1
|
Bury LAD, Fu S, Wynshaw-Boris A. Neuronal lineage tracing from progenitors in human cortical organoids reveals mechanisms of neuronal production, diversity, and disease. Cell Rep 2024; 43:114862. [PMID: 39395167 DOI: 10.1016/j.celrep.2024.114862] [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: 08/20/2023] [Revised: 08/14/2024] [Accepted: 09/25/2024] [Indexed: 10/14/2024] Open
Abstract
The contribution of progenitor subtypes to generating the billions of neurons produced during human cortical neurogenesis is not well understood. We developed the cortical organoid lineage-tracing (COR-LT) system for human cortical organoids. Differential fluorescent reporter activation in distinct progenitor cells leads to permanent reporter expression, enabling the progenitor cell lineage of neurons to be determined. Surprisingly, nearly all excitatory neurons produced in cortical organoids were generated indirectly from intermediate progenitor cells. Additionally, neurons of different progenitor lineages were transcriptionally distinct. Isogenic lines made from an autistic individual with and without a likely pathogenic CTNNB1 variant demonstrated that the variant substantially altered the proportion of neurons derived from specific progenitor cell lineages, as well as the lineage-specific transcriptional profiles of these neurons, suggesting a pathogenic mechanism for this mutation. These results suggest individual progenitor subtypes play roles in generating the diverse neurons of the human cerebral cortex.
Collapse
Affiliation(s)
- Luke A D Bury
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| | - Shuai Fu
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
| | - Anthony Wynshaw-Boris
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| |
Collapse
|
2
|
Yang Q, Patrick M, Lu J, Chen J, Zhang Y, Hemani H, Lehrmann E, De S, Weng NP. Homeodomain-only protein suppresses proliferation and contributes to differentiation- and age-related reduced CD8 + T cell expansion. Front Immunol 2024; 15:1360229. [PMID: 38410516 PMCID: PMC10895957 DOI: 10.3389/fimmu.2024.1360229] [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: 12/22/2023] [Accepted: 01/24/2024] [Indexed: 02/28/2024] Open
Abstract
T cell activation is a tightly controlled process involving both positive and negative regulators. The precise mechanisms governing the negative regulators in T cell proliferation remain incompletely understood. Here, we report that homeodomain-only protein (HOPX), a homeodomain-containing protein, and its most abundant isoform HOPXb, negatively regulate activation-induced proliferation of human T cells. We found that HOPX expression progressively increased from naïve (TN) to central memory (TCM) to effector memory (TEM) cells, with a notable upregulation following in vitro stimulation. Overexpression of HOPXb leads to a reduction in TN cell proliferation while HOPX knockdown promotes proliferation of TN and TEM cells. Furthermore, we demonstrated that HOPX binds to promoters and exerts repressive effects on the expression of MYC and NR4A1, two positive regulators known to promote T cell proliferation. Importantly, our findings suggest aging is associated with increased HOPX expression, and that knockdown of HOPX enhances the proliferation of CD8+ T cells in older adults. Our findings provide compelling evidence that HOPX serves as a negative regulator of T cell activation and plays a pivotal role in T cell differentiation and in age-related-reduction in T cell proliferation.
Collapse
Affiliation(s)
- Qian Yang
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Michael Patrick
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Jian Lu
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Joseph Chen
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Yongqing Zhang
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Humza Hemani
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Elin Lehrmann
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Supriyo De
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Nan-ping Weng
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| |
Collapse
|
3
|
Qu M, Jin Z, Xu Y, Sun W, Luo Y, Zhang N, Huang Z, Han L, Gong Y, Xie C. hsa-miR-1301-3p Promotes the Proliferation and Migration of Nonsmall Cell Lung Cancer Cells and Reduces Radiosensitivity via Targeting Homeodomain-Only Protein Homeobox. Genet Test Mol Biomarkers 2023; 27:393-405. [PMID: 38156905 DOI: 10.1089/gtmb.2022.0214] [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] [Indexed: 01/03/2024] Open
Abstract
Background: There is increasing evidence that abnormal expression of microRNAs is involved in the occurrence and progression of tumors. In previous experiments, we found that the content of hsa-miR-1301-3p in tumor tissues of patients with nonsmall cell lung cancer (NSCLC) showed an obvious upward trend compared with that in normal tissues. We performed a detailed study on the impact and underlying mechanism of hsa-miR-1301-3p in NSCLC cells. Methods: The impact of hsa-miR-1301-3p on NSCLC cell proliferation, apoptosis, migration, and invasion was examined using colony formation, flow cytometry, modified Boyden chamber, and wound healing assays. Different doses of radiation were applied to NSCLC cells to investigate their sensitivity to radiotherapy. The potential target gene of hsa-miR-1301-3p was determined by dual-luciferase reporter assay and immunoblotting. Result: hsa-miR-1301-3p was upregulated in NSCLC tissues and cells. hsa-miR-1301-3p effectively promoted the rapid proliferation, migration, and invasion of NSCLC cells, while inhibiting apoptosis. It also induced radioresistance in NSCLC cells. hsa-miR-1301-3p targeted the homeodomain-only protein homeobox (HOPX) mRNA 3' untranslated region and inhibited its transcription in NSCLC cells. Exogenous HOPX overexpression antagonized the mechanism by which hsa-miR-1301-3p regulates NSCLC cell proliferation, metastasis, and apoptosis. Conclusions: hsa-miR-1301-3p plays an oncogenic role in the occurrence and development of NSCLC. By targeting HOPX, hsa-miR-1301-3p can not only promote the proliferation and metastasis of NSCLC cells, but also alleviate apoptosis and reduce radiosensitivity.
Collapse
Affiliation(s)
- Mei Qu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Oncology, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, China
| | - Zhiliang Jin
- Department of Oncology, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, China
| | - Yanhua Xu
- Department of Oncology, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, China
| | - Wenjie Sun
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yuan Luo
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Nannan Zhang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhengrong Huang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Linzhi Han
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yan Gong
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
- Tumor Precision Diagnosis and Treatment Technology and Translational Medicine, Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Conghua Xie
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
4
|
del Valle I, Young MD, Kildisiute G, Ogunbiyi OK, Buonocore F, Simcock IC, Khabirova E, Crespo B, Moreno N, Brooks T, Niola P, Swarbrick K, Suntharalingham JP, McGlacken-Byrne SM, Arthurs OJ, Behjati S, Achermann JC. An integrated single-cell analysis of human adrenal cortex development. JCI Insight 2023; 8:e168177. [PMID: 37440461 PMCID: PMC10443814 DOI: 10.1172/jci.insight.168177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 05/31/2023] [Indexed: 07/15/2023] Open
Abstract
The adrenal glands synthesize and release essential steroid hormones such as cortisol and aldosterone, but many aspects of human adrenal gland development are not well understood. Here, we combined single-cell and bulk RNA sequencing, spatial transcriptomics, IHC, and micro-focus computed tomography to investigate key aspects of adrenal development in the first 20 weeks of gestation. We demonstrate rapid adrenal growth and vascularization, with more cell division in the outer definitive zone (DZ). Steroidogenic pathways favored androgen synthesis in the central fetal zone, but DZ capacity to synthesize cortisol and aldosterone developed with time. Core transcriptional regulators were identified, with localized expression of HOPX (also known as Hop homeobox/homeobox-only protein) in the DZ. Potential ligand-receptor interactions between mesenchyme and adrenal cortex were seen (e.g., RSPO3/LGR4). Growth-promoting imprinted genes were enriched in the developing cortex (e.g., IGF2, PEG3). These findings reveal aspects of human adrenal development and have clinical implications for understanding primary adrenal insufficiency and related postnatal adrenal disorders, such as adrenal tumor development, steroid disorders, and neonatal stress.
