1
|
Quan H, Zhou H, Chen F, Chen J, He Y, Xiao H, Liu J, Shi L, Xie W, Chen P, Luo J. Acetylated KIAA1429 by TIP60 facilitates metastasis and immune evasion of hepatocellular carcinoma via N6-methyladenosine-KDM5B-mediated regulation of FoxO1. Cell Death Discov 2025; 11:210. [PMID: 40301310 PMCID: PMC12041376 DOI: 10.1038/s41420-025-02462-4] [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: 09/25/2024] [Revised: 03/22/2025] [Accepted: 03/28/2025] [Indexed: 05/01/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is characterized by programmed cell death ligand-1 (PD-L1)-mediated immune escape. This study aimed to elucidate the function and mechanism behind KIAA1429, a component of N6-methyladenosine (m6A) complex, in immune escape of HCC. PD-L1 expression was assessed through immunofluorescence staining, and flow cytometry was used to determine CD8+ T cell percentage. The level of IFN-γ was detected using enzyme-linked immunosorbent assay. Cell proliferation, migration, and invasion were evaluated through CCK-8, colony formation, and Transwell assays, respectively. The m6A modification level was measured using an RNA methylation quantification assay, m6A dot blot, and methylated RNA immunoprecipitation-qPCR. Molecule interaction was validated using RNA pulldown, RNA immunoprecipitation, chromatin immunoprecipitation, and co-immunoprecipitation assays. In vivo HCC growth was evaluated in NOD/SCID mice. We found that TIP60, KIAA1429 and KDM5B were highly expressed in HCC cells, while FoxO1 was poorly expressed. Functionally, TIP60/KIAA1429 silencing inhibited PD-L1-mediated HCC immune evasion, growth, migration, and invasion. Mechanistically, TIP60 led to acetylation of KIAA1429, which promoted KDM5B expression in an m6A-YTHDF1-dependent manner, and subsequently restrained the transcription and expression of FoxO1. Enforcing YTHDF1 expression or depleting FoxO1 expression markedly reversed the suppressive effect of shKIAA1429 on HCC immune evasion, growth, migration, and invasion. Overall, these findings suggest that acetylated KIAA1429-mediated m6A modification endows HCC cells with immune evasion through regulation of KDM5B/FoxO1 axis, which provide a treatment option for HCC by targeting KIAA1429.
Collapse
Affiliation(s)
- Hu Quan
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, P.R. China
- Department of general Surgery, Turpan City People's Hospital, Tulufan, 838000, Xinjiang, P.R. China
| | - Huijun Zhou
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, P.R. China
| | - Fei Chen
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, P.R. China
| | - Jie Chen
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, P.R. China
| | - Yun He
- The Central Hospital of Shaoyang, The Affiliated Shaoyang Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan Province, P.R. China
| | - Hua Xiao
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, P.R. China
| | - Jia Liu
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, P.R. China
| | - Lei Shi
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, P.R. China
| | - Wei Xie
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, P.R. China.
| | - Pan Chen
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, P.R. China.
| | - Jia Luo
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, P.R. China.
| |
Collapse
|
2
|
Dabin LC, Kersey H, Kim B, Acri DJ, Sharify D, Lee‐Gosselin A, Lasagna‐Reeves CA, Oblak AL, Lamb BT, Kim J. Loss of Inpp5d has disease-relevant and sex-specific effects on glial transcriptomes. Alzheimers Dement 2024; 20:5311-5323. [PMID: 38923164 PMCID: PMC11350029 DOI: 10.1002/alz.13901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 06/28/2024]
Abstract
INTRODUCTION Inpp5d is genetically associated with Alzheimer's disease risk. Loss of Inpp5d alters amyloid pathology in models of amyloidosis. Inpp5d is expressed predominantly in microglia but its function in brain is poorly understood. METHODS We performed single-cell RNA sequencing to study the effect of Inpp5d loss on wild-type mouse brain transcriptomes. RESULTS Loss of Inpp5d has sex-specific effects on the brain transcriptome. Affected genes are enriched for multiple neurodegeneration terms. Network analyses reveal a gene co-expression module centered around Inpp5d in female mice. Inpp5d loss alters Pleotrophin (PTN), Prosaposin (PSAP), and Vascular Endothelial Growth Factor A (VEGFA) signaling probability between cell types. DISCUSSION Our data suggest that the normal function of Inpp5d is entangled with mechanisms involved in neurodegeneration. We report the effect of Inpp5d loss without pathology and show that this has dramatic effects on gene expression. Our study provides a critical reference for researchers of neurodegeneration, allowing separation of disease-specific changes mediated by Inpp5d in disease from baseline effects of Inpp5d loss. HIGHLIGHTS Loss of Inpp5d has different effects in male and female mice. Genes dysregulated by Inpp5d loss relate to neurodegeneration. Total loss of Inpp5d in female mice collapses a conserved gene co-expression module. Loss of microglial Inpp5d affects the transcriptome of other cell types.
