigi D. Notarangelo
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| | - M. Teresa de la Morena
- Division of Immunology, Department of Pediatrics, University of Washington and Seattle Children’s Hospital, Seattle, WA, United States
| |
Collapse
|
23
|
Zhao J, Williams MD, Hernandez M, Kuang G, Goldberg H, Fan J, Ning J, Ferrarotto R, Esmaeli B. Prognostic Impact of Notch1 Intracellular Domain, P63, and c-MYC in Lacrimal Gland Adenoid Cystic Carcinoma. Invest Ophthalmol Vis Sci 2024; 65:4. [PMID: 39230995 PMCID: PMC11379087 DOI: 10.1167/iovs.65.11.4] [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: 03/11/2024] [Accepted: 07/03/2024] [Indexed: 09/06/2024] Open
Abstract
Purpose We assessed whether NICD1 expression, c-MYC expression, and P63 expression by immunohistochemistry (IHC) correlate with prognosis and high-risk clinicopathological features in lacrimal gland adenoid cystic carcinoma (ACC). Methods Records of patients with lacrimal gland ACC who underwent surgery between 1998 to 2018 were reviewed. Clinicopathologic and treatment data were collected. Tumor tissues were subjected to light microscopy and IHC. Results Of 43 patients treated during the study period, 21 had archived tumor tissue available and were included. The median age at diagnosis was 47 years, and 13 patients (62%) were male. Thirteen patients (62%) had T2 disease, and none had nodal or distant metastasis at diagnosis. Tumors were positive for NICD1 expression in eight cases (38%), c-MYC expression in eight (38%), and P63 expression in 11 (52%). Positive NICD1 expression was associated with predominantly solid (vs. cribriform/tubular) pattern (P < 0.001), treatment with orbital exenteration (vs. eye-sparing surgery) (P = 0.008), local recurrence (P = 0.047), and death (P = 0.012). Negative P63 expression was associated with predominantly solid pattern (P = 0.001), local recurrence (P = 0.012), distant metastasis (P = 0.001), and death (P = 0.035). A higher percentage of tumor cells staining for c-MYC was associated with presence of perineural invasion (P = 0.036). Positive NICD1 expression was associated with worse disease-free survival (hazard ratio, 6.27; 95% CI, 1.29-30.46), whereas positive P63 expression was associated with better disease-free survival (hazard ratio, 0.03; 95% CI, 0.0002-0.26). Conclusions IHC for NICD1 and P63 should be considered in lacrimal gland ACC because of their prognostic value and potential as treatment targets.
Collapse
Affiliation(s)
- Jiawei Zhao
- Orbital Oncology & Ophthalmic Plastic Surgery, Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | - Michelle D. Williams
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | - Mike Hernandez
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | - Grace Kuang
- Orbital Oncology & Ophthalmic Plastic Surgery, Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | - Hila Goldberg
- Orbital Oncology & Ophthalmic Plastic Surgery, Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | - Janet Fan
- Orbital Oncology & Ophthalmic Plastic Surgery, Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | - Jing Ning
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | - Renata Ferrarotto
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | - Bita Esmaeli
- Orbital Oncology & Ophthalmic Plastic Surgery, Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| |
Collapse
|
24
|
Suanno G, Genna VG, Maurizi E, Dieh AA, Griffith M, Ferrari G. Cell therapy in the cornea: The emerging role of microenvironment. Prog Retin Eye Res 2024; 102:101275. [PMID: 38797320 DOI: 10.1016/j.preteyeres.2024.101275] [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: 10/11/2023] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
The cornea is an ideal testing field for cell therapies. Its highly ordered structure, where specific cell populations are sequestered in different layers, together with its accessibility, has allowed the development of the first stem cell-based therapy approved by the European Medicine Agency. Today, different techniques have been proposed for autologous and allogeneic limbal and non-limbal cell transplantation. Cell replacement has also been attempted in cases of endothelial cell decompensation as it occurs in Fuchs dystrophy: injection of cultivated allogeneic endothelial cells is now in advanced phases of clinical development. Recently, stromal substitutes have been developed with excellent integration capability and transparency. Finally, cell-derived products, such as exosomes obtained from different sources, have been investigated for the treatment of severe corneal diseases with encouraging results. Optimization of the success rate of cell therapies obviously requires high-quality cultured cells/products, but the role of the surrounding microenvironment is equally important to allow engraftment of transplanted cells, to preserve their functions and, ultimately, lead to restoration of tissue integrity and transparency of the cornea.
Collapse
Affiliation(s)
- Giuseppe Suanno
- Vita-Salute San Raffaele University, Milan, Italy; Eye Repair Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Eleonora Maurizi
- Centre for Regenerative Medicine ''S. Ferrari'', University of Modena and Reggio Emilia, Modena, Italy
| | - Anas Abu Dieh
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, Quebec, Canada
| | - May Griffith
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, Quebec, Canada.
| | - Giulio Ferrari
- Vita-Salute San Raffaele University, Milan, Italy; Eye Repair Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy; Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
25
|
Li W, Yang Y, Huang L, Yu X, Wang T, Zhang N, Yang M. The TDP-43/TP63 Positive Feedback Circuit Promotes Esophageal Squamous Cell Carcinoma Progression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402913. [PMID: 39023169 PMCID: PMC11425248 DOI: 10.1002/advs.202402913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/30/2024] [Indexed: 07/20/2024]
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most prevalent malignancies with a 5-year survival rate of only 15% in patients with advanced diseases. Tumor protein 63 (TP63), a master transcription factor (TF) in ESCC, cooperates with other TFs to regulate enhancers and/or promoters of target oncogenes, which in turn promotes tumorigenesis. TAR-DNA-binding protein-43 (TDP-43) is an RNA/DNA binding protein with elevated expression in several neoplasms. However, it remains unclear how TDP-43 contributes to ESCC progression. In this study, TDP-43 is identified as a novel oncogene with markedly upregulated expression in ESCC tissues through profiling expression levels of one hundred and fifty canonical RNA binding protein (RBP) genes in multiple ESCC patient cohorts. Importantly, TDP-43 boosted TP63 expression via post-transcriptionally stabilizing TP63 mRNAs as a RBP and promoting TP63 transcription as a TF binding to the TP63 promoter in ESCC cells. In contrast, the master TF TP63 also bound to the TDP-43 promoter, accelerated TDP-43 transcription, and caused a noticeable increase in TDP-43 expression in ESCC cells. The findings highlight TDP-43 as a viable therapeutic target for ESCC and uncover a hitherto unrecognized TDP-43/TP63 circuit in cancer.
Collapse
Affiliation(s)
- Wenwen Li
- Shandong Provincial Key Laboratory of Radiation Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
- School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong, 271021, China
| | - Yanting Yang
- Shandong Provincial Key Laboratory of Radiation Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Linying Huang
- Shandong Provincial Key Laboratory of Radiation Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Xinyuan Yu
- Shandong Provincial Key Laboratory of Radiation Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Teng Wang
- Shandong University Cancer Center, Jinan, Shandong, 250117, China
| | - Nasha Zhang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, 250117, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Ming Yang
- Shandong Provincial Key Laboratory of Radiation Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
- School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong, 271021, China
- Shandong University Cancer Center, Jinan, Shandong, 250117, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, 250117, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| |
Collapse
|
26
|
La Banca V, De Domenico S, Nicolai S, Gatti V, Scalera S, Maugeri M, Mauriello A, Montanaro M, Pahnke J, Candi E, D’Amico S, Peschiaroli A. ABCC1 Is a ΔNp63 Target Gene Overexpressed in Squamous Cell Carcinoma. Int J Mol Sci 2024; 25:8741. [PMID: 39201428 PMCID: PMC11354449 DOI: 10.3390/ijms25168741] [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: 07/03/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
The transcription factor ΔNp63 plays a pivotal role in maintaining the integrity of stratified epithelial tissues by regulating the expression of distinct target genes involved in lineage specification, cell stemness, cell proliferation and differentiation. Here, we identified the ABC transporter subfamily member ABCC1 as a novel ΔNp63 target gene. We found that in immortalized human keratinocytes and in squamous cell carcinoma (SCC) cells, ∆Np63 induces the expression of ABCC1 by physically occupying a p63-binding site (p63 BS) located in the first intron of the ABCC1 gene locus. In cutaneous SCC and during the activation of the keratinocyte differentiation program, ∆Np63 and ABCC1 levels are positively correlated raising the possibility that ABCC1 might be involved in the regulation of the proliferative/differentiative capabilities of squamous tissue. However, we did not find any gross alteration in the structure and morphology of the epidermis in humanized hABCC1 knock-out mice. Conversely, we found that the genetic ablation of ABCC1 led to a marked reduction in inflammation-mediated proliferation of keratinocytes, suggesting that ABCC1 might be involved in the regulation of keratinocyte proliferation upon inflammatory/proliferative signals. In line with these observations, we found a significant increase in ABCC1 expression in squamous cell carcinomas (SCCs), a tumor type characterized by keratinocyte hyper-proliferation and a pro-inflammatory tumor microenvironment. Collectively, these data uncover ABCC1 as an additional ∆Np63 target gene potentially involved in those skin diseases characterized by dysregulation of proliferation/differentiation balance.