Collapse
Affiliation(s)
- Ignacio del Valle
- Genetics and Genomic Medicine Research and Teaching Department, University College London (UCL) Great Ormond Street Institute of Child Health, UCL, London, United Kingdom
| | - Matthew D. Young
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Gerda Kildisiute
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Olumide K. Ogunbiyi
- Department of Histopathology, Great Ormond Street Hospital for Children National Health Service (NHS) Foundation Trust, London, United Kingdom
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, UCL, London, United Kingdom
| | - Federica Buonocore
- Genetics and Genomic Medicine Research and Teaching Department, University College London (UCL) Great Ormond Street Institute of Child Health, UCL, London, United Kingdom
| | - Ian C. Simcock
- Department of Clinical Radiology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
- National Institute of Health Research (NIHR) Great Ormond Street Biomedical Research Centre, London, United Kingdom
- Population, Policy and Practice Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, UCL, London, United Kingdom
| | - Eleonora Khabirova
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Berta Crespo
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, UCL, London, United Kingdom
| | - Nadjeda Moreno
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, UCL, London, United Kingdom
| | - Tony Brooks
- UCL Genomics, Zayed Centre for Research, UCL Great Ormond Street Institute of Child Health, UCL, London, United Kingdom
| | - Paola Niola
- UCL Genomics, Zayed Centre for Research, UCL Great Ormond Street Institute of Child Health, UCL, London, United Kingdom
| | - Katherine Swarbrick
- Department of Histopathology, Great Ormond Street Hospital for Children National Health Service (NHS) Foundation Trust, London, United Kingdom
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, UCL, London, United Kingdom
| | - Jenifer P. Suntharalingham
- Genetics and Genomic Medicine Research and Teaching Department, University College London (UCL) Great Ormond Street Institute of Child Health, UCL, London, United Kingdom
| | - Sinead M. McGlacken-Byrne
- Genetics and Genomic Medicine Research and Teaching Department, University College London (UCL) Great Ormond Street Institute of Child Health, UCL, London, United Kingdom
| | - Owen J. Arthurs
- Department of Clinical Radiology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
- National Institute of Health Research (NIHR) Great Ormond Street Biomedical Research Centre, London, United Kingdom
- Population, Policy and Practice Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, UCL, London, United Kingdom
| | - Sam Behjati
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
- Department of Paediatrics, University of Cambridge, Cambridge, United Kingdom
| | - John C. Achermann
- Genetics and Genomic Medicine Research and Teaching Department, University College London (UCL) Great Ormond Street Institute of Child Health, UCL, London, United Kingdom
| |
Collapse
|
5
|
He S, Ding Y, Ji Z, Yuan B, Chen J, Ren W. HOPX is a tumor-suppressive biomarker that corresponds to T cell infiltration in skin cutaneous melanoma. Cancer Cell Int 2023; 23:122. [PMID: 37344870 DOI: 10.1186/s12935-023-02962-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 06/02/2023] [Indexed: 06/23/2023] Open
Abstract
BACKGROUND Skin cutaneous melanoma (SKCM) is the most threatening type of skin cancer. Approximately 55,000 people lose their lives every year due to SKCM, illustrating that it seriously threatens human life and health. Homeodomain-only protein homeobox (HOPX) is the smallest member of the homeodomain family and is widely expressed in a variety of tissues. HOPX is involved in regulating the homeostasis of hematopoietic stem cells and is closely related to the development of tumors such as breast cancer, nasopharyngeal carcinoma, and head and neck squamous cell carcinoma. However, its function in SKCM is unclear, and further studies are needed. METHODS We used the R language to construct ROC (Receiver-Operating Characteristic) curves, KM (Kaplan‒Meier) curves and nomograms based on databases such as the TCGA and GEO to analyze the diagnostic and prognostic value of HOPX in SKCM patients. Enrichment analysis, immune scoring, GSVA (Gene Set Variation Analysis), and single-cell sequencing were used to verify the association between HOPX expression and immune infiltration. In vitro experiments were performed using A375 cells for phenotypic validation. Transcriptome sequencing was performed to further analyze HOPX gene-related genes and their signaling pathways. RESULTS Compared to normal cells, SKCM cells had low HOPX expression (p < 0.001). Patients with high HOPX expression had a better prognosis (p < 0.01), and the marker had good diagnostic efficacy (AUC = 0.744). GO/KEGG (Gene Ontology/ Kyoto Encyclopedia of Genes and Genomes) analysis, GSVA and single-cell sequencing analysis showed that HOPX expression is associated with immune processes and high enrichment of T cells and could serve as an immune checkpoint in SKCM. Furthermore, cellular assays verified that HOPX inhibits the proliferation, migration and invasion of A375 cells and promotes apoptosis and S-phase arrest. Interestingly, tumor drug sensitivity analysis revealed that HOPX also plays an important role in reducing clinical drug resistance. CONCLUSION These findings suggest that HOPX is a blocker of SKCM progression that inhibits the proliferation of SKCM cells and promotes apoptosis. Furthermore, it may be a new diagnostic and prognostic indicator and a novel target for immunotherapy in SKCM patients.
Collapse
Affiliation(s)
- Song He
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, P.R. China
| | - Yu Ding
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, P.R. China
| | - Zhonghao Ji
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, P.R. China
- Department of Basic Medicine, Changzhi Medical College, Changzhi, 046000, Shanxi, P.R. China
| | - Bao Yuan
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, P.R. China
| | - Jian Chen
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, P.R. China.
| | - Wenzhi Ren
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, 130062, Jilin, P.R. China.
| |
Collapse
|
6
|
Jin Y, Arimura H, Cui Y, Kodama T, Mizuno S, Ansai S. CT Image-Based Biopsy to Aid Prediction of HOPX Expression Status and Prognosis for Non-Small Cell Lung Cancer Patients. Cancers (Basel) 2023; 15:2220. [PMID: 37190150 PMCID: PMC10136849 DOI: 10.3390/cancers15082220] [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: 02/06/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 05/17/2023] Open
Abstract
This study aimed to elucidate a computed tomography (CT) image-based biopsy with a radiogenomic signature to predict homeodomain-only protein homeobox (HOPX) gene expression status and prognosis in patients with non-small cell lung cancer (NSCLC). Patients were labeled as HOPX-negative or positive based on HOPX expression and were separated into training (n = 92) and testing (n = 24) datasets. In correlation analysis between genes and image features extracted by Pyradiomics for 116 patients, eight significant features associated with HOPX expression were selected as radiogenomic signature candidates from the 1218 image features. The final signature was constructed from eight candidates using the least absolute shrinkage and selection operator. An imaging biopsy model with radiogenomic signature was built by a stacking ensemble learning model to predict HOPX expression status and prognosis. The model exhibited predictive power for HOPX expression with an area under the receiver operating characteristic curve of 0.873 and prognostic power in Kaplan-Meier curves (p = 0.0066) in the test dataset. This study's findings implied that the CT image-based biopsy with a radiogenomic signature could aid physicians in predicting HOPX expression status and prognosis in NSCLC.
Collapse
Affiliation(s)
- Yu Jin
- Division of Medical Quantum Science, Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Hidetaka Arimura
- Division of Medical Quantum Science, Department of Health Sciences, Faculty of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - YunHao Cui
- Division of Medical Quantum Science, Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Takumi Kodama
- Division of Medical Quantum Science, Department of Health Sciences, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Shinichi Mizuno
- Division of Medical Technology, Department of Health Sciences, Faculty of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Satoshi Ansai
- Laboratory of Genome Editing Breeding, Graduate School of Agriculture, Kyoto University, Kyoto 606-8520, Japan
| |
Collapse
|
7
|
Bourque J, Kousnetsov R, Hawiger D. Roles of Hopx in the differentiation and functions of immune cells. Eur J Cell Biol 2022; 101:151242. [DOI: 10.1016/j.ejcb.2022.151242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/20/2022] [Accepted: 05/20/2022] [Indexed: 11/03/2022] Open
|
8
|
HOPX: A Unique Homeodomain Protein in Development and Tumor Suppression. Cancers (Basel) 2022; 14:cancers14112764. [PMID: 35681746 PMCID: PMC9179269 DOI: 10.3390/cancers14112764] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/25/2022] [Accepted: 05/30/2022] [Indexed: 12/05/2022] Open
Abstract
Simple Summary Homeobox (HOX) genes encode homeodomain proteins that regulate a wide range of molecular pathways. The homeodomain is highly conserved and binds to DNA. One exception is homeodomain-only protein (HOPX) that lacks DNA-binding capacity. HOPX plays a crucial role in development and its functional impairment is associated with a variety of diseases, including cancer. Loss of HOPX function occurs in a wide range of cancer types, where it functions as a tumor suppressor gene. Understanding the molecular mechanisms by which HOPX regulates carcinogenesis will likely lead to the development of new therapeutic approaches. Abstract Homeobox genes are master regulators of morphogenesis and differentiation by acting at the top of genetic hierarchies and their deregulation is associated with a variety of human diseases. They usually contain a highly conserved sequence that codes for the homeodomain of the protein, a specialized motif with three α helices and an N-terminal arm that aids in DNA binding. However, one homeodomain protein, HOPX, is unique among its family members in that it lacks the capacity to bind DNA and instead functions by interacting with transcriptional regulators. HOPX plays crucial roles in organogenesis and is expressed in both embryonic and adult stem cells. Loss of HOPX expression is common in cancer, where it functions primarily as a tumor suppressor gene. In this review, we describe the function of HOPX in development and discuss its role in carcinogenesis.