Collapse
Affiliation(s)
- Luke C. Dabin
- Department of Medical & Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
- Stark Neuroscience Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
| | - Holly Kersey
- Department of Medical & Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
- Stark Neuroscience Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
- Medical Neuroscience Graduate ProgramIndiana University School of MedicineIndianapolisIndianaUSA
| | - Byungwook Kim
- Department of Medical & Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
- Stark Neuroscience Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
| | - Dominic J. Acri
- Department of Medical & Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
- Stark Neuroscience Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
- Medical Neuroscience Graduate ProgramIndiana University School of MedicineIndianapolisIndianaUSA
| | - Daniel Sharify
- Department of Medical & Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
- Stark Neuroscience Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
| | - Audrey Lee‐Gosselin
- Stark Neuroscience Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
| | - Cristian A. Lasagna‐Reeves
- Stark Neuroscience Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
- Medical Neuroscience Graduate ProgramIndiana University School of MedicineIndianapolisIndianaUSA
- Department of AnatomyCell Biology & PhysiologyIndiana University School of MedicineIndianapolisIndianaUSA
- Center for Computational Biology and BioinformaticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Adrian L. Oblak
- Stark Neuroscience Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
- Medical Neuroscience Graduate ProgramIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | - Bruce T. Lamb
- Department of Medical & Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
- Stark Neuroscience Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
- Medical Neuroscience Graduate ProgramIndiana University School of MedicineIndianapolisIndianaUSA
| | - Jungsu Kim
- Department of Medical & Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
- Stark Neuroscience Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
- Medical Neuroscience Graduate ProgramIndiana University School of MedicineIndianapolisIndianaUSA
| |
Collapse
|
3
|
Epigenetic Mechanisms of Epidermal Differentiation. Int J Mol Sci 2022; 23:ijms23094874. [PMID: 35563264 PMCID: PMC9102508 DOI: 10.3390/ijms23094874] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/24/2022] [Accepted: 04/27/2022] [Indexed: 12/12/2022] Open
Abstract
Keratinocyte differentiation is an essential process for epidermal stratification and stratum corneum formation. Keratinocytes proliferate in the basal layer of the epidermis and start their differentiation by changing their functional or phenotypical type; this process is regulated via induction or repression of epidermal differentiation complex (EDC) genes that play a pivotal role in epidermal development. Epidermal development and the keratinocyte differentiation program are orchestrated by several transcription factors, signaling pathways, and epigenetic regulators. The latter exhibits both activating and repressive effects on chromatin in keratinocytes via the ATP-dependent chromatin remodelers, histone demethylases, and genome organizers that promote terminal keratinocyte differentiation, and the DNA methyltransferases, histone deacetylases, and Polycomb components that stimulate proliferation of progenitor cells and inhibit premature activation of terminal differentiation-associated genes. In addition, microRNAs are involved in different processes between proliferation and differentiation during the program of epidermal development. Here, we bring together current knowledge of the mechanisms controlling gene expression during keratinocyte differentiation. An awareness of epigenetic mechanisms and their alterations in health and disease will help to bridge the gap between our current knowledge and potential applications for epigenetic regulators in clinical practice to pave the way for promising target therapies.