Collapse
Affiliation(s)
- Veronica La Banca
- Department of Experimental Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (V.L.B.); (S.D.D.); (A.M.); (E.C.)
| | - Sara De Domenico
- Department of Experimental Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (V.L.B.); (S.D.D.); (A.M.); (E.C.)
| | - Sara Nicolai
- Institute of Translational Pharmacology (IFT), CNR, Via Fosso del Cavaliere 100, 00133 Rome, Italy; (S.N.); (V.G.)
| | - Veronica Gatti
- Institute of Translational Pharmacology (IFT), CNR, Via Fosso del Cavaliere 100, 00133 Rome, Italy; (S.N.); (V.G.)
| | - Stefano Scalera
- UOSD Clinical Trial Center, Biostatistics and Bioinformatics Division, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (S.S.); (M.M.)
| | - Marcello Maugeri
- UOSD Clinical Trial Center, Biostatistics and Bioinformatics Division, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (S.S.); (M.M.)
| | - Alessandro Mauriello
- Department of Experimental Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (V.L.B.); (S.D.D.); (A.M.); (E.C.)
| | - Manuela Montanaro
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy;
| | - Jens Pahnke
- Translational Neurodegeneration Research and Neuropathology Lab/Section of Neuropathology Research, Department of Pathology (PAT), Medical Faculty/Clinical Medicine (KlinMed), Clinics for Laboratory Medicine (KLM), University of Oslo (UiO) and Oslo University Hospital (OUS), Sognsvannsveien 20, 0372 Oslo, Norway;
- Institute of Nutritional Medicine (INUM)/Lübeck Institute of Dermatology (LIED), University of Lübeck (UzL) and University Medical Center Schleswig-Holstein (UKSH), Ratzeburger Allee 160, D-23538 Lübeck, Germany
- Department of Pharmacology, The Faculty of Medicine and Life Sciences, University of Latvia (LU), Jelgavas iela 3, LV-1004 Rīga, Latvia
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics, The Georg S. Wise Faculty of Life Sciences, Tel Aviv University (TAU), Tel Aviv 6997801, Israel
| | - Eleonora Candi
- Department of Experimental Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy; (V.L.B.); (S.D.D.); (A.M.); (E.C.)
- Biochemistry Laboratory, Istituto Dermopatico Immacolata (IDI-IRCCS), 00166 Rome, Italy
| | - Silvia D’Amico
- Institute of Translational Pharmacology (IFT), CNR, Via Fosso del Cavaliere 100, 00133 Rome, Italy; (S.N.); (V.G.)
| | - Angelo Peschiaroli
- Institute of Translational Pharmacology (IFT), CNR, Via Fosso del Cavaliere 100, 00133 Rome, Italy; (S.N.); (V.G.)
| |
Collapse
|
27
|
Dermitzakis I, Chatzi D, Kyriakoudi SA, Evangelidis N, Vakirlis E, Meditskou S, Theotokis P, Manthou ME. Skin Development and Disease: A Molecular Perspective. Curr Issues Mol Biol 2024; 46:8239-8267. [PMID: 39194704 DOI: 10.3390/cimb46080487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/27/2024] [Accepted: 07/28/2024] [Indexed: 08/29/2024] Open
Abstract
Skin, the largest organ in the human body, is a crucial protective barrier that plays essential roles in thermoregulation, sensation, and immune defence. This complex organ undergoes intricate processes of development. Skin development initiates during the embryonic stage, orchestrated by molecular cues that control epidermal specification, commitment, stratification, terminal differentiation, and appendage growth. Key signalling pathways are integral in coordinating the development of the epidermis, hair follicles, and sweat glands. The complex interplay among these pathways is vital for the appropriate formation and functionality of the skin. Disruptions in multiple molecular pathways can give rise to a spectrum of skin diseases, from congenital skin disorders to cancers. By delving into the molecular mechanisms implicated in developmental processes, as well as in the pathogenesis of diseases, this narrative review aims to present a comprehensive understanding of these aspects. Such knowledge paves the way for developing innovative targeted therapies and personalised treatment approaches for various skin conditions.
Collapse
Affiliation(s)
- Iasonas Dermitzakis
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Despoina Chatzi
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Stella Aikaterini Kyriakoudi
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Nikolaos Evangelidis
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Efstratios Vakirlis
- First Department of Dermatology and Venereology, School of Medicine, Aristotle University of Thessaloniki, 54643 Thessaloniki, Greece
| | - Soultana Meditskou
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Paschalis Theotokis
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Maria Eleni Manthou
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| |
Collapse
|
28
|
Sevilla LM, Pons-Alonso O, Gallego A, Azkargorta M, Elortza F, Pérez P. Glucocorticoid receptor controls atopic dermatitis inflammation via functional interactions with P63 and autocrine signaling in epidermal keratinocytes. Cell Death Dis 2024; 15:535. [PMID: 39069531 DOI: 10.1038/s41419-024-06926-w] [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: 01/26/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 07/30/2024]
Abstract
Atopic dermatitis (AD), a prevalent chronic inflammatory disease with multifactorial etiology, features epidermal barrier defects and immune overactivation. Synthetic glucocorticoids (GCs) are widely prescribed for treating AD due to their anti-inflammatory actions; however, mechanisms are incompletely understood. Defective local GC signaling due to decreased production of endogenous ligand and/or GC receptor (GR) levels was reported in prevalent inflammatory skin disorders; whether this is a consequence or contributing factor to AD pathology is unclear. To identify the chromatin-bound cell-type-specific GR protein interactome in keratinocytes, we used rapid immunoprecipitation of endogenous proteins and mass spectrometry identifying 145 interactors that increased upon dexamethasone treatment. GR-interacting proteins were enriched in p53/p63 signaling, including epidermal transcription factors with critical roles in AD pathology. Previous analyses indicating mirrored AD-like phenotypes between P63 overexpression and GR loss in epidermis, and our data show an intricate relationship between these transcription factors in human keratinocytes, identifying TP63 as a direct GR target. Dexamethasone treatment counteracted transcriptional up-regulation of inflammatory markers by IL4/IL13, known to mimic AD, causing opposite shifts in GR and P63 genomic binding. Indeed, IL4/IL13 decreased GR and increased P63 levels in cultured keratinocytes and human epidermal equivalents (HEE), consistent with GR down-regulation and increased P63 expression in AD lesions vs normal skin. Moreover, GR knockdown (GRKD) resulted in constitutive increases in P63, phospho-P38 and S100A9, IL6, and IL33. Also, GRKD culture supernatants showed increased autocrine production of TH2-/TH1-/TH17-TH22-associated factors including IL4, CXCL10, CXCL11, and CXCL8. GRKD HEEs showed AD-like features including hyperplasia and abnormal differentiation, resembling phenotypes observed with GR antagonist or IL4/IL13 treatment. The simultaneous GR/P63 knockdown partially reversed constitutive up-regulation of inflammatory genes in GRKD. In summary, our data support a causative role for GR loss in AD pathogenesis via functional interactions with P63 and autocrine signaling in epidermal keratinocytes.