Collapse
|
9
|
Zheng Y, Zhou Y, Huang Y, Wang H, Guo H, Yuan B, Zhang J. Transcriptome sequencing of black and white hair follicles in the giant panda. Integr Zool 2022; 18:552-568. [PMID: 35500067 DOI: 10.1111/1749-4877.12652] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
With the completion of the draft assembly of the giant panda genome sequence, RNA sequencing technology has been widely used in genetic research on giant pandas. We used RNA-seq to examine black and white hair follicle samples from adult pandas. By comparison with the giant panda genome, 75 963 SNP loci were labeled, 2 426 differentially expressed genes were identified, and 2 029 new genes were discovered, among which 631 were functionally annotated. A cluster analysis of the differentially expressed genes showed that they were mainly related to the Wnt signaling pathway, ECM-receptor interaction, the p53 signaling pathway and ribosome processing. The enrichment results showed that there were significant differences in the regulatory networks of hair follicles with different colors during the transitional stage of hair follicle resting growth, which may play a regulatory role in melanin synthesis during growth. In conclusion, our results provide new insights and more data support for research on the color formation in giant pandas. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Yi Zheng
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, China
| | - Yingmin Zhou
- Key Laboratory of SFGA on Conservation Biology of Rare Animals in The Giant Panda National Park, China
| | - Yijie Huang
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, China
| | - Haoqi Wang
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, China
| | - Haixiang Guo
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, China
| | - Bao Yuan
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, China
| | - Jiabao Zhang
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, China
| |
Collapse
|
10
|
Osakunor DNM, Ishida K, Lamanna OK, Rossi M, Dwomoh L, Hsieh MH. Host tissue proteomics reveal insights into the molecular basis of Schistosoma haematobium-induced bladder pathology. PLoS Negl Trop Dis 2022; 16:e0010176. [PMID: 35167594 PMCID: PMC8846513 DOI: 10.1371/journal.pntd.0010176] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/17/2022] [Indexed: 12/25/2022] Open
Abstract
Urogenital schistosomiasis remains a major public health concern worldwide. In response to egg deposition, the host bladder undergoes gross and molecular morphological changes relevant for disease manifestation. However, limited mechanistic studies to date imply that the molecular mechanisms underlying pathology are not well-defined. We leveraged a mouse model of urogenital schistosomiasis to perform for the first time, proteome profiling of the early molecular events that occur in the bladder after exposure to S. haematobium eggs, and to elucidate the protein pathways involved in urogenital schistosomiasis-induced pathology. Purified S. haematobium eggs or control vehicle were microinjected into the bladder walls of mice. Mice were sacrificed seven days post-injection and bladder proteins isolated and processed for proteome profiling using mass spectrometry. We demonstrate that biological processes including carcinogenesis, immune and inflammatory responses, increased protein translation or turnover, oxidative stress responses, reduced cell adhesion and epithelial barrier integrity, and increased glucose metabolism were significantly enriched in S. haematobium infection. S. haematobium egg deposition in the bladder results in significant changes in proteins and pathways that play a role in pathology. Our findings highlight the potential bladder protein indicators for host-parasite interplay and provide new insights into the complex dynamics of pathology and characteristic bladder tissue changes in urogenital schistosomiasis. The findings will be relevant for development of improved interventions for disease control.
Collapse
Affiliation(s)
- Derick N. M. Osakunor
- Division of Urology, Department of Surgery, Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Hospital, Washington, District of Columbia, United States of America
| | - Kenji Ishida
- Division of Urology, Department of Surgery, Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Hospital, Washington, District of Columbia, United States of America
| | - Olivia K. Lamanna
- Division of Urology, Department of Surgery, Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Hospital, Washington, District of Columbia, United States of America
| | - Mario Rossi
- Institute of Molecular, Cell and Systems Biology, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Louis Dwomoh
- Institute of Molecular, Cell and Systems Biology, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Michael H. Hsieh
- Division of Urology, Department of Surgery, Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Hospital, Washington, District of Columbia, United States of America
- Departments of Urology, Department of Pediatrics, and Department of Microbiology, Immunology, and Tropical Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, District of Columbia, United States of America
| |
Collapse
|
11
|
Lachner J, Ehrlich F, Wielscher M, Farlik M, Hermann M, Tschachler E, Eckhart L. Single-cell transcriptomics defines keratinocyte differentiation in avian scutate scales. Sci Rep 2022; 12:126. [PMID: 34997067 PMCID: PMC8742010 DOI: 10.1038/s41598-021-04082-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 12/09/2021] [Indexed: 11/24/2022] Open
Abstract
The growth of skin appendages, such as hair, feathers and scales, depends on terminal differentiation of epidermal keratinocytes. Here, we investigated keratinocyte differentiation in avian scutate scales. Cells were isolated from the skin on the legs of 1-day old chicks and subjected to single-cell transcriptomics. We identified two distinct populations of differentiated keratinocytes. The first population was characterized by mRNAs encoding cysteine-rich keratins and corneous beta-proteins (CBPs), also known as beta-keratins, of the scale type, indicating that these cells form hard scales. The second population of differentiated keratinocytes contained mRNAs encoding cysteine-poor keratins and keratinocyte-type CBPs, suggesting that these cells form the soft interscale epidermis. We raised an antibody against keratin 9-like cysteine-rich 2 (KRT9LC2), which is encoded by an mRNA enriched in the first keratinocyte population. Immunostaining confirmed expression of KRT9LC2 in the suprabasal epidermal layers of scutate scales but not in interscale epidermis. Keratinocyte differentiation in chicken leg skin resembled that in human skin with regard to the transcriptional upregulation of epidermal differentiation complex genes and genes involved in lipid metabolism and transport. In conclusion, this study defines gene expression programs that build scutate scales and interscale epidermis of birds and reveals evolutionarily conserved keratinocyte differentiation genes.
Collapse
Affiliation(s)
- Julia Lachner
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Florian Ehrlich
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Matthias Wielscher
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Matthias Farlik
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Marcela Hermann
- Department of Medical Biochemistry, Medical University of Vienna, Vienna, Austria
| | - Erwin Tschachler
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Leopold Eckhart
- Department of Dermatology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
12
|
Muhammad K, Xavier D, Klein-Hessling S, Azeem M, Rauschenberger T, Murti K, Avots A, Goebeler M, Klein M, Bopp T, Sielaff M, Tenzer S, Möckel S, Aramburu J, López-Rodríguez C, Kerstan A, Serfling E. NFAT5 Controls the Integrity of Epidermis. Front Immunol 2021; 12:780727. [PMID: 34956208 PMCID: PMC8696207 DOI: 10.3389/fimmu.2021.780727] [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: 09/21/2021] [Accepted: 11/15/2021] [Indexed: 11/13/2022] Open
Abstract
The skin protects the human body against dehydration and harmful challenges. Keratinocytes (KCs) are the most abundant epidermal cells, and it is anticipated that KC-mediated transport of Na+ ions creates a physiological barrier of high osmolality against the external environment. Here, we studied the role of NFAT5, a transcription factor whose activity is controlled by osmotic stress in KCs. Cultured KCs from adult mice were found to secrete more than 300 proteins, and upon NFAT5 ablation, the secretion of several matrix proteinases, including metalloproteinase-3 (Mmp3) and kallikrein-related peptidase 7 (Klk7), was markedly enhanced. An increase in Mmp3 and Klk7 RNA levels was also detected in transcriptomes of Nfat5-/- KCs, along with increases of numerous members of the 'Epidermal Differentiation Complex' (EDC), such as small proline-rich (Sprr) and S100 proteins. NFAT5 and Mmp3 as well as NFAT5 and Klk7 are co-expressed in the basal KCs of fetal and adult epidermis but not in basal KCs of newborn (NB) mice. The poor NFAT5 expression in NB KCs is correlated with a strong increase in Mmp3 and Klk7 expression in KCs of NB mice. These data suggests that, along with the fragile epidermis of adult Nfat5-/- mice, NFAT5 keeps in check the expression of matrix proteases in epidermis. The NFAT5-mediated control of matrix proteases in epidermis contributes to the manifold changes in skin development in embryos before and during birth, and to the integrity of epidermis in adults.
Collapse
Affiliation(s)
- Khalid Muhammad
- Department of Molecular Pathology, Institute of Pathology, University of Wuerzburg, Wuerzburg, Germany
| | - Delicia Xavier
- Department of Molecular Pathology, Institute of Pathology, University of Wuerzburg, Wuerzburg, Germany
| | - Stefan Klein-Hessling
- Department of Molecular Pathology, Institute of Pathology, University of Wuerzburg, Wuerzburg, Germany.,Comprehensive Cancer Centre Mainfranken, Wuerzburg, Germany
| | - Muhammad Azeem
- Department of Molecular Pathology, Institute of Pathology, University of Wuerzburg, Wuerzburg, Germany.,Department of Dermatology, Venereology and Allergology, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Tabea Rauschenberger
- Department of Molecular Pathology, Institute of Pathology, University of Wuerzburg, Wuerzburg, Germany
| | - Krisna Murti
- Department of Molecular Pathology, Institute of Pathology, University of Wuerzburg, Wuerzburg, Germany
| | - Andris Avots
- Department of Molecular Pathology, Institute of Pathology, University of Wuerzburg, Wuerzburg, Germany.,Comprehensive Cancer Centre Mainfranken, Wuerzburg, Germany
| | - Matthias Goebeler
- Department of Dermatology, Venereology and Allergology, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Matthias Klein
- Institute for Immunology, University Medical Center, University of Mainz, Mainz, Germany
| | - Tobias Bopp
- Institute for Immunology, University Medical Center, University of Mainz, Mainz, Germany.,Research Center for Immunotherapy, University Medical Center, University of Mainz, Mainz, Germany.,University Cancer Center Mainz, University Medical Center, University of Mainz, Mainz, Germany
| | - Malte Sielaff
- Institute for Immunology, University Medical Center, University of Mainz, Mainz, Germany
| | - Stefan Tenzer
- Institute for Immunology, University Medical Center, University of Mainz, Mainz, Germany
| | - Sigrid Möckel
- Institute of Pathology, University of Wuerzburg, Wuerzburg, Germany
| | - José Aramburu
- Immunology Unit, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Cristina López-Rodríguez
- Immunology Unit, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Andreas Kerstan
- Department of Dermatology, Venereology and Allergology, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Edgar Serfling
- Department of Molecular Pathology, Institute of Pathology, University of Wuerzburg, Wuerzburg, Germany.,Comprehensive Cancer Centre Mainfranken, Wuerzburg, Germany
| |
Collapse
|
13
|
Bourque J, Opejin A, Surnov A, Iberg CA, Gross C, Jain R, Epstein JA, Hawiger D. Landscape of Hopx expression in cells of the immune system. Heliyon 2021; 7:e08311. [PMID: 34805566 PMCID: PMC8590040 DOI: 10.1016/j.heliyon.2021.e08311] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/30/2021] [Accepted: 10/29/2021] [Indexed: 11/29/2022] Open
Abstract
Homeodomain only protein (Hopx) is a regulator of cell differentiation and function, and it has also emerged as a crucial marker of specific developmental and differentiation potentials. Hopx expression and functions have been identified in some stem cells, tumors, and in certain immune cells. However, expression of Hopx in immune cells remains insufficiently characterized. Here we report a comprehensive pattern of Hopx expression in multiple types of immune cells under steady state conditions. By utilizing single-cell RNA sequencing (scRNA-seq) and flow cytometric analysis, we characterize a constitutive expression of Hopx in specific subsets of CD4+ and CD8+ T cells and B cells, as well as natural killer (NK), NKT, and myeloid cells. In contrast, Hopx expression is not present in conventional dendritic cells and eosinophils. The utility of identifying expression of Hopx in immune cells may prove vital in delineating specific roles of Hopx under multiple immune conditions.