Collapse
|
4
|
Yao T, Xu Z, Hao Z, Yu Y, Liang B, Wang S. KDM5B promotes cell migration by regulating the noncanonical Wnt/PCP pathway in Hirschsprung's disease. Pediatr Surg Int 2022; 38:99-107. [PMID: 34455465 DOI: 10.1007/s00383-021-05005-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/23/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE We measured the expression of the histone demethylase lysine-specific demethylase 5B (KDM5B) in the bowels of patients with Hirschsprung's disease (HSCR) and investigated the molecular mechanism by which KDM5B promotes the migration of neuronal PC12 cells. METHODS KDM5B expression was detected in the ganglionic and aganglionic colon of patients with HSCR (n = 10) and controls (n = 10). The expression and localization of KDM5B were assessed using immunohistochemical and immunofluorescence staining. Real-time PCR and Western blotting were performed to quantify KDM5B expression. The migration was determined using Transwell and wound-healing assays. G-LISA, GTPase pulldown and luciferase-based reporter gene assays were performed to evaluate the key components of Wnt/planar cell polarity (PCP) signaling in vitro. RESULTS Our current study showed that KDM5B colocalized with neurons. KDM5B expression was reduced in HSCR specimens, while the aganglionic segments showed the greatest reduction. KDM5B knockdown inhibited the migration of PC12 cells. Moreover, inhibition of KDM5B decreased the expression of key genes in the Wnt/PCP pathway, and its inhibitory effect on PC12 cell migration was reversed by Wnt5a treatment. CONCLUSIONS KDM5B promotes neuronal migration via the Wnt/PCP pathway. A potential role for KDM5B in altered enteric nervous system development in HSCR warrants further investigation.
Collapse
Affiliation(s)
- Ting Yao
- Department of Pediatric Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Zhilin Xu
- Department of Pediatric Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Zenghui Hao
- Department of Pediatric Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
| | - You Yu
- Department of Pediatric Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Bingxue Liang
- Department of Pediatric Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China
| | - Shuyu Wang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China.
| |
Collapse
|
5
|
Imodoye SO, Adedokun KA, Muhammed AO, Bello IO, Muhibi MA, Oduola T, Oyenike MA. Understanding the Complex Milieu of Epithelial-Mesenchymal Transition in Cancer Metastasis: New Insight Into the Roles of Transcription Factors. Front Oncol 2021; 11:762817. [PMID: 34868979 PMCID: PMC8636732 DOI: 10.3389/fonc.2021.762817] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 10/04/2021] [Indexed: 12/12/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a physiological program during which polarised, immobile epithelial cells lose connection with their neighbours and are converted to migratory mesenchymal phenotype. Mechanistically, EMT occurs via a series of genetic and cellular events leading to the repression of epithelial-associated markers and upregulation of mesenchymal-associated markers. EMT is very crucial for many biological processes such as embryogenesis and ontogenesis during human development, and again it plays a significant role in wound healing during a programmed replacement of the damaged tissues. However, this process is often hijacked in pathological conditions such as tumour metastasis, which constitutes the most significant drawback in the fight against cancer, accounting for about 90% of cancer-associated mortality globally. Worse still, metastatic tumours are not only challenging to treat with the available conventional radiotherapy and surgical interventions but also resistant to several cytotoxic agents during treatment, owing to their anatomically diffuse localisation in the body system. As the quest to find an effective method of addressing metastasis in cancer intervention heightens, understanding the molecular interplay involving the signalling pathways, downstream effectors, and their interactions with the EMT would be an important requisite while the challenges of metastasis continue to punctuate. Unfortunately, the molecular underpinnings that govern this process remain to be completely illuminated. However, it is becoming increasingly clear that EMT, which initiates every episode of metastasis, significantly requires some master regulators called EMT transcription factors (EMT-TFs). Thus, this review critically examines the roles of TFs as drivers of molecular rewiring that lead to tumour initiation, progression, EMT, metastasis, and colonisation. In addition, it discusses the interaction of various signalling molecules and effector proteins with these factors. It also provides insight into promising therapeutic targets that may inhibit the metastatic process to overcome the limitation of "undruggable" cancer targets in therapeutic design and upturn the current spate of drug resistance. More so, it extends the discussion from the basic understanding of the EMT binary switch model, and ultimately unveiling the E/M cellular plasticity along a phenotypic spectrum via multiple trans-differentiations. It wraps up on how this knowledge update shapes the diagnostic and clinical approaches that may demand a potential shift in investigative paradigm using novel technologies such as single-cell analyses to improve overall patient survival.