Collapse
Affiliation(s)
- Lisa M Sevilla
- Instituto de Biomedicina de Valencia (IBV-CSIC), Department of Pathology and Molecular and Cell Therapy, Valencia, Spain
| | - Omar Pons-Alonso
- Instituto de Biomedicina de Valencia (IBV-CSIC), Department of Pathology and Molecular and Cell Therapy, Valencia, Spain
| | - Andrea Gallego
- Instituto de Biomedicina de Valencia (IBV-CSIC), Department of Pathology and Molecular and Cell Therapy, Valencia, Spain
| | - Mikel Azkargorta
- Proteomics Platform, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), CIBERehd, Science and Technology Park of Bizkaia, Derio, Spain
| | - Félix Elortza
- Proteomics Platform, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), CIBERehd, Science and Technology Park of Bizkaia, Derio, Spain
| | - Paloma Pérez
- Instituto de Biomedicina de Valencia (IBV-CSIC), Department of Pathology and Molecular and Cell Therapy, Valencia, Spain.
| |
Collapse
|
29
|
Polito MP, Marini G, Fabrizi A, Sercia L, Enzo E, De Luca M. Biochemical role of FOXM1-dependent histone linker H1B in human epidermal stem cells. Cell Death Dis 2024; 15:508. [PMID: 39019868 PMCID: PMC11255229 DOI: 10.1038/s41419-024-06905-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/19/2024]
Abstract
Epidermal stem cells orchestrate epidermal renewal and timely wound repair through a tight regulation of self-renewal, proliferation, and differentiation. In culture, human epidermal stem cells generate a clonal type referred to as holoclone, which give rise to transient amplifying progenitors (meroclone and paraclone-forming cells) eventually generating terminally differentiated cells. Leveraging single-cell transcriptomic data, we explored the FOXM1-dependent biochemical signals controlling self-renewal and differentiation in epidermal stem cells aimed at improving regenerative medicine applications. We report that the expression of H1 linker histone subtypes decrease during serial cultivation. At clonal level we observed that H1B is the most expressed isoform, particularly in epidermal stem cells, as compared to transient amplifying progenitors. Indeed, its expression decreases in primary epithelial culture where stem cells are exhausted due to FOXM1 downregulation. Conversely, H1B expression increases when the stem cells compartment is sustained by enforced FOXM1 expression, both in primary epithelial cultures derived from healthy donors and JEB patient. Moreover, we demonstrated that FOXM1 binds the promotorial region of H1B, hence regulates its expression. We also show that H1B is bound to the promotorial region of differentiation-related genes and negatively regulates their expression in epidermal stem cells. We propose a novel mechanism wherein the H1B acts downstream of FOXM1, contributing to the fine interplay between self-renewal and differentiation in human epidermal stem cells. These findings further define the networks that sustain self-renewal along the previously identified YAP-FOXM1 axis.
Collapse
Affiliation(s)
- Maria Pia Polito
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Science, University of Modena and Reggio Emilia, Modena, Italy
| | - Grazia Marini
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Science, University of Modena and Reggio Emilia, Modena, Italy
| | - Alessandra Fabrizi
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Science, University of Modena and Reggio Emilia, Modena, Italy
| | - Laura Sercia
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Science, University of Modena and Reggio Emilia, Modena, Italy
| | - Elena Enzo
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Science, University of Modena and Reggio Emilia, Modena, Italy.
| | - Michele De Luca
- Centre for Regenerative Medicine "Stefano Ferrari", Department of Life Science, University of Modena and Reggio Emilia, Modena, Italy.
| |
Collapse
|
30
|
Lin Y, Tang W, Huang P, Wang Z, Duan L, Jia C, Sun R, Liu L, Shen J. Denticleless E3 ubiquitin protein ligase (DTL) maintains the proliferation and differentiation of epidermis and hair follicles during skin development. Dev Dyn 2024; 253:635-647. [PMID: 38131461 DOI: 10.1002/dvdy.682] [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: 03/27/2023] [Revised: 11/16/2023] [Accepted: 11/20/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND A precise balance between the proliferation and differentiation of epidermal progenitors is required to achieve the barrier function during the development of epidermis. During the entire process of skin development, the newly formed basal layer cells divide, differentiate, and migrate outward to the surface of the skin, which is tightly regulated by a series of events related to cell cycle progression. The CRL4DTL complex (Cullin 4 RING ligase, in association with the substrate receptor DTL) has long emerged as a master regulator in various cellular processes, which mediates the degradation of key cell cycle proteins. However, the roles of DTL in regulating epidermal morphogenesis during skin development remain unclear. RESULTS We showed that DTL deficiency in epidermal progenitor cells leads to defects in epidermal stratification and loss of hair follicles accompanied by reduced epidermal progenitor cells and disturbed cell cycle progression during skin development. Transcriptome analysis revealed that p53 pathway is activated in DTL-depleted epidermal progenitor cells. The apoptosis of epidermal cells showed in DTL deficiency mice is rescued by the absence of p53, but the proliferation and differentiation defects were p53-independent. CONCLUSION Our findings indicate that DTL plays a vital role in epidermal malformation during skin development.
Collapse
Affiliation(s)
- Yanhui Lin
- Institute of Life Sciences, College of Life and Environmental Science, Wenzhou University, Wenzhou, China
| | - Weibo Tang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- Laboratory of Tumor Targeted Therapy and Translational Medicine, Jilin Medical University, Jilin, China
| | - Peijun Huang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Zhendong Wang
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lian Duan
- Central Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chonghui Jia
- Department of Endodontics, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ruizhen Sun
- Department of Histology and Embryology, Harbin Medical University, Harbin, China
| | - Li Liu
- Institute of Life Sciences, College of Life and Environmental Science, Wenzhou University, Wenzhou, China
| | - Jingling Shen
- Institute of Life Sciences, College of Life and Environmental Science, Wenzhou University, Wenzhou, China
- Department of Histology and Embryology, Harbin Medical University, Harbin, China
| |
Collapse
|
31
|
Chouhan G, Lewis NS, Ghanekar V, Koti Ainavarapu SR, Inamdar MM, Sonawane M. Cell-size-dependent regulation of Ezrin dictates epithelial resilience to stretch by countering myosin-II-mediated contractility. Cell Rep 2024; 43:114271. [PMID: 38823013 DOI: 10.1016/j.celrep.2024.114271] [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: 06/29/2023] [Revised: 04/22/2024] [Accepted: 05/09/2024] [Indexed: 06/03/2024] Open
Abstract
The epithelial adaptations to mechanical stress are facilitated by molecular and tissue-scale changes that include the strengthening of junctions, cytoskeletal reorganization, and cell-proliferation-mediated changes in tissue rheology. However, the role of cell size in controlling these properties remains underexplored. Our experiments in the zebrafish embryonic epidermis, guided by theoretical estimations, reveal a link between epithelial mechanics and cell size, demonstrating that an increase in cell size compromises the tissue fracture strength and compliance. We show that an increase in E-cadherin levels in the proliferation-deficient epidermis restores epidermal compliance but not the fracture strength, which is largely regulated by Ezrin-an apical membrane-cytoskeleton crosslinker. We show that Ezrin fortifies the epithelium in a cell-size-dependent manner by countering non-muscle myosin-II-mediated contractility. This work uncovers the importance of cell size maintenance in regulating the mechanical properties of the epithelium and fostering protection against future mechanical stresses.
Collapse
Affiliation(s)
- Geetika Chouhan
- Department of Biological Sciences, Tata Institute of Fundamental Research, Colaba, Mumbai, India
| | - Natasha Steffi Lewis
- Department of Biological Sciences, Tata Institute of Fundamental Research, Colaba, Mumbai, India
| | - Vallari Ghanekar
- Department of Biological Sciences, Tata Institute of Fundamental Research, Colaba, Mumbai, India
| | | | - Mandar M Inamdar
- Department of Civil Engineering, Indian Institute of Technology Bombay, Mumbai, India.
| | - Mahendra Sonawane
- Department of Biological Sciences, Tata Institute of Fundamental Research, Colaba, Mumbai, India.