Collapse
Affiliation(s)
- Jessica Bourque
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63118, USA
| | - Adeleye Opejin
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63118, USA
| | - Alexey Surnov
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63118, USA
| | - Courtney A Iberg
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63118, USA
| | - Cindy Gross
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63118, USA
| | - Rajan Jain
- Department of Medicine, Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jonathan A Epstein
- Department of Medicine, Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Daniel Hawiger
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63118, USA
| |
Collapse
|
14
|
Tsan YC, DePalma SJ, Zhao YT, Capilnasiu A, Wu YW, Elder B, Panse I, Ufford K, Matera DL, Friedline S, O'Leary TS, Wubshet N, Ho KKY, Previs MJ, Nordsletten D, Isom LL, Baker BM, Liu AP, Helms AS. Physiologic biomechanics enhance reproducible contractile development in a stem cell derived cardiac muscle platform. Nat Commun 2021; 12:6167. [PMID: 34697315 PMCID: PMC8546060 DOI: 10.1038/s41467-021-26496-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 10/05/2021] [Indexed: 12/29/2022] Open
Abstract
Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) allow investigations in a human cardiac model system, but disorganized mechanics and immaturity of hPSC-CMs on standard two-dimensional surfaces have been hurdles. Here, we developed a platform of micron-scale cardiac muscle bundles to control biomechanics in arrays of thousands of purified, independently contracting cardiac muscle strips on two-dimensional elastomer substrates with far greater throughput than single cell methods. By defining geometry and workload in this reductionist platform, we show that myofibrillar alignment and auxotonic contractions at physiologic workload drive maturation of contractile function, calcium handling, and electrophysiology. Using transcriptomics, reporter hPSC-CMs, and quantitative immunofluorescence, these cardiac muscle bundles can be used to parse orthogonal cues in early development, including contractile force, calcium load, and metabolic signals. Additionally, the resultant organized biomechanics facilitates automated extraction of contractile kinetics from brightfield microscopy imaging, increasing the accessibility, reproducibility, and throughput of pharmacologic testing and cardiomyopathy disease modeling. Investigations of human cardiac disease involving human pluripotent stem cell-derived cardiomyocytes are limited by the disorganized presentation of biomechanical cues resulting in cell immaturity. Here the authors develop a platform of micron-scale 2D cardiac muscle bundles to precisely deliver physiologic cues, improving reproducibility and throughput.
Collapse
Affiliation(s)
- Yao-Chang Tsan
- Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, USA.,Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Samuel J DePalma
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Yan-Ting Zhao
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - Adela Capilnasiu
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Yu-Wei Wu
- Institute of Molecular Biology, Academia Sinica, NanKang, Taipei, Taiwan
| | - Brynn Elder
- Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Isabella Panse
- Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Kathryn Ufford
- Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Daniel L Matera
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Sabrina Friedline
- Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Thomas S O'Leary
- Molecular Physiology and Biophysics, University of Vermont, Burlington, VT, USA
| | - Nadab Wubshet
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Kenneth K Y Ho
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Michael J Previs
- Molecular Physiology and Biophysics, University of Vermont, Burlington, VT, USA
| | - David Nordsletten
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.,Department of Cardiovascular Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Lori L Isom
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA.,Department of Neurology, University of Michigan, Ann Arbor, MI, USA.,Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Brendon M Baker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Allen P Liu
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.,Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA.,Department of Biophysics, University of Michigan, Ann Arbor, MI, USA
| | - Adam S Helms
- Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
15
|
In Silico Analysis to Explore Lineage-Independent and -Dependent Transcriptional Programs Associated with the Process of Endothelial and Neural Differentiation of Human Induced Pluripotent Stem Cells. J Clin Med 2021; 10:jcm10184161. [PMID: 34575270 PMCID: PMC8471316 DOI: 10.3390/jcm10184161] [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: 08/20/2021] [Revised: 09/11/2021] [Accepted: 09/13/2021] [Indexed: 11/17/2022] Open
Abstract
Despite a major interest in understanding how the endothelial cell phenotype is established, the underlying molecular basis of this process is not yet fully understood. We have previously reported the generation of induced pluripotent stem cells (iPS) from human umbilical vein endothelial cells and differentiation of the resulting HiPS back to endothelial cells (Ec-Diff), as well as neural (Nn-Diff) cell lineage that contained both neurons and astrocytes. Furthermore, the identities of these cell lineages were established by gene array analysis. Here, we explored the same arrays to gain insight into the gene alteration processes that accompany the establishment of endothelial vs. non-endothelial neural cell phenotypes. We compared the expression of genes that code for transcription factors and epigenetic regulators when HiPS is differentiated into these endothelial and non-endothelial lineages. Our in silico analyses have identified cohorts of genes that are similarly up- or downregulated in both lineages, as well as those that exhibit lineage-specific alterations. Based on these results, we propose that genes that are similarly altered in both lineages participate in priming the stem cell for differentiation in a lineage-independent manner, whereas those that are differentially altered in endothelial compared to neural cells participate in a lineage-specific differentiation process. Specific GATA family members and their cofactors and epigenetic regulators (DNMT3B, PRDM14, HELLS) with a major role in regulating DNA methylation were among participants in priming HiPS for lineage-independent differentiation. In addition, we identified distinct cohorts of transcription factors and epigenetic regulators whose alterations correlated specifically with the establishment of endothelial vs. non-endothelial neural lineages.
Collapse
|
16
|
Tatomir A, Beltrand A, Nguyen V, Courneya JP, Boodhoo D, Cudrici C, Muresanu DF, Rus V, Badea TC, Rus H. RGC-32 Acts as a Hub to Regulate the Transcriptomic Changes Associated With Astrocyte Development and Reactive Astrocytosis. Front Immunol 2021; 12:705308. [PMID: 34394104 PMCID: PMC8358671 DOI: 10.3389/fimmu.2021.705308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 07/16/2021] [Indexed: 01/14/2023] Open
Abstract
Response Gene to Complement 32 (RGC-32) is an important mediator of the TGF-β signaling pathway, and an increasing amount of evidence implicates this protein in regulating astrocyte biology. We showed recently that spinal cord astrocytes in mice lacking RGC-32 display an immature phenotype reminiscent of progenitors and radial glia, with an overall elongated morphology, increased proliferative capacity, and increased expression of progenitor markers when compared to their wild-type (WT) counterparts that make them incapable of undergoing reactive changes during the acute phase of experimental autoimmune encephalomyelitis (EAE). Here, in order to decipher the molecular networks underlying RGC-32's ability to regulate astrocytic maturation and reactivity, we performed next-generation sequencing of RNA from WT and RGC-32 knockout (KO) neonatal mouse brain astrocytes, either unstimulated or stimulated with the pleiotropic cytokine TGF-β. Pathway enrichment analysis showed that RGC-32 is critical for the TGF-β-induced up-regulation of transcripts encoding proteins involved in brain development and tissue remodeling, such as axonal guidance molecules, transcription factors, extracellular matrix (ECM)-related proteins, and proteoglycans. Our next-generation sequencing of RNA analysis also demonstrated that a lack of RGC-32 results in a significant induction of WD repeat and FYVE domain-containing protein 1 (Wdfy1) and stanniocalcin-1 (Stc1). Immunohistochemical analysis of spinal cords isolated from normal adult mice and mice with EAE at the peak of disease showed that RGC-32 is necessary for the in vivo expression of ephrin receptor type A7 in reactive astrocytes, and that the lack of RGC-32 results in a higher number of homeodomain-only protein homeobox (HOPX)+ and CD133+ radial glia cells. Collectively, these findings suggest that RGC-32 plays a major role in modulating the transcriptomic changes in astrocytes that ultimately lead to molecular programs involved in astrocytic differentiation and reactive changes during neuroinflammation.