Collapse
Affiliation(s)
- Sikiru O. Imodoye
- Department of Medical Laboratory Science, College of Medicine, University of Lagos, Lagos, Nigeria
| | - Kamoru A. Adedokun
- Department of Oral Pathology, Dental University Hospital, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Abdurrasheed Ola Muhammed
- Department of Histopathology, School of Medical Laboratory Science, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Ibrahim O. Bello
- Department of Biological Sciences, Southern Illinois University, Edwardsville, IL, United States
| | - Musa A. Muhibi
- Department of Medical Laboratory Science, Faculty of Applied Sciences, Edo State University, Uzairue, Nigeria
| | - Taofeeq Oduola
- Department of Chemical Pathology, School of Medical Laboratory Sciences, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Musiliu A. Oyenike
- Department of Medical Laboratory Science, Ladoke Akintola University of Technology, Ogbomosho, Nigeria
| |
Collapse
|
6
|
Leśniak W. Epigenetic Regulation of Epidermal Differentiation. EPIGENOMES 2021; 5:1. [PMID: 34968254 PMCID: PMC8594726 DOI: 10.3390/epigenomes5010001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/19/2020] [Accepted: 12/23/2020] [Indexed: 01/22/2023] Open
Abstract
The epidermis is the outer part of the skin that protects the organism from dehydration and shields from external insults. Epidermal cells, called keratinocytes, undergo a series of morphological and metabolic changes that allow them to establish the biochemical and structural elements of an effective epidermal barrier. This process, known as epidermal differentiation, is critical for the maintenance of the epidermis under physiological conditions and also under stress or in various skin pathologies. Epidermal differentiation relies on a highly coordinated program of gene expression. Epigenetic mechanisms, which commonly include DNA methylation, covalent histone modifications, and microRNA (miRNA) activity, modulate various stages of gene expression by altering chromatin accessibility and mRNA stability. Their involvement in epidermal differentiation is a matter of intensive studies, and the results obtained thus far show a complex network of epigenetic factors, acting together with transcriptional regulators, to maintain epidermal homeostasis and counteract adverse effects of environmental stressors.
Collapse
Affiliation(s)
- Wiesława Leśniak
- Laboratory of Calcium Binding Proteins, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| |
Collapse
|
7
|
Huang D, Xiao F, Hao H, Hua F, Luo Z, Huang Z, Li Q, Chen S, Cheng X, Zhang X, Fang W, Hu X, Liu F. JARID1B promotes colorectal cancer proliferation and Wnt/β-catenin signaling via decreasing CDX2 level. Cell Commun Signal 2020; 18:169. [PMID: 33109187 PMCID: PMC7590656 DOI: 10.1186/s12964-020-00660-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 09/10/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Jumonji AT-rich interactive domain 1B(JARID1B) has been shown to be upregulated in many human cancers and plays a critical role in the development of cancers cells. Nevertheless, its functional role in colorectal cancer (CRC) progression is not fully understood. METHODS Herein, JARID1B expression levels were detected in clinical CRC samples by western blotting and qRT-PCR. DLD-1 cells with JARID1B knockdown or overexpression by stably transfected plasmids were used in vitro and in vivo study. Colony formation, 5-ethynyl-20-deoxyuridine (EdU) and Real Time Cellular Analysis (RTCA) assays were used to detect cell proliferation and growth. Transcriptome and CHIP assays were used to examine the molecular biology changes and molecular interaction in these cells. Nude mice was utilized to study the correlation of JARID1B and tumor growth in vivo. RESULTS Here, we first observed that JARID1B was significantly upregulated in CRC tissue compared to adjacent normal tissues. In CRC patients, JARID1B high expression was positively relation with poor overall survival. Multivariate analyses revealed that high JARID1B expression was an independent predictive marker for the poor prognosis of CRC. In addition, we found that JARID1B promoted CRC cells proliferation by Wnt/β-catenin signaling pathway. Further studies demonstrated CDX2 as a downstream target of JARID1B, and our data demonstrated that CDX2 is crucial for JARID1B -mediated Wnt/β-catenin signaling pathway. Mechanistically, we demonstrated that JARID1B regulated CDX2 expression through demethylation of H3K4me3. CONCLUSIONS CDX2 inhibited by JARID1B-derived H3K4me3 methylation promoted cells proliferation of CRC via Wnt/β-catenin signaling pathway. Therefore, our studies provided a novel insight into the role of JARID1B in CRC cells proliferation and potential new molecular target for treating CRC. Video abstract.