| |
Collapse
|
32
|
Jelin AC, Sopko N, Sobreira N, Boyadjiev SA, Wohler E, Morrill C, Witmer PD, Michaud J, Valle D, Gearhart J, Dicarlo H. Rare exonic CELSR3 variants identified in Bladder Exstrophy Epispadias Complex. Front Genet 2024; 15:1266210. [PMID: 38903756 PMCID: PMC11188427 DOI: 10.3389/fgene.2024.1266210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 03/28/2024] [Indexed: 06/22/2024] Open
Abstract
Introduction/background Bladder exstrophy epispadias complex (BEEC) is a rare congenital anomaly of unknown etiology, although, genetic and environmental factors have been associated with its development. Variants in several genes expressed in the urogenital pathway have been reported as causative for bladder exstrophy in human and murine models. The expansion of next-generation sequencing and molecular genomics has improved our ability to identify the underlying genetic causes of similarly complex diseases and could thus assist with the investigation of the molecular basis of BEEC. Objective The objective was to identify the presence of rare heterozygous variants in genes previously implicated in bladder exstrophy and correlate them with the presence or absence of bladder regeneration in our study population. Patients and Methods We present a case series of 12 patients with BEEC who had bladder biopsies performed by pediatric urology during bladder neck reconstruction or bladder augmentation. Cases were classified as "sufficient" or "insufficient" (n = 5 and 7, respectively) based on a bladder volume of greater than or less than 40% of expected bladder size. Control bladder tissue specimens were obtained from patients (n = 6) undergoing biopsies for conditions other than bladder exstrophy. Whole exome sequencing was performed on DNA isolated from the bladder specimens. Based on the hypothesis of de novo mutations, as well as the potential implications of autosomal dominant conditions with incomplete penetrance, each case was evaluated for autosomal dominant variants in a set of genes previously implicated in BEEC. Results Our review of the literature identified 44 genes that have been implicated in human models of bladder exstrophy. Our whole exome sequencing data analysis identified rare variants in two of these genes among the cases classified as sufficient, and seven variants in five of these genes among the cases classified as insufficient. Conclusion We identified rare variants in seven previously implicated genes in our BEEC specimens. Additional research is needed to further understand the cellular signaling underlying this potentially genetically heterogeneous embryological condition.
Collapse
Affiliation(s)
- Angie C. Jelin
- Department of Gynecology and Obstetrics, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Nikolai Sopko
- Department of Pediatric Urology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Nara Sobreira
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Simeon A. Boyadjiev
- Department of Pediatrics, University of California Davis, Davis, CA, United States
| | - Elizabeth Wohler
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Christian Morrill
- Department of Pediatric Urology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - P. Dane Witmer
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Jason Michaud
- Department of Pediatric Urology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - David Valle
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - John Gearhart
- Department of Pediatric Urology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Heather Dicarlo
- Department of Pediatric Urology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| |
Collapse
|
33
|
Wu HH, Leng S, Sergi C, Leng R. How MicroRNAs Command the Battle against Cancer. Int J Mol Sci 2024; 25:5865. [PMID: 38892054 PMCID: PMC11172831 DOI: 10.3390/ijms25115865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/23/2024] [Accepted: 05/26/2024] [Indexed: 06/21/2024] Open
Abstract
MicroRNAs (miRNAs) are small RNA molecules that regulate more than 30% of genes in humans. Recent studies have revealed that miRNAs play a crucial role in tumorigenesis. Large sets of miRNAs in human tumors are under-expressed compared to normal tissues. Furthermore, experiments have shown that interference with miRNA processing enhances tumorigenesis. Multiple studies have documented the causal role of miRNAs in cancer, and miRNA-based anticancer therapies are currently being developed. This review primarily focuses on two key points: (1) miRNAs and their role in human cancer and (2) the regulation of tumor suppressors by miRNAs. The review discusses (a) the regulation of the tumor suppressor p53 by miRNA, (b) the critical role of the miR-144/451 cluster in regulating the Itch-p63-Ago2 pathway, and (c) the regulation of PTEN by miRNAs. Future research and the perspectives of miRNA in cancer are also discussed. Understanding these pathways will open avenues for therapeutic interventions targeting miRNA regulation.
Collapse
Affiliation(s)
- Hong Helena Wu
- 370 Heritage Medical Research Center, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2S2, Canada;
| | - Sarah Leng
- Department of Laboratory Medicine and Pathology (5B4. 09), University of Alberta, Edmonton, AB T6G 2B7, Canada (C.S.)
| | - Consolato Sergi
- Department of Laboratory Medicine and Pathology (5B4. 09), University of Alberta, Edmonton, AB T6G 2B7, Canada (C.S.)
- Division of Anatomical Pathology, Children’s Hospital of Eastern Ontario (CHEO), University of Ottawa, 401 Smyth Road, Ottawa, ON K1H 8L1, Canada
| | - Roger Leng
- 370 Heritage Medical Research Center, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2S2, Canada;
| |
Collapse
|
34
|
Seager RJ, Ko H, Pabla S, Senosain MF, Kalinski P, Van Roey E, Gao S, Strickland KC, Previs RA, Nesline MK, Hastings S, Zhang S, Conroy JM, Jensen TJ, Eisenberg M, Caveney B, Severson EA, Ramkissoon S, Gandhi S. Immunologic Factors Associated with Differential Response to Neoadjuvant Chemoimmunotherapy in Triple-Negative Breast Cancer. J Pers Med 2024; 14:481. [PMID: 38793063 PMCID: PMC11122407 DOI: 10.3390/jpm14050481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 04/17/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
Background: KEYNOTE-522 resulted in FDA approval of the immune checkpoint inhibitor pembrolizumab in combination with neoadjuvant chemotherapy for patients with early-stage, high-risk, triple-negative breast cancer (TNBC). Unfortunately, pembrolizumab is associated with several immune-related adverse events (irAEs). We aimed to identify potential tumor microenvironment (TME) biomarkers which could predict patients who may attain pathological complete response (pCR) with chemotherapy alone and be spared the use of anti-PD-1 immunotherapy. Methods: Comprehensive immune profiling, including RNA-seq gene expression assessment of 395 immune genes, was performed on matched FFPE tumor samples from 22 stage I-III TNBC patients (14 patients treated with neoadjuvant chemotherapy alone (NAC) and 8 treated with neoadjuvant chemotherapy combined with pembrolizumab (NAC+I)). Results: Differential gene expression analysis revealed that in the NAC group, IL12B and IL13 were both significantly associated with pCR. In the NAC+I group, LCK and TP63 were significantly associated with pCR. Patients in both treatment groups exhibiting pCR tended to have greater tumor inflammation than non-pCR patients. In the NAC+I group, patients with pCR tended to have greater cell proliferation and higher PD-L1 expression, while in the NAC group, patients with pCR tended to have lower cancer testis antigen expression. Additionally, the NAC+I group trended toward a lower relative dose intensity averaged across all chemotherapy drugs, suggesting that more dose reductions or treatment delays occurred in the NAC+I group than the NAC group. Conclusions: A comprehensive understanding of immunologic factors could potentially predict pCR to chemotherapy alone, enabling the avoidance of the unnecessary treatment of these patients with checkpoint inhibitors.
Collapse
Affiliation(s)
- Robert J. Seager
- Labcorp Oncology, Buffalo, NY 14263, USA; (S.P.); (M.-F.S.); (E.V.R.); (S.G.); (S.Z.); (J.M.C.)
| | - Heidi Ko
- Labcorp Oncology, Durham, NC 27710, USA; (H.K.); (K.C.S.); (R.A.P.); (M.K.N.); (S.H.); (T.J.J.); (E.A.S.); (S.R.)
| | - Sarabjot Pabla
- Labcorp Oncology, Buffalo, NY 14263, USA; (S.P.); (M.-F.S.); (E.V.R.); (S.G.); (S.Z.); (J.M.C.)
| | - Maria-Fernanda Senosain
- Labcorp Oncology, Buffalo, NY 14263, USA; (S.P.); (M.-F.S.); (E.V.R.); (S.G.); (S.Z.); (J.M.C.)
| | - Pawel Kalinski
- Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
| | - Erik Van Roey
- Labcorp Oncology, Buffalo, NY 14263, USA; (S.P.); (M.-F.S.); (E.V.R.); (S.G.); (S.Z.); (J.M.C.)
| | - Shuang Gao
- Labcorp Oncology, Buffalo, NY 14263, USA; (S.P.); (M.-F.S.); (E.V.R.); (S.G.); (S.Z.); (J.M.C.)
| | - Kyle C. Strickland
- Labcorp Oncology, Durham, NC 27710, USA; (H.K.); (K.C.S.); (R.A.P.); (M.K.N.); (S.H.); (T.J.J.); (E.A.S.); (S.R.)
- Department of Pathology, Duke University Medical Center, Duke Cancer Institute, Durham, NC 27710, USA
| | - Rebecca Ann Previs
- Labcorp Oncology, Durham, NC 27710, USA; (H.K.); (K.C.S.); (R.A.P.); (M.K.N.); (S.H.); (T.J.J.); (E.A.S.); (S.R.)