Collapse
Affiliation(s)
- Alexandru Tatomir
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
- Department of Neurosciences, “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Austin Beltrand
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Vinh Nguyen
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Jean-Paul Courneya
- Health Sciences and Human Services Library, University of Maryland, Baltimore, MD, United States
| | - Dallas Boodhoo
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Cornelia Cudrici
- Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Dafin F. Muresanu
- Department of Neurosciences, “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Violeta Rus
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Tudor C. Badea
- Retinal Circuit Development and Genetics Unit, N-NRL, National Eye Institute, Bethesda, MD, United States
- Research and Development Institute, Faculty of Medicine, Transylvania University of Brasov, Brasov, Romania
| | - Horea Rus
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
- Research Service, Veterans Administration Maryland Health Care System, Baltimore, MD, United States
| |
Collapse
|
17
|
HOPX Exhibits Oncogenic Activity during Squamous Skin Carcinogenesis. J Invest Dermatol 2021; 141:2354-2368. [PMID: 33845078 DOI: 10.1016/j.jid.2020.04.034] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 04/27/2020] [Accepted: 04/27/2020] [Indexed: 11/23/2022]
Abstract
Cutaneous squamous cell carcinomas (SCCs) are frequent heterogeneous tumors arising from sun-exposed regions of the skin and characterized by complex pathogenesis. HOPX is a member of the homeodomain-containing superfamily of proteins holding an atypical homeodomain unable to bind to DNA. First discovered in the heart as a regulator of cardiac development, in the skin, HOPX modulates the terminal differentiation of keratinocytes. There is a particular interest in studying HOPX in squamous skin carcinogenesis because it has the atypical structure and the functional duality as an oncogene and a tumor suppressor gene, reported in different malignancies. In this study, we analyzed the effects of HOPX knockdown and overexpression on SCC tumorigenicity in vitro and in vivo. Our data show that HOPX knockdown in SCC cells inhibits their proliferative and invasive activity through the acceleration of apoptosis. We established that methylation of two alternative HOPX promoters leads to differential expression of HOPX transcripts in normal keratinocytes and SCC cells. Importantly, we report that HOPX acts as an oncogene in the pathogenesis of SCC probably through the activation of the second alternative promoter and the modulation of apoptosis.
Collapse
|
18
|
The formation of pre-effectors in the steady state opens a new perspective for cancer immunosurveillance. Oncotarget 2021; 12:1318-1320. [PMID: 34194629 PMCID: PMC8238249 DOI: 10.18632/oncotarget.27967] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Indexed: 11/25/2022] Open
|
19
|
Opejin A, Surnov A, Misulovin Z, Pherson M, Gross C, Iberg CA, Fallahee I, Bourque J, Dorsett D, Hawiger D. A Two-Step Process of Effector Programming Governs CD4 + T Cell Fate Determination Induced by Antigenic Activation in the Steady State. Cell Rep 2020; 33:108424. [PMID: 33238127 PMCID: PMC7714042 DOI: 10.1016/j.celrep.2020.108424] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 10/01/2020] [Accepted: 10/30/2020] [Indexed: 02/06/2023] Open
Abstract
Various processes induce and maintain immune tolerance, but effector T cells still arise under minimal perturbations of homeostasis through unclear mechanisms. We report that, contrary to the model postulating primarily tolerogenic mechanisms initiated under homeostatic conditions, effector programming is an integral part of T cell fate determination induced by antigenic activation in the steady state. This effector programming depends on a two-step process starting with induction of effector precursors that express Hopx and are imprinted with multiple instructions for their subsequent terminal effector differentiation. Such molecular circuits advancing specific terminal effector differentiation upon re-stimulation include programmed expression of interferon-γ, whose production then promotes expression of T-bet in the precursors. We further show that effector programming coincides with regulatory conversion among T cells sharing the same antigen specificity. However, conventional type 2 dendritic cells (cDC2) and T cell functions of mammalian target of rapamycin complex 1 (mTORC1) increase effector precursor induction while decreasing the proportion of T cells that can become peripheral Foxp3+ regulatory T (pTreg) cells. The mechanisms in the steady state that govern the formation of effector T cells with potentially autoimmune functions remain unclear. Opejin et al. reveal a two-step process starting with induction of effector precursors that express Hopx and are imprinted with multiple instructions for their subsequent terminal effector differentiation.
Collapse
Affiliation(s)
- Adeleye Opejin
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Alexey Surnov
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Ziva Misulovin
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Michelle Pherson
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Cindy Gross
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Courtney A Iberg
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Ian Fallahee
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Jessica Bourque
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Dale Dorsett
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Daniel Hawiger
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
20
|
Jones J, Chen Y, Tiwari M, Li J, Ling J, Sen GL. KLF3 Mediates Epidermal Differentiation through the Epigenomic Writer CBP. iScience 2020; 23:101320. [PMID: 32659720 PMCID: PMC7358749 DOI: 10.1016/j.isci.2020.101320] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 06/02/2020] [Accepted: 06/24/2020] [Indexed: 12/24/2022] Open
Abstract
Impairments in the differentiation process can lead to skin diseases that can afflict ∼20% of the population. Thus, it is of utmost importance to understand the factors that promote the differentiation process. Here we identify the transcription factor KLF3 as a regulator of epidermal differentiation. Knockdown of KLF3 results in reduced differentiation gene expression and increased cell cycle gene expression. Over half of KLF3's genomic binding sites occur at active enhancers. KLF3 binds to active enhancers proximal to differentiation genes that are dependent upon KLF3 for expression. KLF3's genomic binding sites also highly overlaps with CBP, a histone acetyltransferase necessary for activating enhancers. Depletion of KLF3 causes reduced CBP localization at enhancers proximal to differentiation gene clusters, which leads to loss of enhancer activation but not priming. Our results suggest that KLF3 is necessary to recruit CBP to activate enhancers and drive epidermal differentiation gene expression.
Collapse
Affiliation(s)
- Jackson Jones
- Department of Dermatology, Department of Cellular and Molecular Medicine, UCSD Stem Cell Program, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0869, USA
| | - Yifang Chen
- Department of Dermatology, Department of Cellular and Molecular Medicine, UCSD Stem Cell Program, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0869, USA
| | - Manisha Tiwari
- Department of Dermatology, Department of Cellular and Molecular Medicine, UCSD Stem Cell Program, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0869, USA
| | - Jingting Li
- Department of Dermatology, Department of Cellular and Molecular Medicine, UCSD Stem Cell Program, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0869, USA
| | - Ji Ling
- Department of Dermatology, Department of Cellular and Molecular Medicine, UCSD Stem Cell Program, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0869, USA
| | - George L Sen
- Department of Dermatology, Department of Cellular and Molecular Medicine, UCSD Stem Cell Program, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0869, USA.
| |
Collapse
|
21
|
Vaid S, Huttner WB. Transcriptional Regulators and Human-Specific/Primate-Specific Genes in Neocortical Neurogenesis. Int J Mol Sci 2020; 21:ijms21134614. [PMID: 32610533 PMCID: PMC7369782 DOI: 10.3390/ijms21134614] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/09/2020] [Accepted: 06/26/2020] [Indexed: 12/15/2022] Open
Abstract
During development, starting from a pool of pluripotent stem cells, tissue-specific genetic programs help to shape and develop functional organs. To understand the development of an organ and its disorders, it is important to understand the spatio-temporal dynamics of the gene expression profiles that occur during its development. Modifications in existing genes, the de-novo appearance of new genes, or, occasionally, even the loss of genes, can greatly affect the gene expression profile of any given tissue and contribute to the evolution of organs or of parts of organs. The neocortex is evolutionarily the most recent part of the brain, it is unique to mammals, and is the seat of our higher cognitive abilities. Progenitors that give rise to this tissue undergo sequential waves of differentiation to produce the complete sets of neurons and glial cells that make up a functional neocortex. We will review herein our understanding of the transcriptional regulators that control the neural precursor cells (NPCs) during the generation of the most abundant class of neocortical neurons, the glutametergic neurons. In addition, we will discuss the roles of recently-identified human- and primate-specific genes in promoting neurogenesis, leading to neocortical expansion.
Collapse
|
22
|
Eder N, Roncaroli F, Domart MC, Horswell S, Andreiuolo F, Flynn HR, Lopes AT, Claxton S, Kilday JP, Collinson L, Mao JH, Pietsch T, Thompson B, Snijders AP, Ultanir SK. YAP1/TAZ drives ependymoma-like tumour formation in mice. Nat Commun 2020; 11:2380. [PMID: 32404936 PMCID: PMC7220953 DOI: 10.1038/s41467-020-16167-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 04/17/2020] [Indexed: 11/09/2022] Open
Abstract
YAP1 gene fusions have been observed in a subset of paediatric ependymomas. Here we show that, ectopic expression of active nuclear YAP1 (nlsYAP5SA) in ventricular zone neural progenitor cells using conditionally-induced NEX/NeuroD6-Cre is sufficient to drive brain tumour formation in mice. Neuronal differentiation is inhibited in the hippocampus. Deletion of YAP1's negative regulators LATS1 and LATS2 kinases in NEX-Cre lineage in double conditional knockout mice also generates similar tumours, which are rescued by deletion of YAP1 and its paralog TAZ. YAP1/TAZ-induced mouse tumours display molecular and ultrastructural characteristics of human ependymoma. RNA sequencing and quantitative proteomics of mouse tumours demonstrate similarities to YAP1-fusion induced supratentorial ependymoma. Finally, we find that transcriptional cofactor HOPX is upregulated in mouse models and in human YAP1-fusion induced ependymoma, supporting their similarity. Our results show that uncontrolled YAP1/TAZ activity in neuronal precursor cells leads to ependymoma-like tumours in mice.