Collapse
Affiliation(s)
- Da Huang
- Department of Thyroid Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Nanchang, China
| | - Fan Xiao
- Jiangxi Province Key Laboratory of Molecular Medicine, Nanchang, China
- Department of Anesthesiology, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Haibin Hao
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Fuzhou Hua
- Jiangxi Province Key Laboratory of Molecular Medicine, Nanchang, China
- Department of Anesthesiology, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhenzhong Luo
- Jiangxi Province Key Laboratory of Molecular Medicine, Nanchang, China
- Department of Anesthesiology, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhaoxia Huang
- Center for Education Evaluation, Nanchang Normal University, Nanchang, China
| | - Qing Li
- Department of Pathology, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Sha Chen
- Department of Anesthesiology, Second Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Pathology, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiuzhi Cheng
- Jiangxi Province Key Laboratory of Molecular Medicine, Nanchang, China
- Department of Pathology, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xinyue Zhang
- Jiangxi Province Key Laboratory of Molecular Medicine, Nanchang, China
- Department of Pathology, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Weilan Fang
- Jiangxi Province Key Laboratory of Molecular Medicine, Nanchang, China
- Department of Pathology, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaoyun Hu
- Department of General Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Fanrong Liu
- Jiangxi Province Key Laboratory of Molecular Medicine, Nanchang, China
- Department of Pathology, Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
8
|
Saxena K, Srikrishnan S, Celia-Terrassa T, Jolly MK. OVOL1/2: Drivers of Epithelial Differentiation in Development, Disease, and Reprogramming. Cells Tissues Organs 2020; 211:183-192. [PMID: 32932250 DOI: 10.1159/000511383] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 08/26/2020] [Indexed: 11/19/2022] Open
Abstract
OVOL proteins (OVOL1 and OVOL2), vertebrate homologs of Drosophila OVO, are critical regulators of epithelial lineage determination and differentiation during embryonic development in tissues such as kidney, skin, mammary epithelia, and testis. OVOL can inhibit epithelial-mesenchymal transition and/or can promote mesenchymal-epithelial transition. Moreover, they can regulate the stemness of cancer cells, thus playing an important role during cancer cell metastasis. Due to their central role in differentiation and maintenance of epithelial lineage, OVOL overexpression has been shown to be capable of reprogramming fibroblasts to epithelial cells. Here, we review the roles of OVOL-mediated epithelial differentiation across multiple contexts, including embryonic development, cancer progression, and cellular reprogramming.
Collapse
Affiliation(s)
- Kritika Saxena
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | | | - Toni Celia-Terrassa
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Mohit Kumar Jolly
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore, India,
| |
Collapse
|
9
|
Shen YF, Huang JH, Wang KY, Zheng J, Cai L, Gao H, Li XL, Li JF. PTH Derivative promotes wound healing via synergistic multicellular stimulating and exosomal activities. Cell Commun Signal 2020; 18:40. [PMID: 32151266 PMCID: PMC7063786 DOI: 10.1186/s12964-020-00541-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 02/27/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Diabetic wounds are a disturbing and rapidly growing clinical problem. A novel peptide, parathyroid hormone related peptide (PTHrP-2), is assumed as multifunctional factor in angiogenesis, fibrogenesis and re-epithelization. This study aims to test PTHrP-2 efficiency and mechanism in wound healing. METHODS Through repair phenomenon in vivo some problems were detected, and further research on their mechanisms was made. In vivo therapeutic effects of PTHrP-2 were determined by HE, Masson, microfil and immunohistochemical staining. In vitro direct effects of PTHrP-2 were determined by proliferation, migration, Vascular Endothelial Grown Factor and collagen I secretion of cells and Akt/ Erk1/2 pathway change. In vitro indirect effects of PTHrP-2 was study via exosomes. Exosomes from PTHrP-2 untreated and treated HUVECs and HFF-1 cells were insolated and identified. Exosomes were co-cultured with original cells, HUVECs or HFF-1 cells, and epithelial cells. Proliferation and migration and pathway change were observed. PTHrP-2-HUVEC-Exos were added into in vivo wound to testify its hub role in PTHrP-2 indirect effects in wound healing. RESULTS In vivo, PTHrP-2 exerted multifunctional pro-angiogenesis, pro-firbogenesis and re-epithelization effects. In vitro, PTHrP-2 promoted proliferation and migration of endothelial and fibroblast cells, but had no effect on epithelial cells. Therefore, we tested PTHrP-2 indirect effects via exosomes. PTHrP-2 intensified intercellular communication between endothelial cells and fibroblasts and initiated endothelial-epithelial intercellular communication. PTHrP-2-HUVEC-Exos played a hub role in PTHrP-2 indirect effects in wound healing. CONCLUSION These findings of this study indicated that PTHrP-2, a multifunctional factor, could promote wound healing via synergistic multicellular stimulating and exosomal activities.