- Department of Obstetrics & Gynecology, Duke University Medical Center, Duke Cancer Institute, Division of Gynecologic Oncology, Durham, NC 27710, USA
| | - Mary K. Nesline
- Labcorp Oncology, Durham, NC 27710, USA; (H.K.); (K.C.S.); (R.A.P.); (M.K.N.); (S.H.); (T.J.J.); (E.A.S.); (S.R.)
| | - Stephanie Hastings
- Labcorp Oncology, Durham, NC 27710, USA; (H.K.); (K.C.S.); (R.A.P.); (M.K.N.); (S.H.); (T.J.J.); (E.A.S.); (S.R.)
| | - Shengle Zhang
- Labcorp Oncology, Buffalo, NY 14263, USA; (S.P.); (M.-F.S.); (E.V.R.); (S.G.); (S.Z.); (J.M.C.)
| | - Jeffrey M. Conroy
- Labcorp Oncology, Buffalo, NY 14263, USA; (S.P.); (M.-F.S.); (E.V.R.); (S.G.); (S.Z.); (J.M.C.)
| | - Taylor J. Jensen
- Labcorp Oncology, Durham, NC 27710, USA; (H.K.); (K.C.S.); (R.A.P.); (M.K.N.); (S.H.); (T.J.J.); (E.A.S.); (S.R.)
| | | | | | - Eric A. Severson
- Labcorp Oncology, Durham, NC 27710, USA; (H.K.); (K.C.S.); (R.A.P.); (M.K.N.); (S.H.); (T.J.J.); (E.A.S.); (S.R.)
| | - Shakti Ramkissoon
- Labcorp Oncology, Durham, NC 27710, USA; (H.K.); (K.C.S.); (R.A.P.); (M.K.N.); (S.H.); (T.J.J.); (E.A.S.); (S.R.)
- Wake Forest Comprehensive Cancer Center and Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC 27710, USA
| | - Shipra Gandhi
- Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
| |
Collapse
|
35
|
Nagano H, Mizuno N, Sato H, Mizutani E, Yanagida A, Kano M, Kasai M, Yamamoto H, Watanabe M, Suchy F, Masaki H, Nakauchi H. Skin graft with dermis and appendages generated in vivo by cell competition. Nat Commun 2024; 15:3366. [PMID: 38684678 PMCID: PMC11058811 DOI: 10.1038/s41467-024-47527-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 04/03/2024] [Indexed: 05/02/2024] Open
Abstract
Autologous skin grafting is a standard treatment for skin defects such as burns. No artificial skin substitutes are functionally equivalent to autologous skin grafts. The cultured epidermis lacks the dermis and does not engraft deep wounds. Although reconstituted skin, which consists of cultured epidermal cells on a synthetic dermal substitute, can engraft deep wounds, it requires the wound bed to be well-vascularized and lacks skin appendages. In this study, we successfully generate complete skin grafts with pluripotent stem cell-derived epidermis with appendages on p63 knockout embryos' dermis. Donor pluripotent stem cell-derived keratinocytes encroach the embryos' dermis by eliminating p63 knockout keratinocytes based on cell-extracellular matrix adhesion mediated cell competition. Although the chimeric skin contains allogenic dermis, it is engraftable as long as autologous grafts. Furthermore, we could generate semi-humanized skin segments by human keratinocytes injection into the amnionic cavity of p63 knockout mice embryos. Niche encroachment opens the possibility of human skin graft production in livestock animals.
Collapse
Affiliation(s)
- Hisato Nagano
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
- Department of Plastic and Reconstructive Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Naoaki Mizuno
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan.
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan.
- Department of Experimental Animal Model for Human Disease, Center for Experimental Animals, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan.
| | - Hideyuki Sato
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Eiji Mizutani
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
- Laboratory of Stem Cell Therapy, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Ayaka Yanagida
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
- Department of Veterinary Anatomy, The University of Tokyo, Yayoi 1-1-1, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Mayuko Kano
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
- Metabolism and Endocrinology, Department of Medicine, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan
| | - Mariko Kasai
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Hiromi Yamamoto
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Motoo Watanabe
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Fabian Suchy
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Hideki Masaki
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Hiromitsu Nakauchi
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan.
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan.
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
36
|
Biggs LC, Miroshnikova YA. Nuclear mechanotransduction on skin stem cell fate regulation. Curr Opin Cell Biol 2024; 87:102328. [PMID: 38340567 DOI: 10.1016/j.ceb.2024.102328] [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: 11/01/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 02/12/2024]
Abstract
Mammalian skin is a highly dynamic and regenerative organ that has long been recognized as a mechanically active composite of tissues withstanding daily compressive and tensile forces that arise from body movement. Importantly, cell- and tissue-scale mechanical signals are critical regulators of skin morphogenesis and homeostasis. These signals are sensed at the cellular periphery and transduced by mechanosensitive proteins within the plasma membrane to the cytoskeletal networks, and eventually into the nucleus to regulate chromatin organization and gene expression. The role of each of these nodes in producing a coherent mechanoresponse at both cell- and tissue-scales is emerging. Here we focus on the key cytoplasmic and nuclear mechanosensitive structures that are critical for the mammalian skin development and homeostatic maintenance. We propose that the mechanical state of the skin, in particular of its nuclear compartment, is a critical rheostat that fine-tunes developmental and homeostatic processes essential for the proper function of the organ.
Collapse
Affiliation(s)
- Leah C Biggs
- Department of Cell and Tissue Dynamics, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany.
| | - Yekaterina A Miroshnikova
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
37
|
Soğukpınar M, Utine GE, Boduroğlu K, Şimşek-Kiper PÖ. A spectrum of TP63-related disorders with eight affected individuals in five unrelated families. Eur J Med Genet 2024; 68:104911. [PMID: 38281558 DOI: 10.1016/j.ejmg.2024.104911] [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: 01/31/2023] [Revised: 11/08/2023] [Accepted: 01/20/2024] [Indexed: 01/30/2024]
Abstract
TP63-related disdorders broadly involve varying combinations of ectodermal dysplasia (sparse hair, hypohydrosis, tooth abnormalities, nail dysplasia), cleft lip/palate, acromelic malformation, split-hand/foot malformation/syndactyly, ankyloblepharon filiforme adnatum, lacrimal duct obstruction, hypopigmentation, and hypoplastic breasts and/or nipples. TP63-related disorders are associated with heterozygous pathogenic variants in TP63 and include seven overlapping phenotypes; Ankyloblepharon-ectodermal defects-cleft lip/palate syndrome (AEC), Ectrodactyly-ectodermal dysplasia-cleft lip/palate syndrome 3 (EEC3), Limb-mammary syndrome (LMS), Acro-dermo-ungual-lacrimal-tooth syndrome (ADULT), Rapp-Hodgkin syndrome (RHS), Split-hand/foot malformation 4 (SHFM4), and Orofacial cleft 8. We report on five unrelated families with 8 affected individuals in which the probands presented with varying combinations of ectodermal dysplasia, cleft lip/palate, split-hand/foot malformation, lacrimal duct obstruction, and ankyloblepharon filiforme adnatum. The clinical diagnosis involved AEC syndrome (2 patients), EEC3 syndrome (2 patients), and a yet hitherto unclassified TP63-related disorder. Sanger sequence analysis of the TP63 gene was performed revealing five different variants among which four were novel and three were de novo. The identificated TP63 variants co-segregated with the other affected individuals in the families. The abnormalities of ectoderm derived structures including hair, nails, sweat glands, and teeth should alert the physician to the possibility of TP63-related disorders particularly in the presence of orofacial clefting.