Collapse
Affiliation(s)
- Noreen Eder
- Kinases and Brain Development Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
- Protein Analysis and Proteomics Platform, The Francis Crick Institute, London, NW1 1AT, UK
| | - Federico Roncaroli
- Manchester Centre for Clinical Neuroscience, Salford Royal NHS Foundation Trust, Salford and Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, School of Biology, University of Manchester, Manchester, M13 9PT, UK
| | | | - Stuart Horswell
- Bioinformatics and Biostatistics Platform, The Francis Crick Institute, London, NW1 1AT, UK
| | - Felipe Andreiuolo
- Institute of Neuropathology, DGNN Brain Tumour Reference Center, University of Bonn, Bonn, Germany
| | - Helen R Flynn
- Protein Analysis and Proteomics Platform, The Francis Crick Institute, London, NW1 1AT, UK
| | - Andre T Lopes
- Kinases and Brain Development Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Suzanne Claxton
- Kinases and Brain Development Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - John-Paul Kilday
- Centre for Paediatric, Teenage and Young Adult Cancer, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Lucy Collinson
- Electron Microscopy Platform, The Francis Crick Institute, London, NW1 1AT, UK
| | - Jun-Hao Mao
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Torsten Pietsch
- Institute of Neuropathology, DGNN Brain Tumour Reference Center, University of Bonn, Bonn, Germany
| | - Barry Thompson
- Epithelial Biology Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Ambrosius P Snijders
- Protein Analysis and Proteomics Platform, The Francis Crick Institute, London, NW1 1AT, UK
| | - Sila K Ultanir
- Kinases and Brain Development Laboratory, The Francis Crick Institute, London, NW1 1AT, UK.
| |
Collapse
|
23
|
Szabo PA, Levitin HM, Miron M, Snyder ME, Senda T, Yuan J, Cheng YL, Bush EC, Dogra P, Thapa P, Farber DL, Sims PA. Single-cell transcriptomics of human T cells reveals tissue and activation signatures in health and disease. Nat Commun 2019; 10:4706. [PMID: 31624246 PMCID: PMC6797728 DOI: 10.1038/s41467-019-12464-3] [Citation(s) in RCA: 403] [Impact Index Per Article: 67.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 09/11/2019] [Indexed: 01/04/2023] Open
Abstract
Human T cells coordinate adaptive immunity in diverse anatomic compartments through production of cytokines and effector molecules, but it is unclear how tissue site influences T cell persistence and function. Here, we use single cell RNA-sequencing (scRNA-seq) to define the heterogeneity of human T cells isolated from lungs, lymph nodes, bone marrow and blood, and their functional responses following stimulation. Through analysis of >50,000 resting and activated T cells, we reveal tissue T cell signatures in mucosal and lymphoid sites, and lineage-specific activation states across all sites including distinct effector states for CD8+ T cells and an interferon-response state for CD4+ T cells. Comparing scRNA-seq profiles of tumor-associated T cells to our dataset reveals predominant activated CD8+ compared to CD4+ T cell states within multiple tumor types. Our results therefore establish a high dimensional reference map of human T cell activation in health for analyzing T cells in disease.
Collapse
Affiliation(s)
- Peter A Szabo
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Hanna Mendes Levitin
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Michelle Miron
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Mark E Snyder
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Takashi Senda
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Jinzhou Yuan
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Yim Ling Cheng
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Erin C Bush
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Pranay Dogra
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Puspa Thapa
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Donna L Farber
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Surgery, Columbia University Irving Medical Center, New York, NY, USA.
| | - Peter A Sims
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Biochemistry and Molecular Biophysics, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
24
|
Abstract
Deafness or hearing deficits are debilitating conditions. They are often caused by loss of sensory hair cells or defects in their function. In contrast to mammals, nonmammalian vertebrates robustly regenerate hair cells after injury. Studying the molecular and cellular basis of nonmammalian vertebrate hair cell regeneration provides valuable insights into developing cures for human deafness. In this review, we discuss the current literature on hair cell regeneration in the context of other models for sensory cell regeneration, such as the retina and the olfactory epithelium. This comparison reveals commonalities with, as well as differences between, the different regenerating systems, which begin to define a cellular and molecular blueprint of regeneration. In addition, we propose how new technical advances can address outstanding questions in the field.
Collapse
Affiliation(s)
- Nicolas Denans
- Stowers Institute for Medical Research, Kansas City, Missouri 64110, USA;
| | - Sungmin Baek
- Stowers Institute for Medical Research, Kansas City, Missouri 64110, USA;
| | - Tatjana Piotrowski
- Stowers Institute for Medical Research, Kansas City, Missouri 64110, USA;
| |
Collapse
|
25
|
Chen SY, Ishii MA, Cheng B, Otten ABC, Sun BK. HOPX Is a ZNF750 Target that Promotes Late Epidermal Differentiation. J Invest Dermatol 2019; 139:2039-2042.e2. [PMID: 30959041 DOI: 10.1016/j.jid.2019.03.1141] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 02/08/2019] [Accepted: 03/05/2019] [Indexed: 11/25/2022]
Affiliation(s)
- Selena Y Chen
- Department of Dermatology, University of California-San Diego, La Jolla, California, USA
| | - Mitsuhiro A Ishii
- Department of Dermatology, University of California-San Diego, La Jolla, California, USA
| | - Binbin Cheng
- Department of Dermatology, University of California-San Diego, La Jolla, California, USA
| | - Auke B C Otten
- Department of Dermatology, University of California-San Diego, La Jolla, California, USA
| | - Bryan K Sun
- Department of Dermatology, University of California-San Diego, La Jolla, California, USA.
| |
Collapse
|
26
|
Regional methylome profiling reveals dynamic epigenetic heterogeneity and convergent hypomethylation of stem cell quiescence-associated genes in breast cancer following neoadjuvant chemotherapy. Cell Biosci 2019; 9:16. [PMID: 30774927 PMCID: PMC6367786 DOI: 10.1186/s13578-019-0278-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 01/28/2019] [Indexed: 12/17/2022] Open
Abstract
Background Neoadjuvant chemotherapy (NAC) induces a pathological complete response (pCR) in ~ 30% of patients with breast cancer. However, aberrant DNA methylation alterations are frequent events during breast cancer progression and acquisition of chemoresistance. We aimed to characterize the inter- and intra-tumor methylation heterogeneity (MH) in breast cancer following NAC. Methods DNA methylation profiles of spatially separated regions of breast tumors before and after NAC treatment were investigated using high-density methylation microarray. Methylation levels of genes of interest were further examined using multiplexed MethyLight droplet digital PCR (ddPCR). Results We have discovered different levels of intra-tumor MH in breast cancer patients. Moreover, NAC dramatically altered the methylation profiles and such changes were highly heterogeneous between the patients. Despite the high inter-patient heterogeneity, we identified that stem cell quiescence-associated genes ALDH1L1, HOPX, WNT5A and SOX9 were convergently hypomethylated across all the samples after NAC treatment. Furthermore, by using MethyLight ddPCR, we verified that the methylation levels of these 4 genes were significantly lower in breast tumor samples after NAC than those before NAC. Conclusions Our study has revealed that NAC dramatically alters epigenetic heterogeneity in breast cancer and induces convergent hypomethylation of stem cell quiescence-associated genes, ALDH1L1, HOPX, WNT5A and SOX9, which can potentially be developed as therapeutic targets or biomarkers for chemoresistance. Electronic supplementary material The online version of this article (10.1186/s13578-019-0278-y) contains supplementary material, which is available to authorized users.
Collapse
|
27
|
Miksiunas R, Mobasheri A, Bironaite D. Homeobox Genes and Homeodomain Proteins: New Insights into Cardiac Development, Degeneration and Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1212:155-178. [PMID: 30945165 DOI: 10.1007/5584_2019_349] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cardiovascular diseases are the most common cause of human death in the developing world. Extensive evidence indicates that various toxic environmental factors and unhealthy lifestyle choices contribute to the risk, incidence and severity of cardiovascular diseases. Alterations in the genetic level of myocardium affects normal heart development and initiates pathological processes leading to various types of cardiac diseases. Homeobox genes are a large and highly specialized family of closely related genes that direct the formation of body structure, including cardiac development. Homeobox genes encode homeodomain proteins that function as transcription factors with characteristic structures that allow them to bind to DNA, regulate gene expression and subsequently control the proper physiological function of cells, tissues and organs. Mutations in homeobox genes are rare and usually lethal with evident alterations in cardiac function at or soon after the birth. Our understanding of homeobox gene family expression and function has expanded significantly during the recent years. However, the involvement of homeobox genes in the development of human and animal cardiac tissue requires further investigation. The phenotype of human congenital heart defects unveils only some aspects of human heart development. Therefore, mouse models are often used to gain a better understanding of human heart function, pathology and regeneration. In this review, we have focused on the role of homeobox genes in the development and pathology of human heart as potential tools for the future development of targeted regenerative strategies for various heart malfunctions.