Collapse
Affiliation(s)
- Yi-Fan Shen
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
| | - Jing-Huan Huang
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
| | - Kai-Yang Wang
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
| | - Jin Zheng
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Lin Cai
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, People’s Republic of China
| | - Hong Gao
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
| | - Xiao-Lin Li
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
| | - Jing-Feng Li
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, People’s Republic of China
| |
Collapse
|
10
|
Sun X, Li Z, Niu Y, Zhao L, Huang Y, Li Q, Zhang S, Chen T, Fu T, Yang T, An X, Jiang Y, Zhang J. Jarid1b promotes epidermal differentiation by mediating the repression of Ship1 and activation of the AKT/Ovol1 pathway. Cell Prolif 2019; 52:e12638. [PMID: 31152465 PMCID: PMC6797505 DOI: 10.1111/cpr.12638] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 04/25/2019] [Accepted: 04/26/2019] [Indexed: 12/13/2022] Open
Abstract
Objectives Terminally differentiated stratified squamous epithelial cells play an important role in barrier protection of the skin. The integrity of epidermal cells is maintained by tight regulation of proliferation and differentiation. The aim of this study was to investigate the role of epigenetic regulator H3K4me3 and its demethylase Jarid1b in the control of epithelial cell differentiation. Materials and methods RT‐qPCR, Western blotting and IHC were used to detect mRNA and protein levels. We analysed cell proliferation by CCK8 assay and cell migration by wound healing assay. ChIP was used to measure H3K4me3 enrichment. A chamber graft model was established for epidermal development. Results Our studies showed that H3K4me3 was decreased during epidermal differentiation. The H3K4me3 demethylase Jarid1b positively controlled epidermal cell differentiation in vitro and in vivo. Mechanistically, we found that Jarid1b substantially increased the expression of mesenchymal‐epithelial transition (MET)‐related genes, among which Ovol1 positively regulated differentiation gene expression. In addition, Ovol1 expression was repressed by PI3K‐AKT pathway inhibitors and overexpression (O/E) of the PI3K‐AKT pathway suppressor Ship1. Knockdown (KD) of Ship1 activated downstream PI3K‐AKT pathway and enhanced Ovol1 expression in HaCaT. Importantly, we found that Jarid1b negatively regulated Ship1 expression, but not that of Pten, by directly binding to its promoter to modulate H3K4me3 enrichment. Conclusion Our results identify an essential role of Jarid1b in the regulation of the Ship1/AKT/Ovol1 pathway to promote epithelial cell differentiation.
Collapse
Affiliation(s)
- Xuewei Sun
- Department of Otolaryngology-Head and Neck Surgery, Key Laboratory, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Zhiyuan Li
- Department of Otolaryngology-Head and Neck Surgery, Key Laboratory, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Yanfang Niu
- Department of Otolaryngology-Head and Neck Surgery, Key Laboratory, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China.,Department of Biochemistry & Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Lijuan Zhao
- Department of Otolaryngology-Head and Neck Surgery, Key Laboratory, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Yichuan Huang
- Department of Otolaryngology-Head and Neck Surgery, Key Laboratory, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Qiang Li
- Department of Andrology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Shengnan Zhang
- Department of Otolaryngology-Head and Neck Surgery, Key Laboratory, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Ting Chen
- Department of Otolaryngology-Head and Neck Surgery, Key Laboratory, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Tao Fu
- Department of Otolaryngology-Head and Neck Surgery, Key Laboratory, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Tao Yang
- Department of Biochemistry & Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Xiaofei An
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yan Jiang
- Department of Otolaryngology-Head and Neck Surgery, Key Laboratory, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Jisheng Zhang
- Department of Otolaryngology-Head and Neck Surgery, Key Laboratory, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China.,Shandong Key Laboratory of Digital Medicine and Computer Assisted Surgery, Qingdao, China.,Shandong College Collaborative Innovation Center of Digital Medicine in Clinical Treatment and Nutrition Health, Qingdao, China
| |
Collapse
|