Collapse
Affiliation(s)
- Merve Soğukpınar
- Department of Pediatric Genetics, Hacettepe University, Faculty of Medicine, Ankara, Turkey.
| | - Gülen Eda Utine
- Department of Pediatric Genetics, Hacettepe University, Faculty of Medicine, Ankara, Turkey
| | - Koray Boduroğlu
- Department of Pediatric Genetics, Hacettepe University, Faculty of Medicine, Ankara, Turkey
| | | |
Collapse
|
38
|
LeFever NM, Katreddi RR, Dolphin NM, Mathias NA, Forni PE. Following the p63/Keratin5 basal cells in the sensory and non-sensory epithelia of the vomeronasal organ. Genesis 2024; 62:e23596. [PMID: 38665067 PMCID: PMC11141727 DOI: 10.1002/dvg.23596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/27/2024] [Accepted: 03/19/2024] [Indexed: 05/01/2024]
Abstract
The vomeronasal organ (VNO) is a part of the accessory olfactory system, which detects pheromones and chemical factors that trigger a spectrum of sexual and social behaviors. The vomeronasal epithelium (VNE) shares several features with the epithelium of the main olfactory epithelium (MOE). However, it is a distinct neuroepithelium populated by chemosensory neurons that differ from the olfactory sensory neurons in cellular structure, receptor expression, and connectivity. The vomeronasal organ of rodents comprises a sensory epithelium (SE) and a thin non-sensory epithelium (NSE) that morphologically resembles the respiratory epithelium. Sox2-positive cells have been previously identified as the stem cell population that gives rise to neuronal progenitors in MOE and VNE. In addition, the MOE also comprises p63 positive horizontal basal cells, a second pool of quiescent stem cells that become active in response to injury. Immunolabeling against the transcription factor p63, Keratin-5 (Krt5), Krt14, NrCAM, and Krt5Cre tracing experiments highlighted the existence of horizontal basal cells distributed along the basal lamina of SE of the VNO. Single cell sequencing and genetic lineage tracing suggest that the vomeronasal horizontal basal cells arise from basal progenitors at the boundary between the SE and NSE proximal to the marginal zones. Moreover, our experiments revealed that the NSE of rodents is, like the respiratory epithelium, a stratified epithelium where the p63/Krt5+ basal progenitor cells self-replicate and give rise to the apical columnar cells facing the lumen of the VNO.
Collapse
Affiliation(s)
| | | | | | | | - Paolo E. Forni
- Department of Biological Sciences
- The RNA Institute
- The Center for Neuroscience Research, University at Albany, State University of New York, Albany, NY, United States
| |
Collapse
|
39
|
Fan Y, Chen S, Chu C, Yin X, Jin J, Zhang L, Yan H, Cao Z, Liu R, Xin M, Li L, Yin C. TP63 truncating mutation causes increased cell apoptosis and premature ovarian insufficiency by enhanced transcriptional activation of CLCA2. J Ovarian Res 2024; 17:67. [PMID: 38528613 PMCID: PMC10962206 DOI: 10.1186/s13048-024-01396-2] [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: 08/31/2023] [Accepted: 03/18/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND Premature ovarian insufficiency (POI) is a severe disorder leading to female infertility. Genetic mutations are important factors causing POI. TP63-truncating mutation has been reported to cause POI by increasing germ cell apoptosis, however what factors mediate this apoptosis remains unclear. METHODS Ninety-three patients with POI were recruited from Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Whole-exome sequencing (WES) was performed for each patient. Sanger sequencing was used to confirm potential causative genetic variants. A minigene assay was performed to determine splicing effects of TP63 variants. A TP63-truncating plasmid was constructed. Real-time quantitative PCR, western blot analyses, dual luciferase reporter assays, immunofluorescence staining, and cell apoptosis assays were used to study the underlying mechanism of a TP63-truncating mutation causing POI. RESULTS By WES of 93 sporadic patients with POI, we found a 14-bp deletion covering the splice site in the TP63 gene. A minigene assay demonstrated that the 14-bp deletion variant led to exon 13 skipping during TP63 mRNA splicing, resulting in the generation of a truncated TP63 protein (TP63-mut). Overexpression of TP63-mut accelerated cell apoptosis. Mechanistically, the TP63-mut protein could bind to the promoter region of CLCA2 and activate the transcription of CLCA2 several times compared to that of the TP63 wild-type protein. Silencing CLCA2 using a specific small interfering RNA (siRNA) or inhibiting the Ataxia Telangiectasia Mutated (ATM) pathway using the KU55933 inhibitor attenuated cell apoptosis caused by TP63-mut protein expression. CONCLUSION Our findings revealed a crucial role for CLCA2 in mediating apoptosis in POI pathogenesis, and suggested that CLCA2 is a potential therapeutic target for POI.
Collapse
Affiliation(s)
- Yali Fan
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100006, China
| | - Shuya Chen
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100006, China
| | - Chunfang Chu
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Xiaodan Yin
- Department of Traditional Chinese Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Jing Jin
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Lingyan Zhang
- Department of Gynaecology and Obstetrics, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Huihui Yan
- Department of Obstetrics, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Zheng Cao
- Department of Laboratory Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Ruixia Liu
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100006, China
| | - Mingwei Xin
- Department of Traditional Chinese Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China.
| | - Lin Li
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100006, China.
| | - Chenghong Yin
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100006, China.
| |
Collapse
|
40
|
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
|
41
|
Wang Y, Meng Z, Liu M, Zhou Y, Chen D, Zhao Y, Zhang T, Zhong N, Dai X, Li S, Zuo W. Autologous transplantation of P63 + lung progenitor cells for chronic obstructive pulmonary disease therapy. Sci Transl Med 2024; 16:eadi3360. [PMID: 38354225 DOI: 10.1126/scitranslmed.adi3360] [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: 04/20/2023] [Accepted: 01/25/2024] [Indexed: 02/16/2024]
Abstract
Adult lung resident stem/progenitor cells, including P63+ progenitor cells, have demonstrated the capacity for regeneration of lung epithelium in preclinical models. Here, we report a clinical trial of intrapulmonary P63+ progenitor cell transplantation in 28 participants with stage II to IV chronic obstructive pulmonary disease (COPD). Autologous P63+ progenitor cells were isolated from the airway basal layer of participants in the intervention group via bronchoscopic brushing, cultured for 3 to 5 weeks, and then transplanted back into the lungs via bronchoscopy at 0.7 × 106 to 5.2 × 106 cells per kilogram of body weight. Twenty patients were evaluable at the end of the study (intervention group, n = 17; control group, n = 3). No grade 3 to 5 adverse events (AEs) or serious AEs occurred. Although bronchoscopy-associated AEs were recorded in participants in the intervention group, other AEs were not substantial different between groups. Twenty-four weeks after transplantation, participants in the intervention group displayed improvement in gas transfer capacity [diffusing capacity of the lung for carbon monoxide (DLCO) change from baseline: +18.2%], whereas the control group experienced a decrease (DLCO change from baseline: -17.4%; P = 0.008). Furthermore, participants in the intervention group showed >30-meter increase in walking distance within 6 minutes. Transcriptomic analysis of progenitor cells isolated from responding and nonresponding individuals in the intervention group showed that higher expression of P63 was associated with treatment efficacy. In conclusion, transplantation of cultured P63+ lung progenitor cells was safe and might represent a potential therapeutic strategy for COPD.
Collapse
Affiliation(s)
- Yujia Wang
- Department of Organ Regeneration, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Zili Meng
- Department of Respiratory and Critical Care Medicine, Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an 223300, China
| | - Ming Liu
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou 510120, China
| | - Yueqing Zhou
- Department of Organ Regeneration, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Super Organ R&D Center, Regend Therapeutics, Shanghai 201318, China
| | - Difei Chen
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou 510120, China
| | - Yu Zhao
- Department of Organ Regeneration, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Ting Zhang
- Super Organ R&D Center, Regend Therapeutics, Shanghai 201318, China
| | - Nanshan Zhong
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou 510120, China
| | - Xiaotian Dai
- Southwest Hospital, Third Military Medical University of PLA, Chongqing 400038, China
| | - Shiyue Li
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou 510120, China
| | - Wei Zuo
- Department of Organ Regeneration, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Super Organ R&D Center, Regend Therapeutics, Shanghai 201318, China
- West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
42
|
Hand AR, Abramson CXG, Dressler KA. Tlx1 regulates acinar and duct development in mouse salivary glands. J Anat 2024; 244:343-357. [PMID: 37837237 PMCID: PMC10780161 DOI: 10.1111/joa.13964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/05/2023] [Accepted: 10/05/2023] [Indexed: 10/15/2023] Open
Abstract
Tlx1 encodes a transcription factor expressed in several craniofacial structures of developing mice. The role of Tlx1 in salivary gland development was examined using morphological and immunohistochemical analyses of Tlx1 null mice. Tlx1 is expressed in submandibular and sublingual glands but not parotid glands of neonatal and adult male and female C57Bl/6J (Tlx1+/+ ) mice. TLX1 protein was localized to the nuclei of terminal tubule cells, developing duct cells and mesenchymal cells in neonatal submandibular and sublingual glands, and to nuclei of duct cells and connective tissue cells in adult glands. Occasionally, TLX1 was observed in nuclei of epithelial cells in or adjacent to the acini. Submandibular glands were smaller and sublingual glands were larger in size in mutant mice (Tlx1-/- ) compared to wild-type mice. Differentiation of terminal tubule and proacinar cells of neonatal Tlx1-/- submandibular glands was abnormal; expression of their characteristic products, submandibular gland protein C and parotid secretory protein, respectively, was reduced. At 3 weeks postnatally, terminal tubule cells at the acinar-intercalated duct junction were poorly developed or absent in Tlx1-/- mice. Granular convoluted ducts in adult mutant mice were decreased, and epidermal growth factor and nerve growth factor expression were reduced. Along with normal acinar cell proteins, adult acinar cells of Tlx1-/- mice continued to express neonatal proteins and expressed parotid proteins not normally present in submandibular glands. Sublingual gland mucous acinar and serous demilune cell differentiation were altered. Tlx1 is necessary for proper differentiation of submandibular and sublingual gland acinar cells, and granular convoluted ducts. The mechanism(s) underlying Tlx1 regulation of salivary gland development and differentiation remains unknown.