Collapse
Affiliation(s)
- Rokas Miksiunas
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Ali Mobasheri
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Daiva Bironaite
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania.
| |
Collapse
|
28
|
Lock MC, Tellam RL, Botting KJ, Wang KCW, Selvanayagam JB, Brooks DA, Seed M, Morrison JL. The role of miRNA regulation in fetal cardiomyocytes, cardiac maturation and the risk of heart disease in adults. J Physiol 2018; 596:5625-5640. [PMID: 29785790 PMCID: PMC6265572 DOI: 10.1113/jp276072] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 05/15/2018] [Indexed: 12/14/2022] Open
Abstract
Myocardial infarction is a primary contributor towards the global burden of cardiovascular disease. Rather than repairing the existing damage of myocardial infarction, current treatments only address the symptoms of the disease and reducing the risk of a secondary infarction. Cardiac regenerative capacity is dependent on cardiomyocyte proliferation, which concludes soon after birth in humans and precocial species such as sheep. Human fetal cardiac tissue has some ability to repair following tissue damage, whereas a fully matured human heart has minimal capacity for cellular regeneration. This is in contrast to neonatal mice and adult zebrafish hearts, which retain the ability to undergo cardiomyocyte proliferation and can regenerate cardiac tissue after birth. In mice and zebrafish models, microRNAs (miRNAs) have been implicated in the regulation of genes involved in cardiac cell cycle progression and regeneration. However, the significance of miRNA regulation in cardiomyocyte proliferation for humans and other large mammals, where the timing of heart development in relation to birth is similar, remains unclear. miRNAs may be valuable targets for therapies that promote cardiac repair after injury. Therefore, elucidating the role of specific miRNAs in large animals, where heart development closely resembles that of humans, remains vitally important for identifying therapeutic targets that may be translated into clinical practice focused on tissue repair.
Collapse
Affiliation(s)
- Mitchell C. Lock
- Early Origins of Adult Health Research Group, School of Pharmacy & Medical SciencesUniversity of South AustraliaAdelaideSA 5001Australia
| | - Ross L. Tellam
- Early Origins of Adult Health Research Group, School of Pharmacy & Medical SciencesUniversity of South AustraliaAdelaideSA 5001Australia
| | - Kimberley J. Botting
- Early Origins of Adult Health Research Group, School of Pharmacy & Medical SciencesUniversity of South AustraliaAdelaideSA 5001Australia
| | - Kimberley C. W. Wang
- Early Origins of Adult Health Research Group, School of Pharmacy & Medical SciencesUniversity of South AustraliaAdelaideSA 5001Australia
- School of Human SciencesUniversity of Western AustraliaCrawleyWA 6009Australia
| | - Joseph B. Selvanayagam
- Cardiac Imaging Research Group, Department of Heart HealthSouth Australian Health & Medical Research Institute, and Flinders UniversityGPO Box 2100AdelaideSA 5001Australia
| | - Doug A. Brooks
- Mechanisms in Cell Biology and Disease Research Group, School of Pharmacy & Medical SciencesUniversity of South AustraliaAdelaideSA 5001Australia
| | - Mike Seed
- Hospital for Sick Children, Division of Cardiology555 University AvenueTorontoON M5G 1X8Canada
| | - Janna L. Morrison
- Early Origins of Adult Health Research Group, School of Pharmacy & Medical SciencesUniversity of South AustraliaAdelaideSA 5001Australia
| |
Collapse
|
29
|
Lima EU, Rubio IGS, Da Silva JC, Galrão AL, Pêssoa D, Oliveira TC, Carrijo F, Silva Campos I, Fonseca Espinheira L, Sampaio LJ, Lima CR, Cerutti JM, Ramos HE. HOPX homeobox methylation in differentiated thyroid cancer and its clinical relevance. Endocr Connect 2018; 7:1333-1342. [PMID: 30400039 PMCID: PMC6280589 DOI: 10.1530/ec-18-0380] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 10/24/2018] [Indexed: 12/29/2022]
Abstract
BACKGROUND The inactivation of the tumor-suppressor homeodomain-only protein X (HOPX) usually involves promoter methylation in several cancer types. This study aimed to investigate the HOPX-β mRNA expression and promoter methylation and their clinical relevance in differentiated thyroid cancer (DTC). PATIENTS AND METHODS Clinicopathological data and paraffin-embedded thyroid tumor tissues from 21 patients with DTC and 6 with benign tumors (T) and their non-tumor parenchyma (NT) were investigated. Tumor cell lines (FTC238, FTC236 and WRO) were treated with demethylating agent. HOPX-β mRNA expression was assessed by qRT-PCR and methylation status by Q-MSP. Thyroid cancer data from Cancer Genome Atlas (TCGA) was also collected. RESULTS HOPX-β mRNA re-expression in two cell lines treated with demethylating agent was observed concomitantly with reduced promoter methylation. Reduced mRNA expression in T group compared to their NT was observed, and reduced protein expression in T compared to NT was observed in three cases. Low mRNA expression with high methylation status was detected in 6/14 DTC samples. High methylation status was associated with older age at diagnosis, recurrent or progressive disease and with the presence of new neoplasm event post initial therapy while hyper-methylation correlated with worse overall survival, worse disease-free status and older age. CONCLUSION A moderate coupling of downregulation of HOPX-β mRNA expression in DTC followed by high HOPX-β promoter methylation was observed however; high HOPX promoter methylation status was associated with the worse prognosis of DTC patients.
Collapse
Affiliation(s)
- Erika Urbano Lima
- Biological Science Department, Thyroid Molecular Sciences Laboratory, Universidade Federal de São Paulo, São Paulo, Brazil
- Structural and Functional Biology Program, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ileana G S Rubio
- Biological Science Department, Thyroid Molecular Sciences Laboratory, Universidade Federal de São Paulo, São Paulo, Brazil
- Structural and Functional Biology Program, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Joaquim Custodio Da Silva
- Department of Bio-regulation, Thyroid Study Laboratory, Health & Science Institute, Federal University of Bahia, Salvador, Brazil
- Post-graduate Program in Interactive Processes of Organs and Systems, Health & Science Institute, Federal University of Bahia, Salvador, Brazil
| | - Ana Luiza Galrão
- Biological Science Department, Thyroid Molecular Sciences Laboratory, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Danielle Pêssoa
- Department of Bio-regulation, Thyroid Study Laboratory, Health & Science Institute, Federal University of Bahia, Salvador, Brazil
- Post-graduate Program in Interactive Processes of Organs and Systems, Health & Science Institute, Federal University of Bahia, Salvador, Brazil
| | - Taise Cerqueira Oliveira
- Department of Bio-regulation, Thyroid Study Laboratory, Health & Science Institute, Federal University of Bahia, Salvador, Brazil
| | - Fabiane Carrijo
- Department of Bio-regulation, Thyroid Study Laboratory, Health & Science Institute, Federal University of Bahia, Salvador, Brazil
- Post-graduate Program in Interactive Processes of Organs and Systems, Health & Science Institute, Federal University of Bahia, Salvador, Brazil
| | | | - Luciano Fonseca Espinheira
- Department of Pathology, Sao Rafael Hospital, Salvador, Brazil
- Department of Anatomic Pathology & Legal Medicine, Bahia Federal Medical School, Federal University of Bahia, Salvador, Brazil
| | | | | | - Janete Maria Cerutti
- Structural and Functional Biology Program, Universidade Federal de São Paulo, São Paulo, Brazil
- Division of Genetics, Department of Morphology and Genetics, Genetic Basis of Thyroid Tumors Laboratory, Paulista School of Medicine, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Helton Estrela Ramos
- Department of Bio-regulation, Thyroid Study Laboratory, Health & Science Institute, Federal University of Bahia, Salvador, Brazil
- Post-graduate Program in Interactive Processes of Organs and Systems, Health & Science Institute, Federal University of Bahia, Salvador, Brazil
- Correspondence should be addressed to H E Ramos:
| |
Collapse
|
30
|
Vaid S, Camp JG, Hersemann L, Eugster Oegema C, Heninger AK, Winkler S, Brandl H, Sarov M, Treutlein B, Huttner WB, Namba T. A novel population of Hopx-dependent basal radial glial cells in the developing mouse neocortex. Development 2018; 145:dev.169276. [PMID: 30266827 DOI: 10.1242/dev.169276] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 09/18/2018] [Indexed: 01/10/2023]
Abstract
A specific subpopulation of neural progenitor cells, the basal radial glial cells (bRGCs) of the outer subventricular zone (OSVZ), are thought to have a key role in the evolutionary expansion of the mammalian neocortex. In the developing lissencephalic mouse neocortex, bRGCs exist at low abundance and show significant molecular differences from bRGCs in developing gyrencephalic species. Here, we demonstrate that the developing mouse medial neocortex (medNcx), in contrast to the canonically studied lateral neocortex (latNcx), exhibits an OSVZ and an abundance of bRGCs similar to that in developing gyrencephalic neocortex. Unlike bRGCs in developing mouse latNcx, the bRGCs in medNcx exhibit human bRGC-like gene expression, including expression of Hopx, a human bRGC marker. Disruption of Hopx expression in mouse embryonic medNcx and forced Hopx expression in mouse embryonic latNcx demonstrate that Hopx is required and sufficient, respectively, for bRGC abundance as found in the developing gyrencephalic neocortex. Taken together, our data identify a novel bRGC subpopulation in developing mouse medNcx that is highly related to bRGCs of developing gyrencephalic neocortex.