Collapse
Affiliation(s)
- Arthur R Hand
- Department of Craniofacial Sciences, University of Connecticut School of Dental Medicine, Farmington, Connecticut, USA
| | - Cailyn X G Abramson
- Department of Craniofacial Sciences, University of Connecticut School of Dental Medicine, Farmington, Connecticut, USA
| | - Keith A Dressler
- Department of Craniofacial Sciences, University of Connecticut School of Dental Medicine, Farmington, Connecticut, USA
| |
Collapse
|
43
|
Napoli M, Deshpande AA, Chakravarti D, Rajapakshe K, Gunaratne PH, Coarfa C, Flores ER. Genome-wide p63-Target Gene Analyses Reveal TAp63/NRF2-Dependent Oxidative Stress Responses. CANCER RESEARCH COMMUNICATIONS 2024; 4:264-278. [PMID: 38165157 PMCID: PMC10832605 DOI: 10.1158/2767-9764.crc-23-0358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 11/14/2023] [Accepted: 12/27/2023] [Indexed: 01/03/2024]
Abstract
The p53 family member TP63 encodes two sets of N-terminal isoforms, TAp63 and ΔNp63 isoforms. They each regulate diverse biological functions in epidermal morphogenesis and in cancer. In the skin, where their activities have been extensively characterized, TAp63 prevents premature aging by regulating the quiescence and genomic stability of stem cells required for wound healing and hair regeneration, while ΔNp63 controls maintenance and terminal differentiation of epidermal basal cells. This functional diversity is surprising given that these isoforms share a high degree of similarity, including an identical sequence for a DNA-binding domain. To understand the mechanisms of the transcriptional programs regulated by each p63 isoform and leading to diverse biological functions, we performed genome-wide analyses using p63 isoform-specific chromatin immunoprecipitation, RNA sequencing, and metabolomics of TAp63-/- and ΔNp63-/- mouse epidermal cells. Our data indicate that TAp63 and ΔNp63 physically and functionally interact with distinct transcription factors for the downstream regulation of their target genes, thus ultimately leading to the regulation of unique transcriptional programs and biological processes. Our findings unveil novel transcriptomes regulated by the p63 isoforms to control diverse biological functions, including the cooperation between TAp63 and NRF2 in the modulation of metabolic pathways and response to oxidative stress providing a mechanistic explanation for the TAp63 knock out phenotypes. SIGNIFICANCE The p63 isoforms, TAp63 and ΔNp63, control epithelial morphogenesis and tumorigenesis through the interaction with distinct transcription factors and the subsequent regulation of unique transcriptional programs.
Collapse
Affiliation(s)
- Marco Napoli
- Department of Molecular Oncology, Division of Basic Science, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Avani A. Deshpande
- Department of Molecular Oncology, Division of Basic Science, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | | | - Kimal Rajapakshe
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | | | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Elsa R. Flores
- Department of Molecular Oncology, Division of Basic Science, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| |
Collapse
|
44
|
Lyu Q, Li Q, Zhou J, Zhao H. Formation and function of multiciliated cells. J Cell Biol 2024; 223:e202307150. [PMID: 38032388 PMCID: PMC10689204 DOI: 10.1083/jcb.202307150] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/29/2023] [Accepted: 11/14/2023] [Indexed: 12/01/2023] Open
Abstract
In vertebrates, multiciliated cells (MCCs) are terminally differentiated cells that line the airway tracts, brain ventricles, and reproductive ducts. Each MCC contains dozens to hundreds of motile cilia that beat in a synchronized manner to drive fluid flow across epithelia, the dysfunction of which is associated with a group of human diseases referred to as motile ciliopathies, such as primary cilia dyskinesia. Given the dynamic and complex process of multiciliogenesis, the biological events essential for forming multiple motile cilia are comparatively unelucidated. Thanks to advancements in genetic tools, omics technologies, and structural biology, significant progress has been achieved in the past decade in understanding the molecular mechanism underlying the regulation of multiple motile cilia formation. In this review, we discuss recent studies with ex vivo culture MCC and animal models, summarize current knowledge of multiciliogenesis, and particularly highlight recent advances and their implications.
Collapse
Affiliation(s)
- Qian Lyu
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Qingchao Li
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Jun Zhou
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, College of Life Sciences, Nankai University, Tianjin, China
| | - Huijie Zhao
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| |
Collapse
|
45
|
Low-Calle AM, Ghoneima H, Ortega N, Cuibus AM, Katz C, Prives C, Prywes R. A Non-Canonical Hippo Pathway Represses the Expression of ΔNp63. Mol Cell Biol 2024; 44:27-42. [PMID: 38270135 PMCID: PMC10829837 DOI: 10.1080/10985549.2023.2292037] [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: 07/13/2023] [Accepted: 11/28/2023] [Indexed: 01/26/2024] Open
Abstract
The p63 transcription factor, a member of the p53 family, plays an oncogenic role in squamous cell carcinomas, while in breast cancers its expression is often repressed. In the canonical conserved Hippo pathway, known to play a complex role in regulating growth of cancer cells, protein kinases MST1/2 and LATS1/2 act sequentially to phosphorylate and inhibit the YAP/TAZ transcription factors. We found that in MCF10A mammary epithelial cells as well as in squamous and breast cancer cell lines, expression of ΔNp63 RNA and protein is strongly repressed by inhibition of the Hippo pathway protein kinases. While MST1/2 and LATS1 are required for p63 expression, the next step of the pathway, namely phosphorylation and degradation of the YAP/TAZ transcriptional activators is not required for p63 repression. This suggests that regulation of p63 expression occurs by a noncanonical version of the Hippo pathway. We identified similarly regulated genes, suggesting the broader importance of this pathway. Interestingly, lowering p63 expression lead to increased YAP protein levels, indicating crosstalk of the YAP/TAZ-independent and -dependent branches of the Hippo pathway. These results, which reveal the intersection of the Hippo and p63 pathways, may prove useful for the control of their activities in cancer cells.
Collapse
Affiliation(s)
- Ana Maria Low-Calle
- Department of Biological Sciences, Columbia University, New York, New York, USA
| | - Hana Ghoneima
- Department of Biological Sciences, Columbia University, New York, New York, USA
| | - Nicholas Ortega
- Department of Biological Sciences, Columbia University, New York, New York, USA
| | - Adriana M. Cuibus
- Department of Biological Sciences, Columbia University, New York, New York, USA
| | - Chen Katz
- Department of Biological Sciences, Columbia University, New York, New York, USA
| | - Carol Prives
- Department of Biological Sciences, Columbia University, New York, New York, USA
| | - Ron Prywes
- Department of Biological Sciences, Columbia University, New York, New York, USA
| |
Collapse
|
46
|
Ramal M, Corral S, Kalisz M, Lapi E, Real FX. The urothelial gene regulatory network: understanding biology to improve bladder cancer management. Oncogene 2024; 43:1-21. [PMID: 37996699 DOI: 10.1038/s41388-023-02876-3] [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: 07/25/2023] [Revised: 10/13/2023] [Accepted: 10/18/2023] [Indexed: 11/25/2023]
Abstract
The urothelium is a stratified epithelium composed of basal cells, one or more layers of intermediate cells, and an upper layer of differentiated umbrella cells. Most bladder cancers (BLCA) are urothelial carcinomas. Loss of urothelial lineage fidelity results in altered differentiation, highlighted by the taxonomic classification into basal and luminal tumors. There is a need to better understand the urothelial transcriptional networks. To systematically identify transcription factors (TFs) relevant for urothelial identity, we defined highly expressed TFs in normal human bladder using RNA-Seq data and inferred their genomic binding using ATAC-Seq data. To focus on epithelial TFs, we analyzed RNA-Seq data from patient-derived organoids recapitulating features of basal/luminal tumors. We classified TFs as "luminal-enriched", "basal-enriched" or "common" according to expression in organoids. We validated our classification by differential gene expression analysis in Luminal Papillary vs. Basal/Squamous tumors. Genomic analyses revealed well-known TFs associated with luminal (e.g., PPARG, GATA3, FOXA1) and basal (e.g., TP63, TFAP2) phenotypes and novel candidates to play a role in urothelial differentiation or BLCA (e.g., MECOM, TBX3). We also identified TF families (e.g., KLFs, AP1, circadian clock, sex hormone receptors) for which there is suggestive evidence of their involvement in urothelial differentiation and/or BLCA. Genomic alterations in these TFs are associated with BLCA. We uncover a TF network involved in urothelial cell identity and BLCA. We identify novel candidate TFs involved in differentiation and cancer that provide opportunities for a better understanding of the underlying biology and therapeutic intervention.
Collapse
Affiliation(s)
- Maria Ramal
- Epithelial Carcinogenesis Group, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Sonia Corral
- Epithelial Carcinogenesis Group, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Mark Kalisz
- Epithelial Carcinogenesis Group, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- CIBERONC, Madrid, Spain
| | - Eleonora Lapi
- Epithelial Carcinogenesis Group, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- CIBERONC, Madrid, Spain
| | - Francisco X Real
- Epithelial Carcinogenesis Group, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.
- CIBERONC, Madrid, Spain.
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain.
| |
Collapse
|
47
|
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
|
48
|
Kim HY, Cooley V, Kim EJ, Li S, Lee JM, Sheyfer D, Liu W, Klein OD, Joester D, Jung HS. Adult dental epithelial stem cell-derived organoids deposit hydroxylapatite biomineral. Int J Oral Sci 2023; 15:55. [PMID: 38062012 PMCID: PMC10703793 DOI: 10.1038/s41368-023-00257-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 11/02/2023] [Accepted: 11/02/2023] [Indexed: 12/18/2023] Open
Abstract
Ameloblasts are specialized cells derived from the dental epithelium that produce enamel, a hierarchically structured tissue comprised of highly elongated hydroxylapatite (OHAp) crystallites. The unique function of the epithelial cells synthesizing crystallites and assembling them in a mechanically robust structure is not fully elucidated yet, partly due to limitations with in vitro experimental models. Herein, we demonstrate the ability to generate mineralizing dental epithelial organoids (DEOs) from adult dental epithelial stem cells (aDESCs) isolated from mouse incisor tissues. DEOs expressed ameloblast markers, could be maintained for more than five months (11 passages) in vitro in media containing modulators of Wnt, Egf, Bmp, Fgf and Notch signaling pathways, and were amenable to cryostorage. When transplanted underneath murine kidney capsules, organoids produced OHAp crystallites similar in composition, size, and shape to mineralized dental tissues, including some enamel-like elongated crystals. DEOs are thus a powerful in vitro model to study mineralization process by dental epithelium, which can pave the way to understanding amelogenesis and developing regenerative therapy of enamel.
Collapse
Affiliation(s)
| | - Victoria Cooley
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA
- X-ray Science Division, Advanced Photon Source, Argonne National Laboratory, Lemont, IL, USA
| | - Eun-Jung Kim
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Oral Science Research Center, BK21 FOUR Project, Yonsei University College of Dentistry, Seoul, Korea
| | - Shujin Li
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Oral Science Research Center, BK21 FOUR Project, Yonsei University College of Dentistry, Seoul, Korea
| | - Jong-Min Lee
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Oral Science Research Center, BK21 FOUR Project, Yonsei University College of Dentistry, Seoul, Korea
| | - Dina Sheyfer
- X-ray Science Division, Advanced Photon Source, Argonne National Laboratory, Lemont, IL, USA
| | - Wenjun Liu
- X-ray Science Division, Advanced Photon Source, Argonne National Laboratory, Lemont, IL, USA
| | - Ophir D Klein
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, CA, USA
- Department of Pediatrics, Cedars-Sinai Guerin Children's, Los Angeles, CA, USA
| | - Derk Joester
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA
| | - Han-Sung Jung
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Oral Science Research Center, BK21 FOUR Project, Yonsei University College of Dentistry, Seoul, Korea.
| |
Collapse
|
49
|
Della Salda L, Bongiovanni L, Massimini M, Romanucci M, Vercelli A, Colosimo A, Di Matteo R, Defourny SVP. p63 immunoexpression in hair follicles of normal and alopecia X-affected skin of Pomeranian dogs. Vet Dermatol 2023; 34:567-575. [PMID: 37518946 DOI: 10.1111/vde.13195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 05/10/2023] [Accepted: 07/14/2023] [Indexed: 08/01/2023]
Abstract
BACKGROUND Alopecia X in Pomeranians is caused by a hair cycle deregulation, associated with downregulation of key regulatory genes of the Wnt and Shh pathways, and stem-cell markers. However, the pathogenesis remains unclear. p63 is an important transcription factor correlated with the aforementioned hair cycle modulating genes. HYPOTHESIS/OBJECTIVES The aim of this study was to highlight possible changes of p63 immunohistochemical expression within the hair follicles in canine alopecia X compared with normal skin. ANIMALS Skin biopsies from 19 alopecia X-affected and six control Pomeranians were analysed. MATERIALS AND METHODS Serial histological sections of skin biopsies harbouring anagen, telogen and kenogen hair follicles were immunohistochemically evaluated for differences in p63 expression in the affected and control samples. RESULTS Dogs with alopecia X had a significantly decreased immunoexpression of p63 in telogen and kenogen hair follicles. CONCLUSIONS AND CLINICAL RELEVANCE The decrease of p63 immunoexpression observed in canine alopecia X suggests an involvement of p63 in hair cycle.
Collapse
Affiliation(s)
| | - Laura Bongiovanni
- Department of Veterinary Medicine, University of Teramo, Teramo, Italy
- Department of Biomolecular Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | | | | | - Antonella Vercelli
- Veterinary Clinic and Analysis Laboratory 'Città di Torino', Turin, Italy
| | - Alessia Colosimo
- Department of Bioscience and Agro-Food and Environmental Technology, University of Teramo, Teramo, Italy
| | - Ramona Di Matteo
- Department of Veterinary Medicine, University of Teramo, Teramo, Italy
| | | |
Collapse
|
50
|
Abstract
Keratinocyte senescence contributes to skin ageing and epidermal dysfunction. According to the existing knowledge, the transcription factor ΔNp63α plays pivotal roles in differentiation and proliferation of keratinocytes. It is traditionally accepted that ΔNp63α exerts its functions via binding to promoter regions to activate or repress gene transcription. However, accumulating evidence demonstrates that ΔNp63α can bind to elements away from promoter regions of its target genes, mediating epigenetic regulation. On the other hand, several epigenetic alterations, including DNA methylation, histone modification and variation, chromatin remodelling, as well as enhancer-promoter looping, are found to be related to cell senescence. To systematically elucidate how ΔNp63α affects keratinocyte senescence via epigenetic regulation, we comprehensively compiled the literatures on the roles of ΔNp63α in keratinocyte senescence, epigenetics in cellular senescence, and the relation between ΔNp63α-mediated epigenetic regulation and keratinocyte senescence. Based on the published data, we conclude that ΔNp63α mediates epigenetic regulation via multiple mechanisms: recruiting epigenetic enzymes to modify DNA or histones, coordinating chromatin remodelling complexes (CRCs) or regulating their expression, and mediating enhancer-promoter looping. Consequently, the expression of genes related to cell cycle is modulated, and proliferation of keratinocytes and renewal of stem cells are maintained, by ΔNp63α. During skin inflammaging, the decline of ΔNp63α may lead to epigenetic dysregulation, resultantly deteriorating keratinocyte senescence.
Collapse
Affiliation(s)
- Linghan Kuang
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu 610041, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| | - Chenghua Li
- Center of Growth, Metabolism and Aging, Key Laboratory of Biological Resources and Ecological Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, China
| |
Collapse
|