Collapse
Affiliation(s)
- Samir Vaid
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, D-01307 Dresden, Germany
| | - J Gray Camp
- Max Planck Institute for Evolutionary Anthropology, Deutscher Platz 6, D-04103 Leipzig, Germany
| | - Lena Hersemann
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, D-01307 Dresden, Germany
| | - Christina Eugster Oegema
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, D-01307 Dresden, Germany
| | - Anne-Kristin Heninger
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, D-01307 Dresden, Germany
| | - Sylke Winkler
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, D-01307 Dresden, Germany
| | - Holger Brandl
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, D-01307 Dresden, Germany
| | - Mihail Sarov
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, D-01307 Dresden, Germany
| | - Barbara Treutlein
- Max Planck Institute for Evolutionary Anthropology, Deutscher Platz 6, D-04103 Leipzig, Germany
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, D-01307 Dresden, Germany
| | - Takashi Namba
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, D-01307 Dresden, Germany
| |
Collapse
|
31
|
Guerrero-Juarez CF, Astrowski AA, Murad R, Dang CT, Shatrova VO, Astrowskaja A, Lim CH, Ramos R, Wang X, Liu Y, Lee HL, Pham KT, Hsi TC, Oh JW, Crocker D, Mortazavi A, Ito M, Plikus MV. Wound Regeneration Deficit in Rats Correlates with Low Morphogenetic Potential and Distinct Transcriptome Profile of Epidermis. J Invest Dermatol 2018; 138:1409-1419. [PMID: 29317265 DOI: 10.1016/j.jid.2017.12.030] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 12/14/2017] [Accepted: 12/22/2017] [Indexed: 01/10/2023]
Abstract
Large excisional wounds in mice prominently regenerate new hair follicles (HFs) and fat, yet humans are deficient for this regenerative behavior. Currently, wound-induced regeneration remains a clinically desirable, but only partially understood phenomenon. We show that large excisional wounds in rats across seven strains fail to regenerate new HFs. We compared wound transcriptomes between mice and rats at the time of scab detachment, which coincides with the onset of HF regeneration in mice. In both species, wound dermis and epidermis share core dermal and epidermal transcriptional programs, respectively, yet prominent interspecies differences exist. Compared with mice, rat epidermis expresses distinct transcriptional and epigenetic factors, markers of epidermal repair, hyperplasia, and inflammation, and lower levels of WNT signaling effectors and regulators. When recombined on the surface of excisional wounds with vibrissa dermal papillae, partial-thickness skin grafts containing distal pelage HF segments, but not interfollicular epidermis, readily regenerated new vibrissa-like HFs. Together, our findings establish rats as a nonregenerating rodent model for excisional wound healing and suggest that low epidermal competence and associated transcriptional profile may contribute to its regenerative deficiency. Future comparison between rat and mouse may lend further insight into the mechanism of wounding-induced regeneration and causes for its deficit.
Collapse
Affiliation(s)
- Christian F Guerrero-Juarez
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, California, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, California, USA
| | - Aliaksandr A Astrowski
- Department of Medical Biology and Genetics, Grodna State Medical University, Grodna, Belarus
| | - Rabi Murad
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, California, USA
| | - Christina T Dang
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, California, USA
| | - Vera O Shatrova
- Department of Medical Biology and Genetics, Grodna State Medical University, Grodna, Belarus
| | - Aksana Astrowskaja
- Department of Medical Biology and Genetics, Grodna State Medical University, Grodna, Belarus
| | - Chae Ho Lim
- The Ronald O. Perelman Department of Dermatology, Department of Cell Biology, New York University School of Medicine, New York, New York, USA
| | - Raul Ramos
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, California, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, California, USA
| | - Xiaojie Wang
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, California, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, California, USA
| | - Yuchen Liu
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, California, USA
| | - Hye-Lim Lee
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, California, USA
| | - Kim T Pham
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, California, USA
| | - Tsai-Ching Hsi
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, California, USA
| | - Ji Won Oh
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, California, USA; Department of Anatomy, School of Medicine, Kyungpook National University, Daegu, Republic of Korea; Biomedical Research Institute, Kyungpook National University Hospital, Daegu, Republic of Korea; Hair Transplantation Center, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Daniel Crocker
- Department of Biology, Sonoma State University, Rohnert Park, California, USA
| | - Ali Mortazavi
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, California, USA
| | - Mayumi Ito
- The Ronald O. Perelman Department of Dermatology, Department of Cell Biology, New York University School of Medicine, New York, New York, USA
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, California, USA; Center for Complex Biological Systems, University of California, Irvine, Irvine, California, USA.
| |
Collapse
|
32
|
Yap LF, Lai SL, Patmanathan SN, Gokulan R, Robinson CM, White JB, Chai SJ, Rajadurai P, Prepageran N, Liew YT, Lopes V, Wei W, Hollows RJ, Murray PG, Lambert DW, Hunter KD, Paterson IC. HOPX functions as a tumour suppressor in head and neck cancer. Sci Rep 2016; 6:38758. [PMID: 27934959 PMCID: PMC5146930 DOI: 10.1038/srep38758] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 11/14/2016] [Indexed: 11/08/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is generalized term that encompasses a diverse group of cancers that includes tumours of the oral cavity (OSCC), oropharynx (OPSCC) and nasopharynx (NPC). Genetic alterations that are common to all HNSCC types are likely to be important for squamous carcinogenesis. In this study, we have investigated the role of the homeodomain-only homeobox gene, HOPX, in the pathogenesis of HNSCC. We show that HOPX mRNA levels are reduced in OSCC and NPC cell lines and tissues and there is a general reduction of HOPX protein expression in these tumours and OPSCCs. HOPX promoter methylation was observed in a subset of HNSCCs and was associated with a worse overall survival in HPV negative tumours. RNAseq analysis of OSCC cells transfected with HOPX revealed a widespread deregulation of the transcription of genes related to epithelial homeostasis and ectopic over-expression of HOPX in OSCC and NPC cells inhibited cell proliferation, plating efficiency and migration, and enhanced sensitivity to UVA-induced apoptosis. Our results demonstrate that HOPX functions as a tumour suppressor in HNSCC and suggest a central role for HOPX in suppressing epithelial carcinogenesis.
Collapse
Affiliation(s)
- Lee Fah Yap
- Department of Oral and Craniofacial Sciences and Oral Cancer Research and Coordinating Centre, Faculty of Dentistry, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Sook Ling Lai
- Department of Oral and Craniofacial Sciences and Oral Cancer Research and Coordinating Centre, Faculty of Dentistry, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Sathya Narayanan Patmanathan
- Department of Oral and Craniofacial Sciences and Oral Cancer Research and Coordinating Centre, Faculty of Dentistry, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Ravindran Gokulan
- Department of Oral and Craniofacial Sciences and Oral Cancer Research and Coordinating Centre, Faculty of Dentistry, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - C. Max Robinson
- Centre for Oral Health Research, Newcastle University, Newcastle, NE2 4BW, United Kingdom
| | - Joe B. White
- Unit of Oral and Maxillofacial Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield, S10 2TA, Unite Kingdom
| | - San Jiun Chai
- Cancer Research Malaysia, Selangor, 47500 Subang Jaya, Malaysia
| | | | - Narayanan Prepageran
- Department of Otorhinolaryngology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Yew Toong Liew
- Department of Otorhinolaryngology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Victor Lopes
- Department of Oral surgery, Edinburgh Postgraduate Dental Institute, University of Edinburgh, Edinburgh, EH3 9HA, United Kingdom
| | - Wenbin Wei
- Institute of Cancer and Genomic Studies, University of Birmingham, Birmingham, B15 2TT, United Kingdom
- Sheffield Institute of Translational Neuroscience, University of Sheffield, Sheffield, S10 2HQ, United Kingdom
| | - Robert J. Hollows
- Institute of Cancer and Genomic Studies, University of Birmingham, Birmingham, B15 2TT, United Kingdom
| | - Paul G. Murray
- Institute of Cancer and Genomic Studies, University of Birmingham, Birmingham, B15 2TT, United Kingdom
| | - Daniel W. Lambert
- Unit of Oral and Maxillofacial Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield, S10 2TA, Unite Kingdom
| | - Keith D. Hunter
- Unit of Oral and Maxillofacial Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield, S10 2TA, Unite Kingdom
| | - Ian C. Paterson
- Department of Oral and Craniofacial Sciences and Oral Cancer Research and Coordinating Centre, Faculty of Dentistry, University of Malaya, 50603 Kuala Lumpur, Malaysia
| |
Collapse
|